Cost-effectiveness analysis of PET-CT positron emission ... - AOTM

Cost-effectiveness analysis of PET-CT positron emission ... - AOTM Cost-effectiveness analysis of PET-CT positron emission ... - AOTM

29.06.2013 Views

Cost-effectiveness analysis of PET-CT positron emission tomography and the diagnostic technologies financed from public sources in oncological diagnostics in Poland. Clinical and epidemiological aspects Health Technology Assessment Report commissioned by Agency for Health Technology Assessment in Poland prepared by Public Health and Social Insurance Institute WyŜsza Szkoła Biznesu National-Louis University Warszawa – Nowy Sącz 2006 1

<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong><br />

tomography and the diagnostic technologies financed from public<br />

sources in oncological diagnostics in Poland. Clinical and<br />

epidemiological aspects<br />

Health Technology Assessment Report<br />

commissioned by<br />

Agency for Health Technology Assessment in Poland<br />

prepared by<br />

Public Health and Social Insurance Institute<br />

WyŜsza Szkoła Biznesu<br />

National-Louis University<br />

Warszawa – Nowy Sącz 2006<br />

1


PRINCIPAL<br />

AGENCY FOR HEALTH TECHNOLOGY ASSESSMENT IN POLAND<br />

22 Lotników Av., 02-668 Warsaw<br />

tel. +48 22 56 67 200<br />

fax +48 22 56 67 202<br />

www.aotm.gov.pl<br />

CONTRA<strong>CT</strong>OR<br />

Public Health and Social Insurance Institute<br />

WYśSZA SZKOŁA BIZNESU<br />

NATIONAL-LOUIS UNIVERISTY<br />

27 Zielona Str., 33-300 Nowy Sącz<br />

tel. +48 18 44 99 120<br />

fax +48 18 44 99 121<br />

e-mail. wsb-nlu@wsb-nlu.edu.pl<br />

www.wsb-nlu.edu.pl<br />

2


THIS REPORT WAS ASSISTED BY SPECIALISTS FROM<br />

MARIA SKLODOWSKA-CURIE MEMORIAL CANCER CENTRE AND INSTITUTE OF<br />

ONCOLOGY<br />

pr<strong>of</strong>. dr hab. med. Marek Nowacki<br />

pr<strong>of</strong>. dr hab. med. Witold Bartnik<br />

doc. dr hab. med. Mariusz Bidziński<br />

doc. dr hab. med. Andrzej Kawecki<br />

doc. dr hab. med. Włodzimierz Ruka<br />

doc. dr hab. med. Jan Walewski<br />

dr med. Wiesław Lasota<br />

dr med. Janusz Meder<br />

dr med. Andrzej Pietraszek<br />

dr med. Piotr Siedlecki<br />

dr med. Piotr Rutkowski<br />

INSTITUTE OF PULMONARY DISEASES AND TUBERCULOSIS IN WARSAW<br />

pr<strong>of</strong>. dr hab. med. Kazimierz Roszkowski-ŚliŜ<br />

3


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

LIST OF CONTENTS<br />

1. SUMMARY.............................................................................................................................. 15<br />

2. INDEX OF ABBREVIATIONS.................................................................................................... 27<br />

3. DESCRIPTION OF HEALTH PROBLEM...................................................................................... 29<br />

3.1. Epidemiology <strong>of</strong> cancers...................................................................................................................... 29<br />

3.2. Lung cancer............................................................................................................................................ 34<br />

3.2.1. Epidemiology............................................................................................................................................34<br />

3.1.1. Risk factors.................................................................................................................................................36<br />

3.1.2. Symptoms ..................................................................................................................................................36<br />

3.1.3. Histopathological picture.......................................................................................................................38<br />

3.1.4. Staging.......................................................................................................................................................40<br />

4<br />

3.1.4.1. Non-small cell cancer staging ............................................................................................................. 41<br />

3.1.4.2. Small cell cancer staging ..................................................................................................................... 41<br />

3.1.5. Diagnostics and treatment ....................................................................................................................41<br />

3.1.5.1. Case history ............................................................................................................................................. 41<br />

3.1.5.2. Physical examination............................................................................................................................. 41<br />

3.1.5.3. Diagnostic imaging................................................................................................................................ 42<br />

3.1.5.4. Microscopic diagnosis........................................................................................................................... 42<br />

3.1.5.5. Other tests................................................................................................................................................ 43<br />

3.1.5.6. General principles in small cell lung cancer diagnosis ................................................................... 43<br />

3.1.5.7. General principles <strong>of</strong> treatment .......................................................................................................... 43<br />

3.1.5.8. Non-small cell cancer ........................................................................................................................... 44<br />

3.1.5.9. Small cell carcinoma ............................................................................................................................. 45<br />

3.1.6. Prognosis ....................................................................................................................................................45<br />

3.3. Lymphomas ............................................................................................................................................ 46<br />

3.3.1. Epidemiology............................................................................................................................................46<br />

3.3.1.1 Non-Hodgkin’s lymphomas .................................................................................................................. 46<br />

3.3.1.2 Hodgkin’s disease .................................................................................................................................. 46<br />

3.3.2. Risk factors.................................................................................................................................................46<br />

3.3.3. Symptoms ..................................................................................................................................................47<br />

3.3.4. Histopathological picture.......................................................................................................................47<br />

3.3.4.1 Hodgkin’s disease .................................................................................................................................. 47<br />

3.3.4.2 Non-Hodgkin’s lymphomas .................................................................................................................. 48<br />

3.3.5. Staging.......................................................................................................................................................48<br />

3.3.6. Diagnostics and treatment ....................................................................................................................50<br />

3.3.6.1 Additional tests ....................................................................................................................................... 50<br />

3.3.6.2 Diagnostic imaging................................................................................................................................ 50


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

3.3.6.3 Cytological and histopathological study .......................................................................................... 50<br />

3.3.6.4 Molecular tests........................................................................................................................................ 50<br />

3.3.6.5 Treatment <strong>of</strong> Hodgkin’s disease .......................................................................................................... 51<br />

3.3.6.6 Treatment <strong>of</strong> non-Hodgkin’s lymphomas........................................................................................... 52<br />

3.3.7. Prognosis ....................................................................................................................................................52<br />

3.4. Esophageal cancer ............................................................................................................................... 53<br />

3.4.1. Epidemiology............................................................................................................................................53<br />

3.4.2. Risk factors.................................................................................................................................................54<br />

3.4.3. Symptoms ..................................................................................................................................................54<br />

3.4.4. Histopathological picture.......................................................................................................................54<br />

3.4.5. Staging.......................................................................................................................................................55<br />

3.4.6. Diagnostics and treatment ....................................................................................................................56<br />

3.4.6.1 Physical examination............................................................................................................................. 56<br />

3.4.6.2 Diagnostic imaging................................................................................................................................ 56<br />

3.4.6.3 Endoscopy............................................................................................................................................... 56<br />

3.4.6.4 Cancer markers...................................................................................................................................... 56<br />

3.4.6.5 Histopathological study......................................................................................................................... 56<br />

3.4.6.6 Radical treatment.................................................................................................................................. 56<br />

3.4.6.7 Palliative treatment................................................................................................................................ 57<br />

3.4.7. Prognosis ....................................................................................................................................................57<br />

3.5. Cancer <strong>of</strong> the female genitals ............................................................................................................. 57<br />

3.5.1. Endometrial cancer.................................................................................................................................57<br />

3.5.1.1 Epidemiology .......................................................................................................................................... 57<br />

3.5.1.2 Risk factors ............................................................................................................................................... 58<br />

3.5.1.3 Symptoms ................................................................................................................................................ 58<br />

3.5.1.4 Histopathological picture ..................................................................................................................... 58<br />

3.5.1.5 Staging ..................................................................................................................................................... 58<br />

3.5.1.6 Diagnostics and treatment................................................................................................................... 59<br />

3.5.1.7 Prognosis .................................................................................................................................................. 60<br />

3.5.2. Cervical cancer .......................................................................................................................................60<br />

3.5.2.1 Epidemiology .......................................................................................................................................... 60<br />

3.5.2.2 Risk factors ............................................................................................................................................... 60<br />

3.5.2.3 Symptoms ................................................................................................................................................ 61<br />

3.5.2.4 Histopathological picture ..................................................................................................................... 61<br />

3.5.2.5 Staging ..................................................................................................................................................... 61<br />

3.5.2.6 Diagnostics and treatment................................................................................................................... 62<br />

3.5.2.7 Prognosis .................................................................................................................................................. 63<br />

3.5.3. Ovarian cancer........................................................................................................................................63<br />

3.5.3.1 Epidemiology .......................................................................................................................................... 63<br />

3.5.3.2 Risk factors ............................................................................................................................................... 64<br />

5


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

6<br />

3.5.3.3 Symptoms ................................................................................................................................................ 64<br />

3.5.3.4 Histopathological picture ..................................................................................................................... 64<br />

3.5.3.5 Staging ..................................................................................................................................................... 65<br />

3.5.3.6 Diagnostics and treatment................................................................................................................... 66<br />

3.5.3.7 Prognosis .................................................................................................................................................. 66<br />

3.6. Thyroid gland cancer ............................................................................................................................ 67<br />

3.6.1. Epidemiology............................................................................................................................................67<br />

3.6.2. Risk factors.................................................................................................................................................67<br />

3.6.3. Symptoms ..................................................................................................................................................67<br />

3.6.4. Histopathological picture.......................................................................................................................68<br />

3.6.5. Staging.......................................................................................................................................................68<br />

3.6.6. Diagnostics and treatment ....................................................................................................................69<br />

3.6.7. Prognosis ....................................................................................................................................................70<br />

3.7. Head and neck cancer ........................................................................................................................ 70<br />

3.7.1. Classification .............................................................................................................................................70<br />

3.7.2. Epidemiology............................................................................................................................................70<br />

3.7.3. Risk factors.................................................................................................................................................71<br />

3.7.4. Symptoms ..................................................................................................................................................71<br />

3.7.5. Histopathological picture.......................................................................................................................71<br />

3.7.6. Staging.......................................................................................................................................................71<br />

3.7.7. Diagnostics and treatment ....................................................................................................................72<br />

3.7.8. Prognosis ....................................................................................................................................................73<br />

3.8. Pancreatic cancer................................................................................................................................. 73<br />

3.8.1. Epidemiology............................................................................................................................................73<br />

3.8.2. Risk factors.................................................................................................................................................74<br />

3.8.3. Symptoms ..................................................................................................................................................74<br />

3.8.4. Histopathological picture.......................................................................................................................74<br />

3.8.5. Staging.......................................................................................................................................................75<br />

3.8.6. Diagnostics and treatment ....................................................................................................................75<br />

3.8.7. Prognosis ....................................................................................................................................................77<br />

3.9. Gastrointestinal stromal tumor.............................................................................................................. 78<br />

3.9.1. Epidemiology............................................................................................................................................78<br />

3.9.2. Symptoms ..................................................................................................................................................79<br />

3.9.3. Histopathological picture.......................................................................................................................79<br />

3.9.4. Staging.......................................................................................................................................................79<br />

3.9.5. Diagnostics and treatment ....................................................................................................................79<br />

3.9.6. Prognosis ....................................................................................................................................................80<br />

3.10. Colon cancer ................................................................................................................................. 81


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

3.10.1. Epidemiology................................................................................................................................81<br />

3.10.2. Risk factors.....................................................................................................................................81<br />

3.10.3. Symptoms ......................................................................................................................................82<br />

3.10.4. Histopathological picture...........................................................................................................82<br />

3.10.5. Staging...........................................................................................................................................82<br />

3.10.6. Diagnostics and treatment........................................................................................................83<br />

3.10.6.1 Screening examinations........................................................................................................................ 83<br />

3.10.6.2 Physical examination............................................................................................................................. 83<br />

3.10.6.3 Laboratory tests ...................................................................................................................................... 83<br />

3.10.6.4 Endoscopy............................................................................................................................................... 83<br />

3.10.6.5 Diagnostic imaging................................................................................................................................ 84<br />

3.10.6.6 Surgery...................................................................................................................................................... 84<br />

3.10.6.7 Supplementary treatment .................................................................................................................... 84<br />

3.10.6.8 Treatment <strong>of</strong> disseminated disease.................................................................................................... 84<br />

3.10.6.9 Evaluation <strong>of</strong> response to treatment .................................................................................................. 84<br />

3.10.6.10 Follow-up.................................................................................................................................................. 84<br />

3.10.7. Prognosis ........................................................................................................................................85<br />

4. DESCRIPTION OF INTERVENTION........................................................................................... 86<br />

4.1. Description <strong>of</strong> method ........................................................................................................................... 86<br />

4.2. Examination protocol, absorbed dose................................................................................................ 87<br />

4.3. Limitations <strong>of</strong> method ............................................................................................................................ 88<br />

4.4. Uses <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> ......................................................................................................................................... 89<br />

4.4.1. Oncology...................................................................................................................................................89<br />

4.4.2. Neurology..................................................................................................................................................90<br />

4.4.3. Cardiology ................................................................................................................................................91<br />

4.5. Results...................................................................................................................................................... 91<br />

5. METHODOLOGY .................................................................................................................... 92<br />

5.1. Purpose <strong>of</strong> study ..................................................................................................................................... 92<br />

5.2. Method <strong>of</strong> clinical efficacy assessment.............................................................................................. 92<br />

5.3. Search strategy for primary studies ..................................................................................................... 92<br />

5.4. Medical database search .................................................................................................................... 93<br />

5.5. Criteria for the inclusion <strong>of</strong> primary studies in the <strong>analysis</strong> ............................................................... 94<br />

5.5.1. Population .................................................................................................................................................95<br />

5.5.2. Intervention ...............................................................................................................................................95<br />

5.5.3. Technologies compared (comparators) ............................................................................................95<br />

5.5.4. End points ..................................................................................................................................................95<br />

5.6. Trial quality assessment......................................................................................................................... 95<br />

7


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

5.7. Statistical <strong>analysis</strong> .................................................................................................................................. 96<br />

5.8. Conflict <strong>of</strong> interest .................................................................................................................................. 98<br />

6. SEARCH RESULTS.................................................................................................................... 99<br />

7. COMPARATIVE ANALYSIS OF THE EFFICACY OF <strong>PET</strong>-<strong>CT</strong> VS <strong>CT</strong> IN LUNG CANCER<br />

STAGING.............................................................................................................................. 101<br />

7.1. Results <strong>of</strong> trial search ........................................................................................................................... 101<br />

7.2. Population characteristics .................................................................................................................. 101<br />

7.3. Description <strong>of</strong> Intervention.................................................................................................................. 102<br />

7.3.1. <strong>PET</strong>-<strong>CT</strong> imaging .......................................................................................................................................102<br />

7.3.2. Diagnostic test compared ...................................................................................................................103<br />

7.3.3. Reference test ........................................................................................................................................104<br />

7.4. Findings ................................................................................................................................................. 106<br />

7.4.1. T-staging...................................................................................................................................................106<br />

7.4.2. Assessment <strong>of</strong> lymph node involvement (N feature)......................................................................109<br />

7.4.3. Assessment <strong>of</strong> distant metastases (M feature).................................................................................114<br />

7.4.4. TNM staging system ...............................................................................................................................115<br />

7.4.5. Impact on therapy ................................................................................................................................120<br />

7.4.6. Safety .......................................................................................................................................................120<br />

7.5. Results.................................................................................................................................................... 120<br />

8. COMPARATIVE ANALYSIS OF THE EFFICACY OF <strong>PET</strong>-<strong>CT</strong> VS <strong>CT</strong> IN LYMPHOMA<br />

STAGING.............................................................................................................................. 122<br />

8.1. Results <strong>of</strong> primary trial search............................................................................................................. 122<br />

8.2. Population characteristics .................................................................................................................. 122<br />

8.3. Description <strong>of</strong> intervention .................................................................................................................. 123<br />

8.3.1. <strong>PET</strong>-<strong>CT</strong> imaging .......................................................................................................................................123<br />

8.3.2. Diagnostic technology compared ....................................................................................................123<br />

8.3.3. Reference test ........................................................................................................................................124<br />

8.4. Findings ................................................................................................................................................. 124<br />

8.4.1. Diagnosis efficacy in disease staging ................................................................................................124<br />

8<br />

8.4.1.1 In nodes.................................................................................................................................................. 124<br />

8.4.1.2 Locations outside <strong>of</strong> nodes ................................................................................................................ 126<br />

8.4.1.3 Total results............................................................................................................................................. 126<br />

8.4.1.3.1 Sensitivity...................................................................................................................................... 127<br />

8.4.1.3.2 Specificity .................................................................................................................................... 128<br />

8.4.1.3.3 Positive likelihood ratio .............................................................................................................. 130<br />

8.4.1.3.4 Negative likelihood ratio........................................................................................................... 132<br />

8.4.1.3.5 Diagnostic odds ratio ................................................................................................................ 134<br />

8.4.1.3.6 Diagnostic accuracy................................................................................................................. 136<br />

8.4.1.3.7 Results........................................................................................................................................... 138


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

8.4.2. Accuracy <strong>of</strong> staging based on Ann Arbor staging system ...........................................................138<br />

8.4.3. Safety .......................................................................................................................................................139<br />

8.5. Results.................................................................................................................................................... 139<br />

9. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS. <strong>CT</strong> IN ESOPHAGEAL CANCER<br />

STAGING.............................................................................................................................. 141<br />

9.1. Disease staging .................................................................................................................................... 141<br />

9.1.1. Results <strong>of</strong> primary trial search ..............................................................................................................141<br />

9.1.2. Population characteristics....................................................................................................................141<br />

9.1.3. Description <strong>of</strong> intervention ...................................................................................................................142<br />

9.1.3.1 <strong>PET</strong>-<strong>CT</strong> imaging ..................................................................................................................................... 142<br />

9.1.3.2 Diagnostic technology compared ................................................................................................... 143<br />

9.1.3.3 Reference test ...................................................................................................................................... 143<br />

9.1.4. Findings ....................................................................................................................................................143<br />

9.1.4.1 Diagnostic efficacy <strong>of</strong> test.................................................................................................................. 143<br />

9.1.4.1.1 T-staging....................................................................................................................................... 143<br />

9.1.4.1.2 N-staging...................................................................................................................................... 146<br />

9.1.4.1.3 M-staging..................................................................................................................................... 147<br />

9.1.4.1.4 CR – complete response........................................................................................................... 149<br />

9.1.4.2 Safety...................................................................................................................................................... 150<br />

9.1.5. Results.......................................................................................................................................................151<br />

9.2. Diagnostics <strong>of</strong> primary esophageal cancer..................................................................................... 152<br />

9.2.1. Primal tests search results .....................................................................................................................152<br />

9.2.2. Population characteristics....................................................................................................................152<br />

9.2.3. Description <strong>of</strong> intervention ...................................................................................................................153<br />

9.2.3.1 <strong>PET</strong>-<strong>CT</strong> imaging ..................................................................................................................................... 153<br />

9.2.3.2 Diagnostic technologies compared ................................................................................................ 153<br />

9.2.3.3 Reference test ...................................................................................................................................... 154<br />

9.2.4. Safety .......................................................................................................................................................154<br />

9.2.5. Findings ....................................................................................................................................................154<br />

9.2.6. Results.......................................................................................................................................................154<br />

10. COMPARATIVE ANALYSIS OF THE EFFICACY OF <strong>PET</strong>-<strong>CT</strong> VS. CONVENTIONAL<br />

IMAGING METHODS (<strong>CT</strong>, MRI, USG) IN CLINICAL STAGING, AND DETE<strong>CT</strong>ING<br />

RECURRENCES OF, MALIGNANT FEMALE GENITAL TUMORS.............................................. 155<br />

10.1. Results <strong>of</strong> primary study search.................................................................................................. 155<br />

10.2. Population characteristics .......................................................................................................... 155<br />

10.3. Description <strong>of</strong> intervention.......................................................................................................... 156<br />

10.3.1. <strong>PET</strong>-<strong>CT</strong> imaging...........................................................................................................................156<br />

10.3.2. Diagnostic technologies compared......................................................................................156<br />

10.3.3. Reference test ............................................................................................................................156<br />

10.4. Findings ......................................................................................................................................... 157<br />

9


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

10.4.1. Disease staging ..........................................................................................................................157<br />

10.4.2. Assessment <strong>of</strong> disease recurrence .........................................................................................159<br />

10.4.3. Total assessment.........................................................................................................................160<br />

10.5. Results............................................................................................................................................ 163<br />

11. COMPARATIVE ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> AND <strong>CT</strong> EFFICACY IN THE DIAGNOSTICS OF<br />

OVARIAN CANCER ............................................................................................................. 164<br />

11.1. Results <strong>of</strong> primary trial search .................................................................................................... 164<br />

11.2. Population characteristics .......................................................................................................... 164<br />

11.3. Description <strong>of</strong> intervention.......................................................................................................... 165<br />

11.3.1. <strong>PET</strong>-<strong>CT</strong> imaging...........................................................................................................................165<br />

11.3.2. Diagnostic technology compared ........................................................................................166<br />

11.3.3. Reference test ............................................................................................................................166<br />

11.4. Findings ......................................................................................................................................... 166<br />

11.4.1. Diagnostic efficacy ...................................................................................................................166<br />

11.4.2. Safety ...........................................................................................................................................170<br />

11.5. Results............................................................................................................................................ 171<br />

12. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS. TRADITIONAL IMAGING METHODS<br />

IN THYROID CANCER DIAGNOSTICS .................................................................................. 172<br />

12.1. Comparative <strong>analysis</strong> <strong>of</strong> the clinical efficacy <strong>of</strong> pet-ct vs. traditional imaging<br />

methods (ct/mri and i131whole-body scintigraphy) in the diagnostics <strong>of</strong> thyroid cancer<br />

recurrences........................................................................................................................................... 172<br />

12.1.1. Results <strong>of</strong> primary trial search ..................................................................................................172<br />

12.1.2. Population characteristics .......................................................................................................172<br />

12.1.3. Description <strong>of</strong> intervention.......................................................................................................173<br />

10<br />

12.1.3.1 <strong>PET</strong>-<strong>CT</strong> imaging ..................................................................................................................................... 173<br />

12.1.3.2 Diagnostic technologies compared ................................................................................................ 174<br />

12.1.3.2.1 <strong>CT</strong>/MRI .......................................................................................................................................... 174<br />

12.1.3.2.2 I131 whole-body scintigraphy (I131 WBS) .............................................................................. 174<br />

12.1.3.3 Reference Test ...................................................................................................................................... 175<br />

12.1.4. Findings ........................................................................................................................................175<br />

12.1.4.1 Safety...................................................................................................................................................... 182<br />

12.1.5. Impact on therapy ....................................................................................................................182<br />

12.1.6. Results...........................................................................................................................................183<br />

12.2. Comparative efficacy <strong>analysis</strong> <strong>of</strong> f pet-ct vs. traditional imaging methods (<strong>CT</strong>, I131<br />

whole-body scintigraphy and USG) in the staging <strong>of</strong> differentiated thyroid cancer .................. 184<br />

12.2.1. Results <strong>of</strong> primary trial search ..................................................................................................184<br />

12.2.2. Population characteristics .......................................................................................................184<br />

12.2.3. Description <strong>of</strong> intervention.......................................................................................................185<br />

12.2.3.1 I 124 <strong>PET</strong>-<strong>CT</strong> imaging ............................................................................................................................... 185


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

12.2.3.2 Diagnostic technology compared ................................................................................................... 186<br />

12.2.3.2.1 Computer tomography ............................................................................................................ 186<br />

12.2.3.2.2 I131 whole-body scintigraphy.................................................................................................. 186<br />

12.2.3.2.3 Ultrasound examination <strong>of</strong> the neck ...................................................................................... 186<br />

12.2.3.3 Reference test ...................................................................................................................................... 186<br />

12.2.4. Findings ........................................................................................................................................186<br />

12.2.4.1 Safety...................................................................................................................................................... 188<br />

12.2.5. Results...........................................................................................................................................188<br />

13. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS. TRADITIONAL IMAGING METHODS<br />

IN HEAD AND NECK CANCER DIAGNOSTICS .................................................................... 190<br />

13.1. Diagnostics <strong>of</strong> malignant head and neck tumors (primary lesions, recurrence after<br />

treatment, cervical lymph node metastasis from an unknown primary focus) ........................... 190<br />

13.1.1. Results <strong>of</strong> primary trial search ..................................................................................................190<br />

13.1.2. Population characteristics .......................................................................................................190<br />

13.1.3. Description <strong>of</strong> intervention.......................................................................................................191<br />

13.1.3.1 <strong>PET</strong>-<strong>CT</strong> imaging ..................................................................................................................................... 191<br />

13.1.3.2 Diagnostic technology compared ................................................................................................... 191<br />

13.1.3.3 Reference test ...................................................................................................................................... 191<br />

13.1.4. Findings ........................................................................................................................................191<br />

13.1.4.1 Diagnostic efficacy.............................................................................................................................. 191<br />

13.1.4.2 Safety...................................................................................................................................................... 193<br />

13.1.5. Results...........................................................................................................................................193<br />

13.2. Detection <strong>of</strong> bone involvement by oral cancer ...................................................................... 194<br />

13.2.1. Results <strong>of</strong> primary trial search ..................................................................................................194<br />

13.2.2. Population characteristics .......................................................................................................194<br />

13.2.3. Description <strong>of</strong> intervention.......................................................................................................194<br />

13.2.3.1 <strong>PET</strong>-<strong>CT</strong> imaging ..................................................................................................................................... 194<br />

13.2.3.2 Diagnostic test compared.................................................................................................................. 195<br />

13.2.3.3 Reference test ...................................................................................................................................... 195<br />

13.2.4. Findings ........................................................................................................................................195<br />

13.2.4.1 Diagnostic efficacy.............................................................................................................................. 195<br />

13.2.4.2 Safety...................................................................................................................................................... 197<br />

13.2.5. Results...........................................................................................................................................197<br />

13.3. Impact <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostics on treatment revisions.............................................................. 198<br />

13.3.1. Results <strong>of</strong> primary trial search ..................................................................................................198<br />

13.3.2. Population characteristics .......................................................................................................198<br />

13.3.3. Intervention description............................................................................................................199<br />

13.3.3.1 <strong>PET</strong>-<strong>CT</strong> imaging ..................................................................................................................................... 199<br />

13.3.3.2 Diagnostic technology compared ................................................................................................... 200<br />

13.3.3.3 Reference test ...................................................................................................................................... 200<br />

11


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

13.3.4. Results...........................................................................................................................................200<br />

12<br />

13.3.4.1 Impact on therapy............................................................................................................................... 200<br />

13.3.4.2 Safety...................................................................................................................................................... 202<br />

13.3.5. Results...........................................................................................................................................203<br />

14. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS. CONVENTIONAL IMAGING<br />

METHODS IN PANCREATIC CANCER DIAGNOSTICS.......................................................... 204<br />

14.1. Results <strong>of</strong> primary trial search .................................................................................................... 204<br />

14.2. Population characteristics .......................................................................................................... 204<br />

14.3. Intervention description .............................................................................................................. 205<br />

14.3.1. <strong>PET</strong>-<strong>CT</strong> imaging...........................................................................................................................205<br />

14.3.2. Diagnostic technology compared ........................................................................................206<br />

14.3.3. Reference test ............................................................................................................................206<br />

14.4. Findings ......................................................................................................................................... 206<br />

14.4.1. Tumor detection and <strong>analysis</strong> .................................................................................................206<br />

14.4.2. Staging.........................................................................................................................................207<br />

14.4.3. Revisions <strong>of</strong> oncological procedures.....................................................................................210<br />

14.4.4. Safety ...........................................................................................................................................211<br />

14.5. Results......................................................................................... Błąd! Nie zdefiniowano zakładki.<br />

15. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS. <strong>CT</strong> IN THE STAGING AND<br />

EVALUATING RESPONSE TO TREATMENT OF GASTROINTESTINAL STROMAL TUMOR......... 213<br />

15.1. Results <strong>of</strong> primary trial search .................................................................................................... 213<br />

15.2. Population characteristics .......................................................................................................... 213<br />

15.3. Description <strong>of</strong> intervention.......................................................................................................... 213<br />

15.3.1. <strong>PET</strong>-<strong>CT</strong> imaging...........................................................................................................................213<br />

15.3.2. Diagnostic technology compared ........................................................................................214<br />

15.3.3. Reference test ............................................................................................................................215<br />

15.4. Findings ......................................................................................................................................... 215<br />

15.4.1. Staging.........................................................................................................................................215<br />

15.4.2. Response to treatment .............................................................................................................215<br />

15.4.3. Safety ...........................................................................................................................................216<br />

15.5. Results............................................................................................................................................ 216<br />

16. COMPARATIVE EFFICACY ANALYSIS OF THE <strong>PET</strong>-<strong>CT</strong> AND <strong>CT</strong> IN ASSESSING RESIDUAL<br />

LESIONS IN COLORE<strong>CT</strong>AL LIVER METASTASIS ..................................................................... 218<br />

16.1. Results <strong>of</strong> primary trial search .................................................................................................... 218<br />

16.2. Population characteristics .......................................................................................................... 218<br />

16.3. Description <strong>of</strong> intervention.......................................................................................................... 218


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

16.3.1. <strong>PET</strong>-<strong>CT</strong> imaging...........................................................................................................................218<br />

16.3.2. Diagnostic technology compared ........................................................................................219<br />

16.3.3. Reference test ............................................................................................................................220<br />

16.4. Findings ......................................................................................................................................... 220<br />

16.5. Results............................................................................................................................................ 221<br />

17. COMPARATIVE ANALYSIS OF THE EFFICACY OF <strong>PET</strong>-<strong>CT</strong> VS OTHER IMAGING<br />

TECHNIQUES (MRI OR <strong>CT</strong>) IN THE STAGING OF NEOPLASM IN VARIED LOCATIONS........ 222<br />

17.1. Neoplasm in varied locations – disease staging (<strong>PET</strong>-<strong>CT</strong> vs. MRI) ......................................... 222<br />

17.1.1. Results <strong>of</strong> trial search .................................................................................................................222<br />

17.1.2. Population characteristics .......................................................................................................222<br />

17.1.3. Description <strong>of</strong> intervention.......................................................................................................223<br />

17.1.3.1 <strong>PET</strong>-<strong>CT</strong> ..................................................................................................................................................... 223<br />

17.1.3.2 Compared diagnostic test ................................................................................................................. 224<br />

17.1.3.3 Reference test ..................................................................................................................................... 225<br />

17.1.4. Findings ........................................................................................................................................226<br />

17.1.4.1 Diagnostic accuracy in detecting primary and recurring carcinomas..................................... 226<br />

17.1.4.2 T-staging................................................................................................................................................. 227<br />

17.1.4.3 N-staging................................................................................................................................................ 231<br />

17.1.4.4 Assessment <strong>of</strong> distant metastases (M feature)................................................................................ 235<br />

17.1.4.5 TNM staging system.............................................................................................................................. 240<br />

17.1.4.6 Impact on therapy............................................................................................................................... 243<br />

17.1.4.7 Safety...................................................................................................................................................... 244<br />

17.1.5. Results...........................................................................................................................................244<br />

17.2. Neoplasm in varied locations: disease staging(<strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong>)........................................ 246<br />

17.2.1. Results <strong>of</strong> trial search .................................................................................................................246<br />

17.2.2. Population characteristics .......................................................................................................246<br />

17.2.3. Description <strong>of</strong> intervention.......................................................................................................247<br />

17.2.3.1 <strong>PET</strong>-<strong>CT</strong> ..................................................................................................................................................... 247<br />

17.2.3.2 Diagnostic trial compared.................................................................................................................. 248<br />

17.2.3.3 Reference test ...................................................................................................................................... 248<br />

17.2.4. Findings ........................................................................................................................................249<br />

17.2.4.1 T-staging................................................................................................................................................. 249<br />

17.2.4.2 N-staging................................................................................................................................................ 249<br />

17.2.4.3 Assessment <strong>of</strong> distant metastasis....................................................................................................... 250<br />

17.2.4.4 TNM staging system.............................................................................................................................. 252<br />

17.2.4.5 Impact on therapy............................................................................................................................... 253<br />

17.2.4.6 Safety...................................................................................................................................................... 254<br />

17.2.5. Results...........................................................................................................................................254<br />

17.3. Cancers <strong>of</strong> unknown primary origin: diagnostic efficacy ...................................................... 255<br />

13


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

17.3.1. Results <strong>of</strong> trial search .................................................................................................................255<br />

17.3.2. Population characteristics .......................................................................................................255<br />

17.3.3. Description <strong>of</strong> intervention.......................................................................................................256<br />

14<br />

17.3.3.1 <strong>PET</strong>-<strong>CT</strong> ..................................................................................................................................................... 256<br />

17.3.3.2 Diagnostic test compared.................................................................................................................. 257<br />

17.3.3.3 Reference test ...................................................................................................................................... 258<br />

17.3.4. Findings ........................................................................................................................................258<br />

17.3.4.1 Diagnostic accuracy in detecting primary cancer <strong>of</strong> unknown origin...................................... 258<br />

17.3.4.2 Safety...................................................................................................................................................... 262<br />

17.3.5. Results...........................................................................................................................................262<br />

18. APPENDICES ........................................................................................................................ 264<br />

18.1. Tool for quality assessment <strong>of</strong> studies........................................................................................ 264<br />

18.2. Detailed search results................................................................................................................ 290<br />

19. LITERATURE USED IN THE ANALYSIS ..................................................................................... 296<br />

19.1. Lung cancer ................................................................................................................................. 296<br />

19.2. Neoplasm in varied locations .................................................................................................... 296<br />

19.3. Lymphomas .................................................................................................................................. 296<br />

19.4. Esophageal cancer ..................................................................................................................... 297<br />

19.5. Cancer in the female genitals.................................................................................................... 297<br />

19.6. Ovarian cancer............................................................................................................................ 297<br />

19.7. Thyroid gland cancer .................................................................................................................. 297<br />

19.8. Head and neck cancer .............................................................................................................. 297<br />

19.9. Pancreatic cancer....................................................................................................................... 298<br />

19.10. Sarcomas ...................................................................................................................................... 298<br />

19.11. Colon cancer ............................................................................................................................... 298<br />

20. TRIALS EXCLUDED FROM THE ANALYSIS ............................................................................. 300<br />

21. TABLE OF ILLUSTRATIONS..................................................................................................... 321<br />

22. TABLE OF GRAPHS ............................................................................................................... 326<br />

23. TABLE OF FIGURES ............................................................................................................... 328


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

1. SUMMARY<br />

AIM OF STUDY<br />

The aim <strong>of</strong> this report is a comparative cost-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong> technology (<strong>positron</strong> <strong>emission</strong><br />

tomography - <strong>PET</strong> fused with computed tomography - <strong>CT</strong>) and the diagnostic technologies financed from public<br />

sources in oncological diagnostics in Poland. This part <strong>of</strong> the report focuses on a clinical and epidemiological<br />

<strong>analysis</strong>.<br />

The study was prepared for the Agency for Health Technology Assessment in Poland.<br />

METHODOLOGY<br />

Clinical efficacy evaluation methods<br />

The search strategy for primary studies, the evaluation <strong>of</strong> studies reliability, as well as data extraction,<br />

statistical <strong>analysis</strong> and results interpretation methods were designed based on guidelines <strong>of</strong> the Medical Services<br />

Advisory Committee [„Guidelines for the assessment <strong>of</strong> diagnostic technologies” August 2005, Australia].<br />

Search strategy for primary studies<br />

As the first step <strong>of</strong> searching for trials a general search strategy was adopted without further specifying the<br />

target population, in order to locate a group <strong>of</strong> trials concerning the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in oncological diagnostics.<br />

Additionally, for 12 indications individual searches were conducted with targeted population (breast cancer, lung<br />

cancer, colorectal cancer, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, gastric and<br />

esophageal cancer, melanoma, glioma, sarcoma, head and neck cancer). In the process <strong>of</strong> searching for reports<br />

on the technology under assessment, results <strong>of</strong> a high specificity strategy that identified only those publications<br />

where the hybrid term “<strong>PET</strong>-<strong>CT</strong>” or its equivalents appeared (#8), were combined with results <strong>of</strong> a high sensitivity<br />

strategy that identified all publications describing the diagnostic method under discussion (search for publications<br />

with “<strong>PET</strong>” and “<strong>CT</strong>” titles, abstracts or keywords - #8).<br />

ID search strategy<br />

#1 „Positron-Emission Tomography” [MeSH] OR „Tomography, Emission-Computed” [MeSH]<br />

#2 <strong>PET</strong> OR <strong>positron</strong> <strong>emission</strong> tomography<br />

#3 #1 OR #2<br />

#4 („Tomography, Spiral Computed” [MeSH] OR „Tomography, X-Ray Computed” [MeSH])<br />

#5 <strong>CT</strong> OR computed tomography<br />

#6 #4 OR #5<br />

#7<br />

dual OR integral OR integration OR combination OR combined OR fusion OR fused OR hybrid OR coincidental OR<br />

combining OR coincidence OR coregistered<br />

#8 #3 AND #6<br />

#9 #3 AND #7<br />

#10 #8 OR #9<br />

15


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

#11 „Neoplasms” [MeSH]<br />

#12 „Medical Oncology” [MeSH]<br />

#13 #11 OR #12<br />

#14 #10 AND #13<br />

#15 #10 AND #13 Limits: English, Polish, Publication Date from 1998/01/01, Humans<br />

Last search date: 20 Mar 2006<br />

Search in medical databases<br />

The above search strategy was applied to the following databases:<br />

Medline via Pubmed,<br />

Cochrane Library (The Cochrane Database <strong>of</strong> Systematic Reviews, The Cochrane Controlled Trials Register),<br />

EmBase,<br />

And medical web sites:<br />

16<br />

• NICE (National Institute for Clinical Excellence),<br />

SBU (Statens beredning för medicinsk utvärdering),<br />

NCCHTA (The National Coordinating Centre for Health Technology Assessment),<br />

CADTH (The Canadian Agency for Drugs and Technologies in Health),<br />

INAHTA (International Network <strong>of</strong> Agencies for Health Technology Assessment),<br />

MSAC (Medical Services Advisory Committee).<br />

Also, the bibliographies <strong>of</strong> the primary studies were searched. Secondary studies were reviewed (object articles,<br />

systematic reviews, meta-analyses, agency reviews) for possible additional primary studies. Moreover, to make the list<br />

<strong>of</strong> publications complete, manufacturers <strong>of</strong> diagnostic equipment were requested to provide all their studies,<br />

especially those concerning the pre-marketing period, as well as marketing information on <strong>PET</strong>-<strong>CT</strong> scanners.<br />

The search was conducted independently by two researchers. Trial reviews were also reviewed by two persons.<br />

Criteria for the inclusion <strong>of</strong> primary studies in the <strong>analysis</strong><br />

Initially, titles and abstracts in the publications found were analyzed. If information on the use <strong>of</strong> <strong>PET</strong> (including <strong>PET</strong>-<br />

<strong>CT</strong>) imaging or <strong>CT</strong> in a trial was found, it was qualified for further verification depending on how the full text was<br />

evaluated.<br />

As the second stage, full texts in an electronic version were analyzed with a special focus on trial methodology, in<br />

order to identify information on the use <strong>of</strong> hybrid <strong>PET</strong>-<strong>CT</strong> scanners. Reports where the use <strong>of</strong> the technique under<br />

discussion was confirmed were further verified. Based on the guidelines <strong>of</strong> the Medical Services Advisory Committee,<br />

the verification process focused on the selection <strong>of</strong> trials, where two strategies were assessed at the same time: <strong>PET</strong>-<br />

<strong>CT</strong> and another strategy financed in Poland. On this basis a decision was made whether to include or exclude a trial,<br />

stating the reasons.<br />

In case <strong>of</strong> primary studies, there were no restrictions on the period <strong>of</strong> follow-up or size <strong>of</strong> population. Trials done on<br />

humans, for which full texts <strong>of</strong> reports are accessible in Polish libraries in English or Polish were included. Restrictions<br />

were defined for the date <strong>of</strong> publication, which narrowed down the scope <strong>of</strong> search to studies published after 1 Jan<br />

1998. This criterion was accommodated based on the results <strong>of</strong> database searches (publication <strong>of</strong> first reports on the


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

hybrid <strong>PET</strong>-<strong>CT</strong> scanner) and the date <strong>of</strong> launching the technology under assessment on the diagnostic procedures<br />

market.<br />

Population<br />

The population were patients diagnosed for oncological indications.<br />

Intervention<br />

The <strong>analysis</strong> included primary prospective or retrospective clinical trials, where <strong>PET</strong>-<strong>CT</strong> results were assessed<br />

(regardless <strong>of</strong> the type <strong>of</strong> radiopharmaceutical used). The essential criterion for the inclusion <strong>of</strong> a publication into the<br />

<strong>analysis</strong> was whether the diagnostic techniques compared were verified using an independent reference test (gold<br />

standard).<br />

Technologies compared (comparators)<br />

Diagnostic technologies financed in Poland from public sources and used in oncological diagnostics.<br />

End points<br />

• Diagnostic efficacy <strong>of</strong> testing methods in comparison to the respective reference method or clinical<br />

observation;<br />

• Change <strong>of</strong> therapeutic decision;<br />

• Impact on clinical end points;<br />

• Safety.<br />

Trial quality assessment<br />

The adopted scale <strong>of</strong> trial quality helps evaluate publications with regard to whether they are properly designed<br />

and conducted, and ensures reliability <strong>of</strong> results.<br />

In compliance with EBM requirements (Evidence Based Medicine), the process <strong>of</strong> trial quality assessment used the<br />

QUADAS form, which consisted <strong>of</strong> 14 questions. The first four questions check whether the choice <strong>of</strong> population was<br />

right; questions 5-9 assess whether the measurements and the reference test were executed correctly, the remaining<br />

questions rated the interpretation and presentations <strong>of</strong> the results.<br />

In order to standardize the assessment procedure, a single form was prepared to verify all the publications<br />

included in the <strong>analysis</strong>.<br />

Each study was rated independently by two analysts. In case <strong>of</strong> discrepancies, the final decision was made<br />

through formal consensus.<br />

Each question was evaluated “yes”, “no”, or “unclear”. Checklists <strong>of</strong> trial quality assessment for each study are given<br />

in an appendix hereto.<br />

Statistical <strong>analysis</strong><br />

Included in the <strong>analysis</strong> are trials that sought information on the results <strong>of</strong> the diagnostic methods under assessment<br />

in comparison with a reference method. The results were grouped according to the parameters <strong>of</strong> a four-field table:<br />

TP, TN, FP and FN.<br />

17


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

18<br />

Next, in order to assess the diagnostic efficacy, parameters such as sensitivity (Se), specificity (Sp), accuracy (Acc),<br />

positive likelihood ratio (LR+), negative likelihood ratio (LR-) and diagnostic odds ratio (DOR) were calculated with a<br />

95% confidence interval. Whenever possible, a meta-<strong>analysis</strong> <strong>of</strong> results <strong>of</strong> studies was also conducted. To evaluate<br />

differences in diagnostic efficacy <strong>of</strong> trials, McNemar’s test was done, provided data were available (test <strong>of</strong><br />

proportions with matched data array).<br />

For a comparative rating <strong>of</strong> odds <strong>of</strong> accurate diagnosis (consistent with the reference standard) for the diagnostic<br />

methods under assessment, the OR parameters statistical significance was rated based on EBM guidelines. For<br />

statistically significant results, the NNT parameter was additionally calculated, specifying confidence intervals.<br />

When no statistically significant discrepancies <strong>of</strong> study results were identified, a meta-<strong>analysis</strong> <strong>of</strong> the results was<br />

conducted using the fixed effect model. For end points that were statistically significant in a heterogeneity test, the<br />

random effect model was used. The statistical s<strong>of</strong>tware MetaDiSc v.1.3 was used to calculate these parameters and<br />

to carry out a meta-analyses <strong>of</strong> sensitivity and specificity. The meta-<strong>analysis</strong> <strong>of</strong> accuracy, OR and NNT, and graphs<br />

representing this results were done in Stats Direct v. 2.5.2.<br />

Conflict is interest<br />

None <strong>of</strong> the authors <strong>of</strong> the study reported any conflict <strong>of</strong> interest.<br />

Results<br />

Lung cancer – T-staging (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>)<br />

As a result <strong>of</strong> searching medical databases 4 primary prospective trials were identified (Antoch 2003, Lardinois<br />

2003, Cerfolio 2005 and Shim 2005), where the <strong>PET</strong>-<strong>CT</strong> method was compared to <strong>CT</strong> in patients suffering from non-<br />

small cell lung cancer (N=570). Both trials were verified histopathologically, however in case <strong>of</strong> metastasis clinical<br />

follow-up, MRI as well as ultrasonography was used. Indications for scanning were based on results <strong>of</strong> T-staging.<br />

In the assessment <strong>of</strong> T-status in all the trials under assessment, the proportion <strong>of</strong> correctly diagnosed patients is<br />

higher when <strong>PET</strong>-<strong>CT</strong> is adopted in comparison to <strong>CT</strong> only. The diagnostic accuracy calculated by meta-<strong>analysis</strong> is<br />

86% (95% CI: 81; 91) for <strong>PET</strong>-<strong>CT</strong> and 71% (95% CI: 55; 84) for <strong>CT</strong>.<br />

Calculated by meta-<strong>analysis</strong> <strong>of</strong> three studies, the odds ratio <strong>of</strong> consistency between the reference test and T-<br />

staging results for <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong> is 2.42 (95% CI: 1.36; 4.29). The value is statistically significant; NNT = 8 (95% CI: 5; 20).<br />

Based on the analyses <strong>of</strong> all the diagnostic parameters established (sensitivity, specificity, accuracy), <strong>PET</strong>-<strong>CT</strong> is<br />

more efficacious than <strong>CT</strong> in rating the involvement <strong>of</strong> lymph nodes (N-status). The diagnostic accuracy calculated<br />

by meta-<strong>analysis</strong> is 85% (95% CI: 76; 93) for <strong>PET</strong>-<strong>CT</strong> and 61% (95% CI: 49; 72) for <strong>CT</strong>. Sensitivity is 85-89%, for <strong>PET</strong>-<strong>CT</strong> and<br />

70% for <strong>CT</strong>, and specificity is 84-94% for <strong>PET</strong>-<strong>CT</strong> and 59-69% for <strong>CT</strong>.<br />

The odds <strong>of</strong> N-staging results being consistent with the reference test is nearly 4 times higher for <strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong><br />

alone, and the value is statistically significant: OR=3.95 (95% CI: 1.65; 9.44); NNT = 4 (95% CI: 3; 10).<br />

The accuracy <strong>of</strong> the TNM staging system evaluated by meta-<strong>analysis</strong> is 88% (95% CI: 83; 93) for <strong>PET</strong>-<strong>CT</strong> and 67%<br />

(95% CI: 59; 74) for <strong>CT</strong>.<br />

The odds <strong>of</strong> obtaining small cell lung cancer TNM staging results consistent with the reference test is higher for <strong>PET</strong>-<br />

<strong>CT</strong> versus <strong>CT</strong> alone, and the differences observed are statistically significant; OR = 3.91 (95% CI: 2.04; 7.50); NNT = 5<br />

(95% CI: 4; 9).<br />

In summary, the <strong>PET</strong>-<strong>CT</strong> method has a higher diagnostic efficacy than <strong>CT</strong> in the staging <strong>of</strong> clinical non-small cell<br />

lung cancer (NSCLC).


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Neoplasms in varied locations<br />

1. Disease Staging (<strong>PET</strong>-<strong>CT</strong> vs. MRI)<br />

Two primary clinical trials (Antoch 2003 and Schmidt 2005) were found, where the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in<br />

the staging <strong>of</strong> neoplasms with varied locations was directly compared to magnetic resonance imaging – MRI<br />

(N=136). Both trials were qualified based on a reference methods, which in this part <strong>of</strong> the study were<br />

histopathological examination and/or clinical follow-up.<br />

A single-trial-based rating <strong>of</strong> the efficacy <strong>of</strong> primary and secondary carcinoma detection showed that the<br />

sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is higher in comparison to magnetic resonance imaging: 100% (95% CI: 59; 100) vs. 86% (95% CI:<br />

42; 100). The specificity <strong>of</strong> both methods was 100%. The differences between groups were not statistically significant.<br />

For T-staging, the diagnostic accuracy established by meta-<strong>analysis</strong> <strong>of</strong> two trials was 89% (95% CI: 68; 100) for <strong>PET</strong>-<br />

<strong>CT</strong> and 79% (95% CI: 26; 99) for MRI. Calculated by meta-<strong>analysis</strong>, the odds ratio for T-staging results being consistent<br />

with the reference test using <strong>PET</strong>-<strong>CT</strong> versus MRI, is 3.29 (95% CI: 1.37; 7.90), and the value is statistically significant;<br />

NNT=7 (95% CI: 4; 20).<br />

Analysis showed that the <strong>PET</strong>-<strong>CT</strong> method was more accurate compared to MRI in the assessment <strong>of</strong> the<br />

involvement <strong>of</strong> lymph nodes by cancer metastasis (N status). The accuracy established by meta-<strong>analysis</strong> is 94% (95%<br />

CI: 89; 97) for <strong>PET</strong>-<strong>CT</strong>, as compared to 80% for MRI (95% CI: 73; 86). The odds <strong>of</strong> the staging results for the involvement<br />

<strong>of</strong> lymph nodes by cancer metastasis being consistent with the reference test were four times higher for the <strong>PET</strong>-<strong>CT</strong><br />

method than for magnetic resonance. The result is statistically significant; OR = 4.00 (95% CI: 1.74; 9.19); NNT = 8 (95%<br />

CI: 5; 17). The diagnostic sensitivity reported by the authors was 95% and 100% for <strong>PET</strong>-<strong>CT</strong>, and 79% for MRI, while<br />

specificity is 92%-95% for <strong>PET</strong>-<strong>CT</strong> and 78%-95% for MRI.<br />

Established by meta-<strong>analysis</strong> <strong>of</strong> two trials, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong> in distant metastasis staging (M<br />

status) is 94% (95% CI: 90; 98) for <strong>PET</strong>-<strong>CT</strong> and 96% (95% CI: 86; 100) for magnetic resonance. The odds <strong>of</strong> obtaining<br />

reference method-consistent staging results for distant metastasis using the <strong>PET</strong>-<strong>CT</strong> method are the same as the odds<br />

for magnetic resonance: OR = 1.00 (95% CI: 0.34; 2.95). The result is statistically significant. Based on two trials included<br />

in this <strong>analysis</strong>, sensitivity was 93% and 100% for <strong>PET</strong>-<strong>CT</strong>, and was 90% and 100% for MRI. Specificity was 95% and 98%<br />

for <strong>PET</strong>-<strong>CT</strong>, and 95% and 100% for MRI.<br />

A review <strong>of</strong> the diagnostic accuracy <strong>of</strong> the methods in total staging <strong>of</strong> cancer (TNM staging) was conducted by<br />

meta-<strong>analysis</strong> <strong>of</strong> two trials, which showed that the accuracy is higher for <strong>PET</strong>-<strong>CT</strong> than for MRI. The values are 86%<br />

(95% CI: 65; 98) and 75% (95% CI: 33; 99) respectively. The odds <strong>of</strong> obtaining results consistent with reference test are<br />

2.61 times higher for <strong>PET</strong>-<strong>CT</strong> than for MRI. The result is statistically significant; OR = 2.61 (95% CI: 1.46; 4.67); NNT = 6 (95%<br />

CI: 4; 15).<br />

The use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> MRI in the staging <strong>of</strong> cancer was conducive to changes in therapeutic<br />

recommendations in 12% <strong>of</strong> patients suffering from cancer with varied locations, while the use <strong>of</strong> MRI instead <strong>of</strong> <strong>PET</strong>-<br />

<strong>CT</strong> was conducive to changes in therapeutic recommendations in 2% <strong>of</strong> patients.<br />

In summary, in patients with various cancer locations, the <strong>PET</strong>-<strong>CT</strong> method shows higher diagnostic efficacy in<br />

comparison to MRI in detecting primary and recurring cancer, as well as in the staging <strong>of</strong> tumor (T-status), lymph<br />

nodes involvement (N-status), and distant metastasis (M-status) and combined staging (TNM staging).<br />

2. Cancer staging (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>)<br />

One primary clinical trial (Antoch 2004) was identified, where the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in the staging <strong>of</strong><br />

neoplasm in varied locations was compared directly with <strong>CT</strong> (N=260). Both testing methods were verified against the<br />

reference methods, i.e. histopathological tests and/or clinical follow-up.<br />

The diagnostic accuracy <strong>of</strong> T-staging was 82% (95% CI: 71; 90) for <strong>PET</strong>-<strong>CT</strong>, and 66% (95% CI: 55; 77) for <strong>CT</strong>. The<br />

differences between the groups under discussion are statistically significant (p = 0.0018). The odds <strong>of</strong> obtaining a T-<br />

19


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

status consistent with the reference method is 2.29 times higher for <strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong>. The result is statistically<br />

significant; OR = 2.29 (95% CI: 1.03; 5.25), NNT = 7 (95% CI: 4; 59).<br />

20<br />

Analysis showed that the <strong>PET</strong>-<strong>CT</strong> method is statistically significantly more accurate than <strong>CT</strong> in the staging <strong>of</strong> the<br />

involvement <strong>of</strong> lymph nodes (N status). The diagnostic accuracy is 92% (95% CI: 88; 95) for <strong>PET</strong>-<strong>CT</strong> and 76% (95% CI:<br />

70; 81) for <strong>CT</strong>. The odds <strong>of</strong> obtaining lymph nodes involvement staging results consistent with the reference test is<br />

nearly four times higher for the <strong>PET</strong>-<strong>CT</strong> procedure versus <strong>CT</strong>. The result is statistically significant; OR = 3.84 (95% CI: 2.19;<br />

6.93), NNT = 7 (95% CI: 5; 10).<br />

The diagnostic accuracy <strong>of</strong> the methods compared in the staging <strong>of</strong> distant metastasis (M status) is 95% (95% CI:<br />

92; 98) for <strong>PET</strong>-<strong>CT</strong> and 88% (95% CI: 84; 92) for <strong>CT</strong>, and the differences between the methods are statistically<br />

significant (p=0.0001). The odds <strong>of</strong> M-staging results being consistent with the reference method is 2.7 times higher for<br />

<strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong>. The result is statistically significant; OR = 2.70 (95% CI: 1.30; 5.92), NNT = 15 (95% CI: 7; 44).<br />

Sensitivity, specificity and accuracy are higher for <strong>PET</strong>-<strong>CT</strong> compared to <strong>CT</strong>, including in staging metastasis to lungs,<br />

liver and bones, as well as organs total.<br />

Analysis showed that the diagnostic accuracy <strong>of</strong> the methods under assessment in cancer staging (TNM staging) is<br />

higher when the <strong>PET</strong>-<strong>CT</strong> method is used compared to <strong>CT</strong>. The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 84% (95% CI: 79; 88), and for <strong>CT</strong> it<br />

is 63% (95% CI: 57; 69). The odds <strong>of</strong> obtaining staging results consistent with the reference standard is more than three<br />

times higher for <strong>PET</strong>-<strong>CT</strong> compared to <strong>CT</strong>. The result is statistically significant, OR = 3,09 (95% CI 2.00; 4.80), NNT = 5 (95%<br />

CI: 4; 8).<br />

Using <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> <strong>CT</strong> in disease staging led to a different recommendation for further treatment in 15%<br />

patients. Using <strong>CT</strong> instead <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> caused therapeutic decisions to be changed for 0.8% patients.<br />

3. Detecting primary origin <strong>of</strong> cancer in metastasis patients (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>)<br />

Two primary clinical trials were found that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with that <strong>of</strong> <strong>CT</strong> in<br />

locating primary neoplasm in metastasis patients. Both testing methods were verified against a reference test<br />

(histopathological examination and/or clinical observation).<br />

Based on a meta-<strong>analysis</strong>, for patients with metastasis in the neck area the diagnostic sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is higher<br />

than the values established for <strong>CT</strong>: 47% (95% CI: 31; 64) vs. 26% (95% CI: 13; 44). Another meta-<strong>analysis</strong> showed that<br />

the odds <strong>of</strong> determining the origin <strong>of</strong> primary neoplasm in a population <strong>of</strong> patients with metastasis in the neck area<br />

using <strong>PET</strong>-<strong>CT</strong> was almost three times higher compared to <strong>CT</strong>, and the result was statistically significant; OR = 2.87 (95%<br />

CI: 1.08; 7.63), NNT = 5 (95% CI: 3; 35).<br />

A single-trial <strong>analysis</strong> <strong>of</strong> the diagnostic efficacy <strong>of</strong> the technologies compared in locating primary tumor in patients<br />

with cancer metastasis outside the neck indicated no statistically significant differences between <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong>. The<br />

diagnostic sensitivity <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong> method was higher in versus <strong>CT</strong>, and stood at 36% (95% CI: 18; 57) vs. 15% (95% CI:<br />

4; 35).<br />

The results <strong>of</strong> a single trial also warranted the claim that the diagnostic sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> proved higher than the<br />

sensitivity <strong>of</strong> <strong>CT</strong> in locating cancer focuses in the total population <strong>of</strong> patients with cancer metastasis both in and<br />

outside the neck, and was 36% (95% CI: 22; 52) for <strong>PET</strong>-<strong>CT</strong> vs. 19% (95% CI: 9; 34) for <strong>CT</strong>.<br />

Lymphoma staging and restaging (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>)<br />

As a result <strong>of</strong> searching medical databases two primary studies were found: Freudenberg 2003 (N = 27) and<br />

Schaefer 2004 (N = 60), which compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in lymphoma staging and<br />

restaging. The reference tests were histopathological tests, clinical follow-up and conventional staging <strong>of</strong> the disease<br />

(clinical and laboratory tests, USG, MRI, scintigraphy).<br />

A diagnostic efficacy review showed that <strong>PET</strong>-<strong>CT</strong> was diagnostically more efficacious than <strong>CT</strong> both for lymphatic<br />

and extra-lymphatic locations individually, and for the total cohort.<br />

In staging lesions in the lymphical area, the sensitivity and accuracy <strong>of</strong> both <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> were 100% (95% CI: 82;<br />

100).


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

In restaging, <strong>PET</strong>-<strong>CT</strong> sensitivity stood at 85% (95% CI: 55; 98) and specificity at 100% (95% CI: 88; 100); both were<br />

higher than the respective parameters for <strong>CT</strong>. For <strong>CT</strong> the values were: 69% for sensitivity (95% CI: 39; 91) and 86% for<br />

specificity (95% CI: 67; 96). The diagnostic accuracy <strong>of</strong> the methods discussed was 95% (95% CI: 83; 99) for <strong>PET</strong>-<strong>CT</strong> and<br />

80% (95% CI: 65; 91) for <strong>CT</strong>.<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity and specificity in detecting pathological lesions located outside <strong>of</strong> lymph nodes stood at 75%<br />

(95% CI: 19; 99) and 100% (95% CI: 78; 100), respectively. In restaging, both sensitivity and specificity were 100%. With<br />

<strong>CT</strong> imaging, the values were: 25% for sensitivity (95% CI: 1; 81) and 100% for specificity (95% CI: 78; 100) for staging,<br />

and 75% (95% CI: 19; 99) and 86% (95% CI: 71; 95) respectively for restaging. <strong>PET</strong>-<strong>CT</strong> scanning demonstrated higher<br />

accuracy (95% for staging and 100% for restaging) compared to <strong>CT</strong> (84% and 85% respectively).<br />

Meta-<strong>analysis</strong> <strong>of</strong> the tests included in this study showed that in disease staging in patients with lymphoma, <strong>PET</strong>-<strong>CT</strong> is<br />

characterized by higher sensitivity and specificity compared to <strong>CT</strong>. The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 93% (95% CI: 82; 98), vs.<br />

80% for <strong>CT</strong> (95% CI: 66; 89). The specificity <strong>of</strong> the diagnostic methods discussed is 100% (95% CI: 96; 100) and 84% (95%<br />

CI: 75; 91) respectively.<br />

<strong>PET</strong>-<strong>CT</strong> is characterizes by a higher accuracy <strong>of</strong> 97% (95% CI: 93; 99) s. 78% for <strong>CT</strong> (95% CI: 57; 93).<br />

Freudenberg 2003 analyses the consistency between the reference test and staging based on the Ann Arbor<br />

system. Disease was staged correctly by <strong>PET</strong>-<strong>CT</strong> in 26 (96%) patients, and by <strong>CT</strong> in 13 (48%) patients. The difference<br />

between <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> was statistically significant (p=0.002). The odds <strong>of</strong> obtaining the correct staging were 28 times<br />

for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>: OR = 28 (95% CI: 3.35; 1227.94); NNT = 3 (95% CI: 2; 4).<br />

<strong>PET</strong>-<strong>CT</strong> imaging is characterized by higher diagnostic efficacy than <strong>CT</strong> in the staging <strong>of</strong> lymphomas.<br />

Esophageal cancer<br />

1. Staging and response to treatment (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>, EUS<br />

As a result <strong>of</strong> searching medical databases one primary study was found (Cerfolio 2005) that evaluated the<br />

diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong> and transesophageal endoscopic ultrasound-guided biopsy (EUS) in the<br />

restaging <strong>of</strong> esophageal cancer and the assessment <strong>of</strong> response to treatment.<br />

In the diagnostics <strong>of</strong> primary tumor (T status), the highest diagnostic accuracy was demonstrated by <strong>PET</strong>-<strong>CT</strong>: 83%<br />

(95% CI: 68; 93) for stage T0, 80% (95% CI: 65; 91) for stages T1-T3, and 98% (95% CI: 87; 100) for stage T4. In the other<br />

tests under assessment, the accuracy <strong>of</strong> <strong>CT</strong> was 61% (95% CI: 45; 76), 56% (95% CI: 40; 72) and 95% (95% CI: 83; 99)<br />

respectively, and the accuracy <strong>of</strong> EUS was 66% (95% CI: 49; 80), 56% (95% CI: 40; 72) and 90% (95% CI: 77; 97).<br />

<strong>PET</strong>-<strong>CT</strong> scanning allowed for correct (consistent with reference test) T-staging in 80% patients, while <strong>CT</strong> and EUS - in<br />

56% each.<br />

The odds <strong>of</strong> obtaining a correct T-status by using <strong>PET</strong>-<strong>CT</strong> is 3.23 times higher compared to <strong>CT</strong> alone, and to EUS;<br />

OR = 3.23 (95% CI: 1.09; 10.00), both for the comparison <strong>PET</strong>-<strong>CT</strong> vs. EUS and for the comparison <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>; NNT = 5<br />

(95% CI: 3; 24).<br />

In staging lymph nodes involvement (N status), the highest diagnostic accuracy was achieved for <strong>PET</strong>-<strong>CT</strong>: 93% (95%<br />

CI: 80; 98), compared to 78% (95% CI: 62; 80) for <strong>CT</strong> and 78% (95% CI: 62; 80) for EUS. The difference in accuracy was<br />

statistically significant (p=0.04).<br />

The diagnostic efficacy <strong>of</strong> trials in M-staging was evaluated separately for stages M1a and M1b.<br />

The highest accuracy in determining stage M1a metastasis was demonstrated by EUS – 92% (95% CI: 80; 98), while<br />

the accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 90% (95% CI: 77; 97), and the accuracy <strong>of</strong> <strong>CT</strong> was 88% (95% CI: 75; 95). The differences<br />

between the groups were statistically insignificant. The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and EUS was 33% (95% CI: 4; 78), and the<br />

sensitivity <strong>of</strong> <strong>CT</strong> imaging was 0% (95% CI: 0; 40). The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 98% (95% CI: 87; 100), while that <strong>of</strong> <strong>CT</strong><br />

imaging and EUS is 100%.<br />

In the diagnostics <strong>of</strong> stage M1b metastasis, the sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 67% (95% CI: 22; 96) and was higher than<br />

the sensitivity <strong>of</strong> <strong>CT</strong>, which stood at 50% (95% CI: 12; 88).<br />

21


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

22<br />

In the assessment <strong>of</strong> complete response to treatment, <strong>PET</strong>-<strong>CT</strong> showed the highest sensitivity <strong>of</strong> 87% (95% CI: 60; 98).<br />

The sensitivity <strong>of</strong> <strong>CT</strong> was 27% (95% CI: 8; 55), and the sensitivity <strong>of</strong> EUS was 20% (95% CI: 4; 48). The differences<br />

between <strong>PET</strong>-<strong>CT</strong> and the methods compared are statistically significant. The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning stood at<br />

88% (95% CI: 72; 97), <strong>of</strong> <strong>CT</strong> at 91% (95% CI: 76; 98), and <strong>of</strong> EUS-FNA at 94% (95% CI: 80; 99). The highest accuracy was<br />

demonstrated by <strong>PET</strong>-<strong>CT</strong>, and stood at 88% % (95% CI: 75; 95) vs. 71% (95% CI: 56; 83) for <strong>CT</strong> and 71% (95% CI: 56; 83)<br />

for EUS. The differences between <strong>PET</strong>-<strong>CT</strong> and the methods compared are statistically significant (p=0.045 for the<br />

comparison with EUS-FNA, and p=0.05 for the comparison with <strong>CT</strong>).<br />

In summary, in restaging esophageal cancer, and in determining complete response to treatment, <strong>PET</strong>-<strong>CT</strong> imaging<br />

demonstrates higher diagnostic efficacy than <strong>CT</strong> or trans-esophageal ultrasound-guided biopsy.<br />

2. The diagnostics <strong>of</strong> primary esophageal cancer (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>)<br />

As a result <strong>of</strong> searching medical databases one primary trial was found (Kula 2005), which compared the<br />

diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with that <strong>of</strong> <strong>CT</strong> in the staging <strong>of</strong> esophageal cancer (N=12). The reference test was a<br />

histopathological or cytological test.<br />

Based on studies accessible, <strong>PET</strong>-<strong>CT</strong> is characterized by higher sensitivity (100%) in comparison to <strong>CT</strong> (92%) in<br />

detecting esophageal cancer.<br />

Cancer <strong>of</strong> the female genitals<br />

1. Female genital cancer: staging and assessment <strong>of</strong> disease recurrence (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>, MRI,<br />

ultrasonography)<br />

As a result <strong>of</strong> searching medical databases one primary clinical trial was found (Grisaru 2004) that compared<br />

directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> vs. conventional imaging methods (<strong>CT</strong>, magnetic resonance and<br />

ultrasonography) in the diagnostics <strong>of</strong> female genital cancer (N=53). Positive results <strong>of</strong> scanning were verified by<br />

histopathological tests <strong>of</strong> material removed during a surgical procedure or obtained from guided biopsy. Negative<br />

results were verified based on long-term clinical observation and repeated diagnostic imaging.<br />

The assessment <strong>of</strong> the diagnostic efficacy <strong>of</strong> imaging methods discussed included both staging and detecting<br />

recurrences <strong>of</strong> disease.<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostics in clinical staging is higher than the sensitivity <strong>of</strong> the conventional diagnostic<br />

imaging and stands at 100% (95% CI: 66; 100) vs. 56% (95% CI: 21; 86). The difference is statistically insignificant<br />

(p=0.13). The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 100% (95% CI: 66; 100), and that <strong>of</strong> <strong>CT</strong>, MRI and ultrasound scanning is 78% (95%<br />

CI: 40; 97). The difference is statistically insignificant (p=0.48). The diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in the primary<br />

staging <strong>of</strong> disease is 100% (95% CI: 81; 100) and is higher than the accuracy <strong>of</strong> the conventional imaging methods,<br />

which is 67% (95% CI: 41; 87). The difference is statistically significant (p=0.04).<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting disease recurrence is 96% (95% CI: 80; 100) and is higher than the sensitivity <strong>of</strong><br />

the conventional imaging methods, which is 36% (95% CI: 18; 57). The difference is statistically significant, in favor <strong>of</strong><br />

<strong>PET</strong>-<strong>CT</strong> (p


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

As far as the diagnostic accuracy <strong>of</strong> the methods compared in the staging and detecting recurrences <strong>of</strong><br />

malignant female genital cancer, a statistically significant difference was observed in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> versus<br />

conventional methods: 96% (95% CI: 87; 100) vs. 50% (95% CI: 36; 64); p < 0.01, respectively.<br />

2. Ovarian cancer: detecting disease recurrences (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>)<br />

As a result <strong>of</strong> searching medical databases two primary clinical trials were found (Hauth 2005 and Makhija 2001)<br />

that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with <strong>CT</strong> in the assessment <strong>of</strong> ovarian cancer recurrences<br />

(N=27). The reference test was histopathological examination or, if histopathological examination was inaccessible,<br />

clinical follow-up.<br />

<strong>PET</strong>-<strong>CT</strong> is characterized by higher sensitivity than contrast <strong>CT</strong>. The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 84% (95% CI: 60; 97) and<br />

that <strong>of</strong> <strong>CT</strong> is 47% (95% CI: 24; 71).<br />

The specificity <strong>of</strong> these methods, evaluated only in Hauth 2005, was 100% for both <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> (95% CI: 63; 100).<br />

That implies that a negative result was returned by the imaging methods under <strong>analysis</strong> for each patient with no<br />

cancer recurrence.<br />

The data provided testify to the superiority <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostics over contrast <strong>CT</strong> imaging. The small size <strong>of</strong><br />

population in the primary studies limits the reliability <strong>of</strong> the <strong>analysis</strong>.<br />

Thyroid cancer<br />

1. Diagnostics <strong>of</strong> recurrence (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>/MRI; whole-body scintigraphy)<br />

As a result <strong>of</strong> searching through medical databases two primary clinical studies were found (Zimmer 2003, Nahas<br />

2005), in which <strong>PET</strong>-<strong>CT</strong> was compared to conventional diagnostics (<strong>CT</strong>/MRI and iodine 131 whole-body scintigraphy -<br />

WBS) in detecting thyroid cancer (N = 41). The imaging methods compared were verified based on<br />

histopathological tests, while for negative results verification was based on long-term clinical observation and a series<br />

<strong>of</strong> diagnostic imaging.<br />

In detecting recurrences <strong>of</strong> thyroid cancer, the sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 100% (95% CI: 40; 100), i.e. twice the sensitivity<br />

<strong>of</strong> <strong>CT</strong>/MRI, 50% (95% CI: 7; 93). The difference is not statistically significant (p = 0.48). In detecting recurrences <strong>of</strong><br />

thyroid cancer, <strong>PET</strong>-<strong>CT</strong> is characterized by a higher specificity than <strong>CT</strong> and MRI. The calculated values were 100%<br />

(95% CI: 29; 100) vs. 33% (95% CI: 1; 91) respectively. The difference is not statistically significant (p = 0.48). The<br />

diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is higher than that <strong>of</strong> <strong>CT</strong>/MRI, the values being 100% (95% CI: 59; 100) vs. 43% (95% CI:<br />

10; 82) respectively. The difference is not statistically significant (p = 0.13).<br />

Based on a comparison <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with iodine 131 whole body scintigraphy (I 131 WBS), the imaging sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

is higher than that <strong>of</strong> scintigraphy. The established value is 87% (95% CI: 66; 97) for <strong>PET</strong>-<strong>CT</strong>, but for I 131 WBS it is 0-21%. In<br />

detecting thyroid cancer recurrences, specificity is identical for <strong>PET</strong>-<strong>CT</strong> and whole-body scintigraphy and stands at<br />

100%. Similarly, in detecting recurrences <strong>of</strong> thyroid cancer, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 89% (95% CI: 75; 97)<br />

and is higher than that <strong>of</strong> I 131 WBS, which stands at 33% (95% CI: 18; 50).<br />

In one <strong>of</strong> the studies included, the authors reported that <strong>PET</strong>-<strong>CT</strong> scanning supported early revisions <strong>of</strong> treatment<br />

policies in 22 patients (67%). These patients were under observation following standard treatment <strong>of</strong> thyroid cancer.<br />

Additionally, in some patients, the adopted treatment policy was confirmed by the data collected from <strong>PET</strong>-<strong>CT</strong><br />

scanning.<br />

In summary, <strong>PET</strong>-<strong>CT</strong> is characterized by a higher diagnostic efficacy in detecting recurrences <strong>of</strong> thyroid cancer<br />

than convectional imaging methods.<br />

2. Staging (I124 – <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>, I 131 WBS, USG)<br />

23


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

24<br />

As a result <strong>of</strong> searching through medical databases, one primary prospective trial was found (Freudenberg 2004)<br />

that compared the diagnostic efficacy <strong>of</strong> I 124 <strong>PET</strong>-<strong>CT</strong> imaging with convectional diagnostic methods (<strong>CT</strong>, I 131 whole-<br />

body scintigraphy and ultrasonography) in the staging <strong>of</strong> thyroid cancer (N = 12). All tests were verified based on<br />

histopathological diagnostics and through formal consensus based on diagnostic imaging results.<br />

In tumor staging (T-status), the sensitivity <strong>of</strong> I 124 <strong>PET</strong>-<strong>CT</strong> is 100% (95% CI: 80; 100), and is the same as the sensitivity <strong>of</strong><br />

I 131 whole-body scintigraphy (I 131 WBS), but higher than the sensitivity <strong>of</strong> <strong>CT</strong> and ultrasound scanning, for which the<br />

values are 35% (95% CI: 14; 62) and 47% (95% CI: 23; 72) respectively.<br />

In node involvement staging (N-status), I 124 <strong>PET</strong>-<strong>CT</strong> proved to be the most sensitive. The values are: 100% for <strong>PET</strong>-<strong>CT</strong><br />

(95% CI: 54; 100), 83% for I 131 WBS (95% CI: 36; 100), 50% for <strong>CT</strong> (95% CI: 12; 88) and 33% for ultrasonography (95% CI: 4;<br />

78).<br />

In distant metastasis staging (M-status), I 124 <strong>PET</strong>-<strong>CT</strong> is characterized by a higher sensitivity than I 131 WBS or<br />

ultrasonography. The calculated values are 100% (95% CI: 92; 100), 76% (95% CI: 61; 87) and 65% (95% CI: 50; 79)<br />

respectively.<br />

The combined sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> imaging in clinical staging <strong>of</strong> thyroid cancer, irrespective <strong>of</strong> location, is 100%<br />

(95% CI: 94; 100) and is higher than the sensitivity <strong>of</strong> I 131 WBS, <strong>CT</strong> or ultrasound scanning. The values established for<br />

these methods are 83% (95% CI: 72; 91), 57% (95% CI: 44; 68) and 43% (95% CI: 23; 66) respectively. The differences<br />

between <strong>PET</strong>-<strong>CT</strong> and each <strong>of</strong> the scanning method compared are statistically significant (p < 0.01).<br />

In summary, I 124 <strong>PET</strong>-<strong>CT</strong> is characterized by a higher diagnostic efficacy than the conventional imaging methods in<br />

primary diagnostics <strong>of</strong> clinical staging <strong>of</strong> thyroid cancer.<br />

Head and neck cancer<br />

1. Diagnostic <strong>of</strong> malignant head and neck tumors (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>)<br />

As a result <strong>of</strong> searching through medical databases one primary study was found (Branstetter 2005) that<br />

compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with <strong>CT</strong> in the diagnostics <strong>of</strong> malignant head and neck tumors<br />

(primary lesions, recurrences following therapy, metastasis to cervical lymph nodes from an unknown primary focus;<br />

N = 65). Biopsy <strong>of</strong> suspicious lesions, clinical observation, and additional diagnostic imaging were used as the<br />

reference standard.<br />

The sensitivity and specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> were 98% (95% CI: 88; 100) and 92% (95% CI: 84; 97) respectively, while for <strong>CT</strong><br />

scans the values were 74% (95% CI: 59; 86) and 75% (95% CI: 64; 84). The accuracy was rated 94% for <strong>PET</strong>-<strong>CT</strong> (95% CI:<br />

89; 98) and 74% for <strong>CT</strong> (95% CI: 66; 82). The difference is statistically significant.<br />

2. Detection <strong>of</strong> bone involvement by oral cavity cancer (<strong>PET</strong>-<strong>CT</strong> vs. SPE<strong>CT</strong>/<strong>CT</strong>, <strong>CT</strong>)<br />

As a result <strong>of</strong> searching through medical databases one primary trial was found (Goerres 2005) that compared<br />

directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with SPE<strong>CT</strong>/<strong>CT</strong> and <strong>CT</strong> in identifying bone involvement by oral cavity<br />

cancer (N = 34). Histopathological examination following a surgical procedure was used as the reference standard.<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was the highest and stood at 100% (95% CI: 74; 100), the sensitivity <strong>of</strong> SPE<strong>CT</strong>/<strong>CT</strong> was 92%<br />

(95% CI: 62; 100). The sensitivity <strong>of</strong> <strong>CT</strong> was 92% (95% CI: 62; 100). The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 91% (95% CI: 71; 99),<br />

compared to 86% for SPE<strong>CT</strong>/<strong>CT</strong> (95% CI: 65; 97) and 100% for <strong>CT</strong> (95% CI: 85; 100).<br />

The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning was 94% (95% CI: 80; 99), compared to 88% for SPE<strong>CT</strong>/<strong>CT</strong> (95% CI: 73; 97) and 97%<br />

for <strong>CT</strong> (95% CI: 85; 100), which represents 94%, 88% and 97%, <strong>of</strong> properly diagnosed patients for <strong>PET</strong>-<strong>CT</strong>, SPE<strong>CT</strong>/<strong>CT</strong> and


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

<strong>CT</strong> respectively. The differences in accuracy, sensitivity and specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>, <strong>CT</strong> and SPE<strong>CT</strong>/<strong>CT</strong> tests in the<br />

diagnostics <strong>of</strong> local involvement <strong>of</strong> bones by oral cavity cancer are not statistically significant.<br />

3. The impact <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostics on therapy (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>, MRI)<br />

As a result <strong>of</strong> searching through medical databases two primary studies were found (Wild 2005, Koshy 2005) which<br />

compared directly the impact <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning on therapeutic decisions with standard head and neck cancer<br />

staging methods (<strong>CT</strong>, MRI, medical examination; N = 57).<br />

Histopathological examination was used as the reference standard in Wild 2005, while in Koshy 2005, when distant<br />

metastasis or synchronic tumors were suspected, histopathological tests or a combination <strong>of</strong> additional diagnostic<br />

imaging and clinical observation were executed (no data are available on the reference standard in the remaining<br />

patients).<br />

In Wild 2005, <strong>PET</strong>-<strong>CT</strong> imaging results triggered revisions <strong>of</strong> treatment suggested based on conventional imaging in<br />

43% patients, while in Koshy 2005 the value was 25%, which produced the total <strong>of</strong> 32% cases (95% CI: 21; 44).<br />

Pancreatic cancer: diagnostics <strong>of</strong> primary focus and clinical staging system (<strong>PET</strong>-<strong>CT</strong><br />

vs <strong>CT</strong>, ERCP, MRI, EUS, laparoscopy)<br />

As a result <strong>of</strong> searching through medical databases one primary clinical trial was found (Heinrich 2005) that<br />

compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with conventional imaging methods (contrast <strong>CT</strong>, ERCP, MRI,<br />

EUS, diagnostic laparoscopy) in diagnosing primary lesions and in staging pancreatic cancer (N = 59). The<br />

comparison <strong>of</strong> imaging methods was verified by histopathological tests <strong>of</strong> material removed during a surgical<br />

procedure or bioptates, or by long-term clinical observation.<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning in detecting primary lesions <strong>of</strong> pancreatic cancer is 89% (95% CI: 76; 96), which is<br />

lower than the sensitivity <strong>of</strong> contrast <strong>CT</strong>, which is 93%. <strong>PET</strong>-<strong>CT</strong> imaging has a higher sensitivity rating than contrast <strong>CT</strong> in<br />

detecting pancreatic cancer, the estimated values being 69% (95% CI: 39; 91) vs. 21%, respectively. The accuracy <strong>of</strong><br />

<strong>PET</strong>-<strong>CT</strong> is 85% (95% CI: 73; 93). No statistically significant differences were observed between the groups for any <strong>of</strong> the<br />

parameters <strong>of</strong> efficacy discussed.<br />

In M-staging, <strong>PET</strong>-<strong>CT</strong> is characterized by a higher sensitivity than conventional imaging methods, the estimated<br />

values being 81% (95% CI: 54; 96) vs. 56% (95% CI: 30; 80) respectively. The difference in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is not<br />

statistically significant (p = 0.22). Imaging specificity favors <strong>PET</strong>-<strong>CT</strong>, for which it stands at 100% (95% CI: 92; 100) vs. 95%<br />

(95% CI: 84; 99) for the conventional methods compared. The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in M-staging is 95% (95% CI: 86; 99)<br />

and is higher than the accuracy <strong>of</strong> the conventional methods (<strong>CT</strong>, NMR and USG), 85% (95% CI: 73; 93). The<br />

difference, although close to the threshold, did not reach statistical significance (p = 0.08).<br />

A revision <strong>of</strong> oncological procedures as a result <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning was reported for 16% patients (95% CI: 6; 32)<br />

with resectable pancreatic cancer. Procedure revision is more likely if <strong>PET</strong>-<strong>CT</strong> scanning is used in clinical staging than<br />

if the conventional methods are used, the estimated values being 33% (95% CI: 20; 48) vs. 20% (9; 34) respectively.<br />

The difference in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is statistically significant (p = 0.03).<br />

25


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Gastrointestinal stromal tumor: staging and evaluation <strong>of</strong> response to treatment (<strong>PET</strong>-<br />

<strong>CT</strong> vs <strong>CT</strong>)<br />

26<br />

A single clinical trial was found (Antoch 2004) that compared the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with that <strong>of</strong> <strong>CT</strong><br />

imaging in GIST staging and in the evaluation <strong>of</strong> treatment response in patients with gastrointestinal stromal tumor<br />

(N = 20). Long-term clinical observation was used as a reference standard.<br />

In primary disease staging, <strong>PET</strong>-<strong>CT</strong> imaging is more effective as a diagnostics method than <strong>CT</strong>. The difference<br />

between the diagnostic methods compared was statistically significant (p < 0.0001). In terms <strong>of</strong> response to<br />

treatment, significant congruence <strong>of</strong> results between the method compared and the reference standard was<br />

identified only for <strong>PET</strong>-<strong>CT</strong>. When <strong>PET</strong>-<strong>CT</strong> scanning was used, response to treatment was diagnosed correctly in 95%,<br />

100% and 100% patients in 1, 3 and 6-month follow-up periods respectively. For <strong>CT</strong>, the respective values were 44%,<br />

60% and 57%. The difference between particular diagnostic methods reached statistical significance in the 1 st month<br />

(p = 0.001).<br />

Colorectal liver metastasis: residual disease after ablation (<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>)<br />

As a result <strong>of</strong> searching through medical databases, a single primary prospective clinical trial was found (Veit 2006)<br />

that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> imaging in detecting residual lesions <strong>of</strong> colorectal<br />

liver metastasis following a therapy that used high-current radio-frequency current ablation (N = 13).<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting residual disease is 65%, and is higher than that <strong>of</strong> <strong>CT</strong>, which stands at 44%. The<br />

authors provide no data on the statistical significance <strong>of</strong> the results above.<br />

The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting residual disease is 68%, and is higher than the accuracy <strong>of</strong> <strong>CT</strong>, which stands at<br />

47%.<br />

Analysis <strong>of</strong> imaging methods compared showed that in detecting residual lesions <strong>of</strong> colorectal liver metastasis<br />

following high-current radio-frequency ablation, a higher efficacy is demonstrated by <strong>PET</strong>-<strong>CT</strong> diagnostics than by <strong>CT</strong><br />

imaging.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

2. INDEX OF ABBREVIATIONS<br />

18 F-FDG [18F]-Fluoro-Deoxy-Glucose<br />

3D 3 Dimensional Imaging<br />

Acc Accuracy<br />

AJCC American Joint Committee on Cancer<br />

BAC Fine-needle aspiration biopsy<br />

BD, bd No data available<br />

CA 19-9 Carbohydrate antigen 19-9<br />

CEA Carcinoembryonic antigen<br />

CI Confidence Interval<br />

CR Complete response<br />

<strong>CT</strong> Computed Tomography<br />

DOR Diagnostics Odds Ratio<br />

EBM Evidence Based Medicine<br />

ERCP Endoscopic retrograde cholangio- pancreatography<br />

EUS-FNA Endoscopic ultrasound-guided fine-needle aspiration biopsy<br />

FIGO International Federation <strong>of</strong> Gynecology and Obstetrics<br />

FN False Negative<br />

FP False Positive<br />

GIST Gastrointestinal stromal tumor<br />

I 124 Radioactive isotope <strong>of</strong> iodine-124<br />

I 131 Radioactive isotope <strong>of</strong> iodine-131<br />

IS Statistically significant<br />

kg Kilogram<br />

l Liter<br />

LR- Negative likelihood ratio<br />

LR+ Positive likelihood ratio<br />

M Metastasis staging<br />

M0 Absence <strong>of</strong> distant metastasis<br />

M1 Presence <strong>of</strong> distant metastasis<br />

MBq Becquerel, unit <strong>of</strong> radioactivity<br />

MRI Magnetic resonance imaging<br />

N Number <strong>of</strong> patients in group<br />

n Number <strong>of</strong> patients with end point<br />

N Llymph nodes involvement staging<br />

N0 Absence <strong>of</strong> metastasis to lymph nodes<br />

27


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

28<br />

nd Not applicable<br />

Ng Nanogram<br />

NNT Number <strong>of</strong> patients needed to treat<br />

NPV Negative predictive value<br />

NS Not statistically significant<br />

NSCLC Non-small cell lung cancer<br />

NSE Neurospecific enolase<br />

OR Odds ratio<br />

pp. Percentage points<br />

<strong>PET</strong>-<strong>CT</strong>, <strong>PET</strong>/<strong>CT</strong> Positron Emission Tomography and Computed Tomography<br />

pg Pikogram<br />

PPV Positive predictive value<br />

R<strong>CT</strong> Randomized controlled trial<br />

Re Reference method<br />

rtg X-rays trial<br />

SD Standard deviation<br />

Se Sensitivity<br />

Sp Specificity<br />

T Identification <strong>of</strong> tumor size<br />

Tis In situ tumor<br />

TN True negative<br />

TNM<br />

Stage <strong>of</strong> disease: T – tumor size identification; N – assessment <strong>of</strong> lymph nodes involvement, M<br />

– detecting metastasis<br />

TP True positive<br />

Tx Latent tumor<br />

USG Ultrasonography<br />

WBS Whole body scintigraphy<br />

WMD Weighted mean difference


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

3. DESCRIPTION OF HEALTH PROBLEM<br />

3.1. Epidemiology <strong>of</strong> cancers<br />

In 2002, 10.9 million <strong>of</strong> new cases <strong>of</strong> cancer, including 26 most common tumors and 6.7<br />

million related deaths were reported in the world. The most common tumor is lung cancer,<br />

with respect to both disease incidence (1.35 million) and mortality (1.18 million). This cancer<br />

type is typified by poor prognosis, which is indicated by a high mortality to incidence ratio<br />

(0.87). Breast cancer is the second leading malignant tumor in the world (1.15 million <strong>of</strong> new<br />

cases), but is typified by more optimistic prognosis (mortality to incidence ration at 0.35).<br />

Colorectal cancer was diagnosed in 1.02 million people, gastric cancer in 934,000, and liver<br />

cancer in 626,000.<br />

The most common malignant tumor among men is lung cancer, however in developed<br />

countries it is second to prostate cancer. Cervical cancer is the second leading malignant<br />

cancer among women in developing countries; in developed countries it ranks seventh.<br />

Morbidity, i.e. the prevalence <strong>of</strong> cancer (number <strong>of</strong> patients) in a population in a specific<br />

period <strong>of</strong> time, depends on the incidence <strong>of</strong> disease and its mortality. As regards morbidity,<br />

the most common tumor types are breast cancer (17.9%), colorectal cancer (11.5%), and<br />

prostate cancer (9.6%). The morbidity to disease incidence quotient is an index that<br />

represents prognosis. For example, the world’s morbidity rate for breast cancer is the highest,<br />

although it breast cancer has a lower incidence rate than lung cancer, which has a poor<br />

prognosis.<br />

Figure 1 shows disease incidence and death rates <strong>of</strong> the world’s most common tumors, in<br />

developed and developing countries [1].<br />

29


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Figure 1.<br />

Global incidence and mortality rates for the most common tumors types in developing and developed countries [1]<br />

30<br />

After World War II in Europe, a rising trend in the incidence <strong>of</strong> malignant tumors was<br />

observed, not only in women but also in men. Recently, in the countries <strong>of</strong> Western Europe,<br />

the incidence <strong>of</strong> the prostate cancer in men has been going up in the first place, while the<br />

incidence <strong>of</strong> lung cancer has been following a downward trend or staying flat. In these<br />

countries, the incidence <strong>of</strong> gastric cancer has significantly dropped in men. The incidence<br />

rates <strong>of</strong> breast cancer calculated for women in Western Europe keep rising, so does the


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

incidence <strong>of</strong> lung cancer, especially in young women. The incidence <strong>of</strong> cervical cancer and<br />

gastric cancer is going significantly down in this group; however the incidence <strong>of</strong> cancer <strong>of</strong><br />

the large intestine and colon is increasing slightly regardless <strong>of</strong> gender. In Eastern Europe, the<br />

rates <strong>of</strong> incidence <strong>of</strong> lung cancer in men and breast cancer in women keep growing. High<br />

incidences <strong>of</strong> gastric cancer in both sexes and <strong>of</strong> cervical cancer in women are still<br />

observed. The frequency <strong>of</strong> developing cancer is expected to grow gradually over the next<br />

20 years, the reasons being, among other things, the ageing <strong>of</strong> the population and increased<br />

exposure to risk factors [2]. In the member states <strong>of</strong> the European Union, a decrease in<br />

cancer-related mortality has been observed, but in a number <strong>of</strong> countries <strong>of</strong> Eastern Europe,<br />

including Poland, the trend continues along the rising path [2].<br />

Based on data from the Central Statistical Office, in 2001 the incidence <strong>of</strong> malignant<br />

cancer in Poland was 298 per 100,000 people annually. For a number <strong>of</strong> years this ratio has<br />

been growing faster than the population. The disease is the cause <strong>of</strong> about 40% <strong>of</strong> deaths in<br />

women and about 30% deaths in men aged 45–64 years [3].<br />

Table 1 presents the morbidity and mortality rates for selected malignant cancer types in<br />

men and women in Poland (2002) [4].<br />

Table 1.<br />

Morbidity and mortality rates for selected malignant cancer types in Poland in 2002 [4]<br />

Type <strong>of</strong><br />

tumor<br />

Incidence<br />

<strong>of</strong> disease<br />

Morbidity<br />

(death<br />

rate)*<br />

Men Women<br />

Number<br />

<strong>of</strong> deaths<br />

Mortality*<br />

Incidence<br />

<strong>of</strong> disease Morbidity*<br />

Number<br />

<strong>of</strong> deaths<br />

Mortality<br />

(death<br />

rate)*<br />

Oral cavity 1719 7.3 800 3.4 441 1.4 221 0.7<br />

Nasal part<br />

<strong>of</strong> the<br />

pharynx<br />

Other parts<br />

<strong>of</strong> the<br />

pharynx<br />

141 0.6 81 0.3 63 0.2 37 0.1<br />

1383 6.0 523 2.2 204 0.7 89 0.3<br />

Esophagus 1408 6.0 1113 4.7 305 0.9 260 0.8<br />

Stomach 4962 20.7 4017 16.6 2634 7.8 2188 6.2<br />

Colorectum 7671 31.9 4432 18.2 7909 23.5 4082 11.4<br />

Liver 851 3.6 1045 4.3 884 2.6 1141 3.2<br />

Pancreas 2254 9.5 1914 8.0 2103 6.1 1849 5.3<br />

Larynx 3062 13.2 1539 6.5 370 1.3 151 0.5<br />

Lung 19478 82.0 16354 68.4 4534 14.6 3960 12.3<br />

Skin<br />

melanoma<br />

881 3.8 463 2.0 1250 4.5 451 1.5<br />

31


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

32<br />

Prostate 6016 24.1 3114 12.4<br />

Testicle 869 4.1 118 0.5<br />

Breast 14358 50.3 4781 15.5<br />

Uterine<br />

cervix<br />

Uterine<br />

corpus<br />

4901 18.4 2278 7.8<br />

3824 13.0 963 2.8<br />

Ovary 3570 12.5 2071 6.8<br />

Kidney 3151 13.5 1477 6.2 1662 5.3 855 2.5<br />

Gall<br />

bladder<br />

Central<br />

nervous<br />

system<br />

Thyroid<br />

gland<br />

Non-<br />

Hodgkin’s<br />

lymphoma<br />

Hodgkin's<br />

disease<br />

Multiple<br />

myeloma<br />

5215 21.8 2065 8.4 1181 3.6 498 1.4<br />

1812 8.2 1291 5.8 1546 5.9 1148 4.1<br />

422 1.8 82 0.3 1086 4.1 216 0.6<br />

1574 7.0 781 3.4 1088 3.8 578 1.8<br />

565 2.7 249 1.1 670 3.0 159 0.6<br />

721 3.1 418 1.7 731 2.3 454 1.3<br />

Leukaemias 1575 7.2 1252 5.4 1293 4.6 1050 3.3<br />

Total 71349 301.8 48765 203.5 63220 210.3 36317 110.6<br />

Prognosis varies depending on type <strong>of</strong> cancer as well as on expenditure on health care. In<br />

1995, expenditure ranged from US$420 in Poland (parity <strong>of</strong> USD purchasing power per person<br />

in population) to US$2555 (Switzerland) [5].<br />

European 5-year mean survival rates range from 94% for lip cancer to < 4% for pancreatic<br />

cancer. For 21 out <strong>of</strong> 42 cancer types in adults, the 5-year mean survival rate is ≥ 50%.<br />

Generally, survival rates are high for lip, testicle and thyroid cancer, skin melanoma and<br />

Hodgkin’s disease (the European 5-year mean survival rate is ≥ 80%). These high survival rates<br />

are attributable to possibilities <strong>of</strong> efficacious treatment, as well as a good availability <strong>of</strong> tumor<br />

location and early diagnosis opportunities. For a larger group <strong>of</strong> cancers, the 5-year mean<br />

survival rate ranges from 60 to 79%, including breast cancer, prostate cancer, cancer <strong>of</strong> the<br />

gall bladder, cervical cancer, uterine cancer and cancer <strong>of</strong> the larynx. This group represents<br />

one-third <strong>of</strong> all tumors. Moderate prognosis (5-year survival rates <strong>of</strong> 40–59%) is common for<br />

colorectal cancer, non-Hodgkin’s lymphomas, and renal cancer. Poor prognosis is associated<br />

with about 10% <strong>of</strong> tumors (5-year survival rates <strong>of</strong> 20–39%), among them gastric cancer,<br />

ovarian cancer, or multiple myeloma. The poorest prognosis (5-year survival rates < 20%) is<br />

associated with tumors such as lung cancer, pancreatic cancer, esophageal cancer, brain<br />

cancer, and liver cancer. Liver cancer represents one-fourth <strong>of</strong> all tumors. They are not as<br />

easily accessible for diagnostic procedures and are usually diagnosed late.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Prognostic factors include the patient’s age. For example, the highest 5-year survival rate<br />

in breast cancer in women is observed for ages 45-54, and for prostate cancer for ages 55-64.<br />

Sex is another factor. A Higher 5-year survival rate has been observed in women compared<br />

to men for four head and neck tumors: cancer <strong>of</strong> the salivary gland, oral cavity, tongue and<br />

<strong>of</strong> oral part <strong>of</strong> pharynx (difference in 5-year survival rate ≥ 15%), for thyroid cancer (10%) and<br />

for skin melanoma (10%). These differences probably stem from the fact that women and<br />

tumors are diagnosed earlier on, hence their stage is lower.<br />

5-year survivals differ between European countries. The lowest rate has been reported in<br />

five countries <strong>of</strong> Eastern Europe (the Czech Republic, Estonia, Poland, Slovakia and Slovenia)<br />

and in six countries <strong>of</strong> Western Europe (Germany, Great Britain, Wales, Scotland, Malta and<br />

Portugal). Of all the countries <strong>of</strong> Eastern Europe, Poland has the lowest survival rates for<br />

cancer [5].<br />

Figure 2 shows the relative 5-year survival rates for selected cancer types in Europe [5] 1 .<br />

1 1. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin 2005; 55 (2): pp 74–108.<br />

2. Evidence-based Cancer Prevention: Strategies for NGOs – A UICC Handbook for Europe. http://www.uicc.org<br />

3. Narodowy program zwalczania chorób nowotworowych. ZałoŜenia i cele operacyjne 2006–2015;<br />

http://www.mz.gov.pl/wwwfiles/ma_struktura/docs/zalozenia_ustawy_o_npzchn.pdf<br />

4. The Globocan 2002 database: http://www-dep.iarc.fr<br />

5. Coleman MO et al. Eurocare-3 summary: cancer survival in Europe at the end <strong>of</strong> 20th century. Ann Oncol 2003; 1:<br />

pp 128–149.<br />

33


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Figure 2.<br />

European average relative 5-year survival rates (%) for 42 selected tumor types; adults aged 15–99, diagnosed in<br />

1990–1994, with the follow-up period till 1999 [5]<br />

34<br />

3.2. Lung cancer<br />

3.2.1. Epidemiology<br />

Malignant lung tumors are the most common tumor types in the world. In 2002, 1.35 million<br />

persons had lung cancer and 1.18 million related deaths were reported globally. The high


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

morbidity to incidence ration (0.87) proves the disease has a poor prognosis. Currently, these<br />

tumors represent 12.4% <strong>of</strong> all newly diagnosed malignant tumors [1. 14]. Since 1985, a 51%<br />

growth in lung cancer incidence has been observed, the reasons seem to be the growing<br />

and ageing population [1]. In Europe, growth in the incidence <strong>of</strong> lung cancer was observed<br />

in the 1960’s and 1970’s; later, lung cancer began to fall (mostly in Great Britain) [2]. The<br />

incidence <strong>of</strong> lung cancer varies significantly between men and women. It is estimated that<br />

every 4th European men diagnosed with malignant tumor will develop lung cancer, while in<br />

women this tumor type represents only 6% <strong>of</strong> all tumors diagnosed [14]. Unlike in men, for who<br />

the growth <strong>of</strong> lung cancer incidence has been curbed, lung cancer incidence and mortality<br />

rate in European women is increasing dramatically [2]. In Eastern Europe lung cancer is the<br />

most frequent malignant tumor in men and the seventh leading cancer type in women [2].<br />

Poland is among the countries with the highest incidence and mortality rate <strong>of</strong> lung<br />

cancer. In 1999, the rates were: 83/100000 and 81.9/100000 in men, and 19.6/100000 and<br />

18.4/100000 in women. Before 1995, an upsurge in the incidence and mortality caused by this<br />

tumor had been observed. Since 1995, the growth pace has been slower [1, 2, 4, 14]. At the<br />

same time, over the last 30 years, a significant increase <strong>of</strong> incidence and the mortality rate<br />

has been reported in Poland for younger groups, both men and women [4]. 2002<br />

epidemiological reports indicate that age-standardized lung cancer incidence and mortality<br />

rates were 82 and 68.4 per 100,000 men, and 14.6 and 12.3 respectively per 100,000 women<br />

[3].<br />

Graph 1 illustrates mortality trends in 20 selected European countries.<br />

35


ASRW (age-standardized incidece rate per world population) on 100000 persons<br />

<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Chart 1.<br />

Mortality trends associated with malignant lung cancer in 20 European countries (age-standardized rate); source:<br />

WHO [2]<br />

36<br />

3.1.1. Risk factors<br />

Source: WHO<br />

The relation between smoking or exposure to carcinogenic substances contained in<br />

cigarettes smoke and malignant lung tumor has been known for a long time. Addiction to<br />

nicotine may be the cause <strong>of</strong> up to 90% <strong>of</strong> new cases. If a smoker quits smoking, cancer risk<br />

decreases gradually. Also, second-hand smoking increases the risk <strong>of</strong> disease. Other<br />

carcinogenic substances include industrial pollution, heavy metals, ionizing radiation, and<br />

exposure to radon or asbestos, but their impact is not as high. Genetic predisposition to lung<br />

tumors also exists [4, 6, 14].<br />

3.1.2. Symptoms<br />

Years: 1953–57 to 1993–97<br />

Initially, lung cancer is asymptomatic; possible symptoms are associated in the main with<br />

the location <strong>of</strong> the tumor. Squamous and small cell cancers are located centrally, while small<br />

adenocarcinoma or giant cell cancer are more <strong>of</strong>ten situated peripherally in the lung.<br />

Tumors located centrally block the bronchi, cause cough and haemoptysis. Peripheral tumors


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

cause pain, interstitial pneumonia or dyspnoea. The most common symptoms are cough<br />

(>50% <strong>of</strong> patients), dyspnoea (30–40%), chest pain (25–35%), haemoptysis (15–30%), recurring<br />

pneumonia (as the first symptom in 15–20% <strong>of</strong> patients) and atelectasis caused by poor<br />

bronchial patency due to obstruction from tumor mass. Symptoms <strong>of</strong> local spread <strong>of</strong> the<br />

tumor are: superior vena cava syndrome (compression <strong>of</strong> the superior vena cava), hoarse<br />

throat (caused by paralysis <strong>of</strong> the recurrent laryngeal nerve), Horner’s syndrome. Pancoast’s<br />

syndrome is associated with the presence <strong>of</strong> tumor in the apex <strong>of</strong> the lung, and is<br />

accompanied by shoulder, arm and forearm pain. Other symptoms are disability <strong>of</strong> trachea<br />

and loss <strong>of</strong> esophageal patency, which may lead to breathing disturbances or problems with<br />

swallowing. Heart infiltration may lead to its functional disturbances, while carcinogenic<br />

infiltration <strong>of</strong> the pleura may be conducive to accumulation <strong>of</strong> exudative fluid in the pleural<br />

cavity, which additionally decreases the vital capacity <strong>of</strong> the lung and impedes ventilation.<br />

Infiltration <strong>of</strong> the phrenic nerve leads to diaphragm paralysis, which additionally impedes lung<br />

ventilation. Cancer invasion in the thoracic wall causes strong pain. A number <strong>of</strong> ailments<br />

beside those listed above are related to metastasis, the most frequent being osseal pains<br />

(bone metastasis), headaches and neurological symptoms (brain metastasis), pain in<br />

hypergastrium, nausea, loss <strong>of</strong> body weight, jaundice (liver metastasis) [4, 6, 8].<br />

Paraneoplastic syndromes can be observed in the course <strong>of</strong> lung tumors, mostly as<br />

hormonal or neurological disorders. Endocrinological disorders occur mainly in small cell<br />

cancer. The most common endocrinological paraneoplastic syndrome is the syndrome <strong>of</strong><br />

inappropriate antidiuretic hormone secretion (ADH). Secretion <strong>of</strong> atrial natriuretic peptide<br />

manifests itself by a low content <strong>of</strong> sodium in urine and low blood pressure. Increased<br />

concentration <strong>of</strong> corticotrophin may cause Cushing’s syndrome. In patients suffering from<br />

non-small cell lung cancer, hypocalcaemia is observed. Neurological paraneoplastic<br />

syndromes include Lambert and Eaton syndrome (i.e. weakness <strong>of</strong> the extremities, which<br />

manifests itself during exercise), polymiositis and dermatomyositis. The disease may also be<br />

accompanied by cerebritis and inflammation <strong>of</strong> the spinal cord, cerebellum degeneration,<br />

sensory neuropathy, or blunt vision. It is believed that all the neurological syndromes<br />

mentioned are <strong>of</strong> autoimmune origin [9, 10].<br />

At the advanced stages <strong>of</strong> the disease, syndromes are associated mainly with distant<br />

metastasis. Cancer spreads most <strong>of</strong> all to the liver, bones, brain, skin, suprarenal glands, and<br />

small cell lung cancer can also invade bone marrow [4, 6 and 8]. At initial diagnosis, small cell<br />

cancer <strong>of</strong> lung is usually diffused; only in 1/3 <strong>of</strong> patient the tumor is low stage cancer [10].<br />

General cancer symptoms include weakness, cachexia, loss <strong>of</strong> body weight and fever.<br />

37


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

38<br />

3.1.3. Histopathological picture<br />

The classification <strong>of</strong> primary lung tumors is based on the morphologic picture, biological<br />

features, clinical course and treatment procedures. Of all the numerous existing histological<br />

classifications <strong>of</strong> lung cancer, the most widespread is the classification accepted by WHO<br />

in 1999 [4]. Diagram 1 presents the classification <strong>of</strong> lung tumors based on WHO<br />

recommendations.<br />

Diagram 1.<br />

WHO classification <strong>of</strong> lung tumors, 1999<br />

1. Squmous cell carconoma<br />

a. Papillary<br />

b. Clear cell<br />

c. Small cell<br />

d. Basaloid<br />

2. Small cell cancer<br />

a. Combined small cell carcinoma<br />

3. Adenocarcinoma<br />

a. Acinar<br />

b. Papillary<br />

c. Bronchoalveolar carcinoma<br />

I. Nonmucinous<br />

II. Mucinous<br />

III. Mixed mucinous and nonmucinous or indeterminate cell type<br />

d. Solid adenocarcinoma with mucin formation<br />

e. Adenocarcinoma with mixed subtypes and variants (mucinous cystadenocarcinoma, signet ring<br />

adenocarcinoma, clear cell adenocarcinoma)<br />

4. Large cell carcinoma<br />

a. Large-cell neuroendocrine carcinoma<br />

I. Combined large cell neuroendocrine carcinoma<br />

b. Basaloid carcinoma<br />

c. Lymphoepithelioma-like carcinoma<br />

d. Clear cell carcinoma<br />

e. Large cell carcinoma with rhabdoid phenotype<br />

5. Adenosquamous carcinoma<br />

6. Carcinomas with pleomorphic, sarcomatoid or sarcomatous elements<br />

a. Carcinomas with spindle and/or giant cells<br />

I. Pleomorphic carcinoma<br />

II. Spindle cell carcinoma<br />

III. Giant cell carcinoma<br />

b. Carcinosarcoma<br />

c. Pulmonary blastoma<br />

d. Others<br />

7. Carcinoid tumors<br />

a. Typical carcinoid


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

b. Atypical carcinoid<br />

8. Carcinomas <strong>of</strong> salivary gland type<br />

a. Mucoepidermoid carcinoma<br />

b. Adenoid cystic carcinoma<br />

c. Others<br />

9. Unclassified carcinoma<br />

Considering different clinical courses, prognosis and treatment, lung cancers are, in most<br />

cases, divided into non-small cell cancers, which represent 80% <strong>of</strong> all lung tumors (including<br />

squamous cell cancer, adenocarcinoma and giant cell carcinoma), and small cell cancers<br />

(20% <strong>of</strong> all).<br />

Non-small cell cancers are tumors <strong>of</strong> small chemosensivity. The first-choice therapy is<br />

surgical excision <strong>of</strong> the tumor. Small cell cancer is characteristic for its fast growth, tendency<br />

to rapid metastasis formation and high vulnerability to chemotherapy.<br />

Squamous cell carcinoma (carcinoma planoepitheliale) represents about 40% <strong>of</strong> all<br />

primary lung tumors. Most commonly it is located in the large bronchi, and associated above<br />

all with cigarettes smoking. It develops from metaplasia <strong>of</strong> respiratory track epithelium, and is<br />

slow-growing. Symptoms <strong>of</strong> bronchus narrowing, such as atelectasis, or pneumonia are<br />

common in the course <strong>of</strong> the disease.<br />

Adenocarcinoma represents about 30% <strong>of</strong> primary lung tumors. Most <strong>of</strong>ten it is located in<br />

the small bronchi and is not as strongly associated with smoking. Atypical alveolar<br />

hyperplasia is considered to precede the development <strong>of</strong> adenocarcinoma. A special type<br />

<strong>of</strong> this tumor is bronchoalveolar cancer (carcinoma bronchoalveolare). This cancer can<br />

occur multifocally.<br />

Giant cell cancer (carcinoma macrocellulare) is the most rare (10%) <strong>of</strong> all cancer types. Its<br />

localization is the large bronchi. The clinical course is similar to adenocarcinoma.<br />

Small cell cancer (carcinoma microcellulare) is distinguishable for the highest cellular<br />

proliferation index. At diagnosis, haematogenous metastases are usually formed. This cancer<br />

type is particularly strongly associated with cigarettes smoking. As a rule, it is located close to<br />

pulmonary hiluses. Metastasis occurs mainly in the liver, bones, bone marrow, central nervous<br />

system, skin and s<strong>of</strong>t tissues. Symptoms <strong>of</strong> endocrinological or neurological nature are <strong>of</strong>ten<br />

present [4, 6, 7].<br />

A histopathological picture helps determine the histological malignancy grade (G trait).<br />

Table 2 presents the classification.<br />

39


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Table 2.<br />

histological malignancy grades <strong>of</strong> lung cancer<br />

Gx Grade <strong>of</strong> differentiation cannot be assessed<br />

G1 Highly differentiated<br />

G2 Moderate degree <strong>of</strong> differentiation<br />

G3 Low degree <strong>of</strong> differentiation<br />

G4 Undifferentiated<br />

40<br />

3.1.4. Staging<br />

Table 3 presents the commonly used TNM system for the staging <strong>of</strong> lung cancers [15].<br />

Table 3.<br />

TNM staging system <strong>of</strong> the clinical advancement <strong>of</strong> the lung cancer<br />

Primary tumor<br />

Tx Primary tumor cannot be evaluated<br />

T0 No evidence <strong>of</strong> primary tumor<br />

Tis Carcinoma in situ<br />

T1<br />

T2<br />

T3<br />

T4<br />

Regional lymph nodes<br />

Tumor 3 centimeters (< 3 cm) or less in greatest dimension, surrounded by<br />

lung or pleura<br />

Tumor more than 3 centimeters (> 3 cm) in greatest dimension, or tumor<br />

invading the visceral pleura, atelectasis or lung infection<br />

Tumor that invades the chest wall, diaphragm, pleura, or pericardium, or the<br />

main stem bronchus less than 2 centimeters (< 2 cm) from the carina but<br />

without involvement <strong>of</strong> the carina<br />

Invasion <strong>of</strong> mediastinum, heart, great vessels, trachea, carina, esophagus,<br />

vertebral body, tumor with a malignant pleural effusion<br />

N0 No regional lymph node metastasis<br />

N1 Metastasis to same-side peribronchial and/or hilar lymph nodes tumor<br />

N2 Metastasis to same-side mediastinal and/or subcarinal lymph nodes<br />

N3<br />

Distant metastasis<br />

M0 No distant metastasis<br />

M1 Distant metastasis present<br />

Staging<br />

Latent cancer Tx N0 M0<br />

Stage 0 Tis<br />

Stage I<br />

Stage II<br />

Stage IIIA<br />

Stage IIIB<br />

Metastasis to opposite-side mediastinal or hilar nodes or to same- or oppositeside<br />

supracalvicular lymph nodes.<br />

T1 N0 M0<br />

T2 N0 M0<br />

T1 N1 M0<br />

T2 N1 M0<br />

T3 N0 M0<br />

T3 N1 M0<br />

T1-3 N2 M0<br />

Every T N3 M0<br />

T4, N (any), M0<br />

Stage IV T (any), N (any), M1


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

It should be noted that the classification above particularly important for non-small cell<br />

lung cancers, because small cell cancers are usually diagnosed in a diffused form. For small<br />

cell cancers, a two-stage classification <strong>of</strong> tumor is used:<br />

1. Limited stage – tumor is confined to one side <strong>of</strong> the chest, also with same-side<br />

carcinogenic exudation; involvement <strong>of</strong> same-side hilar region, and the involvement<br />

<strong>of</strong> mediastinal and supraclavicular, lymph nodes on both sides are possible,<br />

2. Extensive stage – tumor outside the limited disease area.<br />

3.1.4.1. Non-small cell cancer staging<br />

In patients for whom surgical excision <strong>of</strong> tumor is considered, it is particularly important to<br />

determine the local and regional cancer stage. For this purpose, <strong>CT</strong> with contrast is<br />

indispensable. The study covers the upper part <strong>of</strong> the chest to examine the liver, suprarenal<br />

glands, lymph nodes (frequent metastatic location). Other imagining studies are performed if<br />

clinical symptoms occur suggesting involvement <strong>of</strong> these organs.<br />

3.1.4.2. Small cell cancer staging<br />

As small cell cancer diffuses early, it is particularly important to identify cancer foci outside<br />

the chest. If at least one focus <strong>of</strong> distant metastasis is present general disease is diagnosed<br />

positively. Therefore further screening for distant metastasis is pointless as is has no effect on<br />

the procedure with the patient. It is necessary to perform <strong>CT</strong> <strong>of</strong> the abdominal cavity, and <strong>CT</strong><br />

or magnetic resonance imagining <strong>of</strong> the brain because asymptomatic brain metastasis is<br />

frequent. As a result <strong>of</strong> frequent bone marrow metastasis, scintigraphic study <strong>of</strong> the skeletal<br />

system is recommended. Trepanobiopsy <strong>of</strong> bone marrow is performed in patients with<br />

positive results <strong>of</strong> scintigraphy, with high lactic dehydrogenase level in plasma or with<br />

thrombocytopenia.<br />

3.1.5. Diagnostics and treatment<br />

3.1.5.1. Case history<br />

Identifies the disorders described above.<br />

3.1.5.2. Physical examination<br />

Symptoms depend mainly on the disease stage, location <strong>of</strong> primary tumor, as well as<br />

metastatic location. Examination may help detect auscultatory changes over lung fields,<br />

which correspond to pneumonia, atelectasis, fluids in pleura or infiltration by tumor. In<br />

advanced cases, enlarged cervical, supraclavicular, and axillary lymph nodes are<br />

detectable. Liver metastasis produces liver enlargement, tenderness under the right costal<br />

arch or jaundice. Brain metastasis leads to neurological disorders such as focal symptoms or<br />

41


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

sensory disorders. Strong bone pains and tenderness may be caused by metastasis. Quite<br />

common symptoms are clubbed fingers [4, 6, 8].<br />

42<br />

3.1.5.3. Diagnostic imaging<br />

Apart from case history and physical study, the basic method <strong>of</strong> lung diagnostics is chest X-<br />

ray in the anterolateral position, with additional lateral projections. It helps detect lesions that<br />

<strong>of</strong>ten do not produce clinical symptoms typical <strong>of</strong> cancer yet. Diagnostic imaging is not<br />

recommended as lung cancer screening examinations in asymptomatic patients or patients<br />

without tumor in history. In a routine chest radiogram, small, 1cm-big tumors can be seen [9,<br />

12]. Apart from tumor-like lesions, a chest radiogram may reveal other lesions that are<br />

manifestations <strong>of</strong> cancer: inflammatory interstitial densities, atelectatic symptoms, exudation<br />

to the pleural cavity. If tumor-like changes are present in a chest radiogram, the radiogram<br />

needs to be compared with previous imaging studies <strong>of</strong> the chest. If the picture <strong>of</strong> the lesion<br />

remains stable within a period ≥ 2 years, or if changes typical <strong>of</strong> benign tumors, such as<br />

calcifications within tumor limits are visible, further diagnostics is not recommended [9].<br />

Contrast <strong>CT</strong> is more accurate and it reveals a few millimeter large tumor-like changes, or<br />

carcinogenic changes that are invisible in X-ray pictures, where they are overshadowed by<br />

mediastinum or interstitial densities. <strong>CT</strong> helps identify not only primary tumors, but also lymph<br />

node metastasis. However, tomography scanning has minor importance in tumor staging in<br />

atelectatic areas (tumor is hard to separate from the atelectatic focus), or in the staging <strong>of</strong><br />

small infiltrations by cancer <strong>of</strong> the mediastinum (which is essential for diagnostics and<br />

therapy). The value <strong>of</strong> <strong>CT</strong> is also limited in the evaluation metastasis to mediastinal lymph<br />

nodes because morphologic criteria (enlargement <strong>of</strong> lymph node > 1cm as a criterion <strong>of</strong><br />

metastatic diagnosis) are characterized by low sensitivity. Magnetic resonance is not<br />

commonly used in the diagnostics <strong>of</strong> lung changes. Recently, <strong>PET</strong> has been used more and<br />

more <strong>of</strong>ten for N-staging. <strong>PET</strong> helps to find small metastasis to mediastinal lymph nodes, and<br />

to determine the extension <strong>of</strong> the tumor in atelectatic lung areas with more precision than<br />

<strong>CT</strong>. Whole-body <strong>PET</strong> helps detect metastatic foci outside <strong>of</strong> the chest. As the availability <strong>of</strong><br />

<strong>PET</strong> is still low in Poland, N-staging <strong>of</strong> mediastinal lymph nodes is done using invasive methods,<br />

such as mediastinoscopy [6, 9].<br />

3.1.5.4. Microscopic diagnosis<br />

Microscopic diagnosis can be assessed on the basis <strong>of</strong> cytological or histopathological<br />

examinations. In 50–70% <strong>of</strong> cases, cytological evaluation <strong>of</strong> sputum is sufficient, in 80–90% <strong>of</strong><br />

cases microscopic diagnosis can be made by bronch<strong>of</strong>iberoscopy. Bronchoscopy permits<br />

macroscopic evaluation <strong>of</strong> the bronchi and sampling for histopathological tests. What’s<br />

more, in this procedure bronchial stem washings are collected, which, when assessed<br />

cytologically, may help diagnose changes invisible to imaging studies or bronchoscopy.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Image-guided (<strong>CT</strong> or ultrasound-guided) biopsy <strong>of</strong> peripheral lesions through the chest wall is<br />

recommended. When fluid is detected in the pleural cavity, it can be sampled using<br />

transthoracal biopsy to be examined cytologically. If the studies above are insufficient for<br />

diagnosis, supplemental mediastinoscopy, thoracoscopy or thoracotomy should be carried<br />

out. If lymph nodes are enlarged, biopsy or surgical excision may be recommendable [4, 6, 8,<br />

9].<br />

3.1.5.5. Other tests<br />

Biochemical tests identify anemia, or high concentration <strong>of</strong> alkaline phosphatase (for<br />

bone metastasis) or high levels <strong>of</strong> liver enzymes in blood (for liver metastasis), as well as<br />

improper sodium concentration or low serum calcium concentration. For small cell cancer,<br />

bone marrow trepanobiopsy to evaluate bone marrow infiltration is indicated. Also,<br />

increased concentration level <strong>of</strong> cancer markers, such as carcinoembryonic antigen (CEA),<br />

CK 19 section <strong>of</strong> cytokeratin (CYFRA 21.1), or neuron-specific enolase (NSE) can be found In<br />

cancer patients [6].<br />

3.1.5.6. General principles in small cell lung cancer diagnosis<br />

The diagnostic procedure depends on the disease stage, clinical condition, and outcome<br />

<strong>of</strong> additional studies. If based on the clinical picture, small cell lung cancer is suspected,<br />

diagnosis should be obtained using the least invasive method possible. In patients with<br />

suspected non-carcinogenic exudation in pleura, fluid should be taken for cytological<br />

examination using biopsy through the chest wall, and if infiltration <strong>of</strong> the chest wall and<br />

parietal pleura is suspected, pleura can be sampled by blind biopsy through the chest wall<br />

with the use <strong>of</strong> a special needle. When histopathological tests do not confirm suspicions in<br />

patients with fluid in the pleural cavity, thoracoscopy should be done for sampling as the next<br />

step. In patients with suspected lung cancer and single out-<strong>of</strong>-lung, presumably metastatic,<br />

lesions, diagnosis should be done by large-needle biopsy, or a specimen <strong>of</strong> the lesion should<br />

be taken. In patients with extensive infiltration <strong>of</strong> the mediastinum, diagnosis should be<br />

established by examining tissue specimens taken from the mediastinum using the most<br />

efficacious method possible (bronchoscopy with peribronchial biopsy, biopsy through the<br />

chest wall or mediastinoscopy). Patients diagnosed with a single peripheral tumor-like,<br />

presumably carcinogenic, lesion, in whom the disease seems to be at an early stage, are<br />

qualified to surgery and intraoperational tumor evaluation [6, 8, 9].<br />

3.1.5.7. General principles <strong>of</strong> treatment<br />

Primary prevention <strong>of</strong> lung cancer by promoting non-smoking attitudes is <strong>of</strong> paramount<br />

importance. Smokers should be identified, because quitting smoking curbs the risk <strong>of</strong> lung<br />

cancer and as such it should be recommended. Smokers should have access to<br />

psychosocial and behavioral therapies whenever recommended [4, 6, 8, 9, 11].<br />

43


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

44<br />

3.1.5.8. Non-small cell cancer<br />

The choice <strong>of</strong> treatment method depends mainly on the tumor stage. Surgery is the basic<br />

type <strong>of</strong> treatment. For stages I and II, complete tumor resection with lobotomy or<br />

pulmectomy is performed. Stage IIIA tumors have border-line operability. Stage IIIB tumors are<br />

no longer operable. The extent <strong>of</strong> resection is determined by the R-trait: R0 are macro- and<br />

microscopic resections; R1 is macroscopic-only resection; and R2 is incomplete resection in<br />

macroscopic evaluation. Not all patients can qualify for operation. The necessary<br />

requirements include: the simple exercise tests such as walking up and down the stairs, the 6-<br />

minute walk test or, finally, spirometric examination (mainly the expiratory volume in the first<br />

second and vital capacity). Postoperative complications include respiratory and<br />

cardiovascular insufficiency. Operation can be supplemented with other therapeutic<br />

procedures such as neoadjuvant radiotherapy, postoperational radiotherapy,<br />

postoperational radiotherapy coupled with chemotherapy, or chemotherapy. The routine<br />

procedure in inoperable patients is radiotherapy. Radical radiotherapy is an option for<br />

patients with limited-mass tumor, without exudation in the pleural cavity and with an efficient<br />

respiratory track. Doses >65 Gy and fraction doses 1.8–2.5 Gy are administered. Because the<br />

radiated tumor is located in the chest in the environment <strong>of</strong> radiosensitive structures <strong>of</strong> critical<br />

importance for life, such as the heart and the great vessels, so-called three-dimensional<br />

conformal radiation therapy id used (3D-CRT). With this technology, the radiation area can<br />

be adjusted to the tumor shape with great precision, ensuring maximum preservation <strong>of</strong> the<br />

surrounding structures. In some cases integrated treatment is used (chemotherapy with<br />

radiotherapy). However, this policy increases the early toxicity <strong>of</strong> therapy. With locally<br />

advanced lung tumors, palliative radiotherapy is used to help shrink the tumor, alleviate pain,<br />

and reduce the symptom <strong>of</strong> tumor pressure on the adjacent structures. Radiation doses are<br />

reduced (20–30 Gy in a few fractions or a single higher dose, e.g. 10 Gy). Chemotherapy uses<br />

platinum derivatives (cisplatin, carboplatinum), ifosphamide, mitomycine, or alcaloids<br />

(vinblastin). New drugs include taxoids (paclitaxel, docetaxel), gemcitabine, vinorelbin. Two-<br />

drug combinations comprising platinum-based agents are currently the preferred standard<br />

regimens. Drugs that act on epithelial cell growth factor receptor tyrosine kinase inhibitors,<br />

such as gefitinib or erlotinib, arouse great expectations. Other additional treatment methods<br />

that may be applied to specific clinical conditions are brachytherapy, electrocoagulation,<br />

phototherapy, cryotherapy, or laser therapy. They affect cancer tissues locally. In the case <strong>of</strong><br />

atelectasis caused by tumor growing into and narrowing the bronchial lumen, bronchial<br />

patency should be improved and a stent placed in the narrowing. In patients with bone<br />

metastasis, radiotherapy may significantly decrease pain. Symptomatic brain metastasis is an<br />

indication for radiation or excision. For patients in good general condition, diagnosed with<br />

disease dissemination, chemotherapy should be considered [4, 6, 8, 9, 11].


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

3.1.5.9. Small cell carcinoma<br />

Small cell carcinoma is the most aggressive type <strong>of</strong> cancer. Chemotherapy is the<br />

fundamental treatment. More active drugs against this cancer type include cisplatin,<br />

etoposide, cyclophosfamide, doxorubicin, vincristin. The most <strong>of</strong>ten applied is a combination<br />

<strong>of</strong> cisplatin with etoposide; or, less frequently, vincristin with doxorubicin and<br />

cyclophosphamide. Chemotherapy leads to total r<strong>emission</strong> in about 80–90% <strong>of</strong> patients. The<br />

therapy is supplemented by radiotherapy <strong>of</strong> the primary cancer focus. The recommended<br />

doses are 55–60 Gy. A combination <strong>of</strong> chemotherapy and radiotherapy helps attain a higher<br />

rate <strong>of</strong> long-term survivals. In patients with total r<strong>emission</strong> <strong>of</strong> cancer in the chest, additional<br />

brain radiation is used. The recommended doses are 25–30 Gy. This procedure reduces the<br />

number <strong>of</strong> distant metastasis.<br />

The efficacy <strong>of</strong> surgical treatment is low with small cell cancer, and is always combined<br />

with chemotherapy. For recurrent limited-stage cancer, chemotherapy can be repeated.<br />

The regimen is repeated not earlier than 3 months after therapy is completed; in other cases<br />

different drug combinations are administered [4, 6, 8, 9, 10, 11].<br />

3.1.6. Prognosis<br />

Lung cancer has a poor prognosis. Epidemiologic studies indicate that the 5-year survival<br />

rate in Poland for malignant lung cancer patients is 6.1% in men and 6.8% in women, and is<br />

lower than in the European population [5.13]. Mortality as a cause <strong>of</strong> malignant lung tumors is<br />

still growing in Poland. However, this factor is expected to stabilize in the nearest years [5].<br />

The survival period in non-small lung cancer depends on the disease stage at diagnosis.<br />

The only efficacious therapeutic method is surgical treatment. 5-year survivals after complete<br />

resection <strong>of</strong> stage I, II and IIIA tumors are 50–70%, 30–50% and 10–30% respectively. Patients<br />

with disseminated non-small lung cancer do not survive a year. In the majority <strong>of</strong> patients<br />

diagnosed with non-small lung cancer, disseminated disease is found at the first diagnosis.<br />

The median period <strong>of</strong> survival without treatment is 6–8 weeks, only a few percent <strong>of</strong> patients<br />

survive 5 years [6] 2 .<br />

2 1. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin 2005; 55 (2): pp 74–108.<br />

2. Evidence-based Cancer Prevention: Strategies for NGOs – A UICC Handbook for Europe. http://www.uicc.org<br />

3. The Globocan 2002 database: http://www-dep.iarc.fr<br />

4. Roszkowski-ŚliŜ K. Cancer płuca – aspekty epidemiologiczne i diagnostyczne. Pneumonologia 2005;<br />

http://www.terapia.com.pl/archiwum<br />

5. Narodowy program zwalczania chorób nowotworowych. ZałoŜenia i cele operacyjne 2006–2015;<br />

http://www.mz.gov.pl/wwwfiles/ma_struktura/docs/zalozenia_ustawy_o_npzchn.pdf<br />

6. Szczeklik A. i wsp. Choroby wewnętrzne. Kraków, Medycyna Praktyczna, 2005.<br />

7. Coran R, Kumar V, Ramzi T. Robbins pathologic basis <strong>of</strong> disease. Elsevier/Saunders, 1999, wydanie 6.<br />

8. Pawlicki M i wsp. Leczenie nowotworów. Bielsko Biała, Alfa Medica Press, 1996.<br />

9. Diagnosis and management <strong>of</strong> lung cancer: ACCP evidence-based guidelines. American College <strong>of</strong> Chest<br />

Physicians. Chest 2003; 123 (1): pp 1S–337S.<br />

10. Adjei AA, Marks RS, Bonner JA. Current guidelines for the management <strong>of</strong> small cell lung cancer. Mayo Clin<br />

Proc 1999; 74: pp 809–881.<br />

45


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

46<br />

3.3. Lymphomas<br />

3.3.1. Epidemiology<br />

3.3.1.1 Non-Hodgkin’s lymphomas<br />

In 2002, 301,000 new cases <strong>of</strong> non-Hodgkin’s lymphomas were reported in the world. They<br />

represents 2.8% <strong>of</strong> all tumors and occur most <strong>of</strong>ten in developed societies (50.5% <strong>of</strong> all cases<br />

in the world), with the highest incidence rates reported in Australia and North America,<br />

average-level incidence in Europe (excluding Eastern Europe) and on Pacific Islands, and<br />

relatively low incidence in Asia and Eastern Europe [1]. Men are affected 1.5 times more ften<br />

than women [2]. In Poland, 7 out <strong>of</strong> 100,000 men and 4 out <strong>of</strong> 100,000 women get non-<br />

Hodgkin’s lymphomas every year [3]. Mortality in men is 3.4, in women it is and 1.8 per<br />

100,000. AIDS Patients suffer from non-Hodgkin’s lymphomas 1000 times more frequently [2].<br />

3.3.1.2 Hodgkin’s disease<br />

Hodgkin’s disease represents 1% <strong>of</strong> all the tumors diagnosed, and 10% <strong>of</strong> haematopoietic<br />

tumors. The disease is more frequent with young people aged 20-30 [4]; men are diagnosed<br />

1.6 times more frequently [1]. In developing countries, the disease affects mainly children and<br />

adults, and the histopathological texture is dominated by the mixed cell type. In developed<br />

countries, mainly young adults suffer from Hodgkin’s disease; in the histopathological picture,<br />

nodular sclerosis Hodgkin’s disease prevails.<br />

The highest incidence was reported in developed countries (North America, Europe), the<br />

lowest in Asian populations [1]. 2002 data indicate that annually, 2.7 out <strong>of</strong> 100,000 men and<br />

3 out <strong>of</strong> 100,000 women develop Hodgkin’s disease in Poland. At the same time, the mortality<br />

in Poland amounts 1.1 in men and 0.6 among women [3].<br />

3.3.2. Risk factors<br />

The etiopathogenesis <strong>of</strong> non-Hodgkin’s lymphomas is unclear. Clinical data suggest a<br />

relationship with viral infections. For example, non-Hodgkin’s lymphoma develops in 10% <strong>of</strong><br />

AIDS patients. Other risk factors are immunological disturbances, such as organ transplants,<br />

immunosuppressive therapy, autoimmune diseases, or congenital immunodeficiency<br />

11. Clinical Guideline 24. Lung cancer: the diagnosis and treatment <strong>of</strong> lung cancer. National Institute for Clinical<br />

Excellence 2005; http://www.nice.org.uk<br />

12. Rojek M i wsp. Biopsja aspiracyjna cienkoigłowa w diagnostyce nowotworów płuc. Medycyna Rodzinna 2002;<br />

19; http://www.borgis.pl/czytenia<br />

13. Coleman MO et al. Eurocare-3 summary: cancer survival in Europe at the end <strong>of</strong> 20th century. Ann Oncol 2003;<br />

1: pp 128–149.<br />

14. Tyczyński JE, Bray F, Parkin DM. Lung cancer In Europe. European Network <strong>of</strong> Cancer Registries, International<br />

Agency for Research on Cancer, ENCR Cancer Fact Sheets 2002; 1; http://www.encr.com.fr/lung-factsheets.pdf<br />

15. AJCC cancer staging manual, sixth edition. http://www.cancerstaging.org/education/tnmschema/lung.ppt


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

syndromes. The hypothesis whereby ultraviolet radiation increased the risk <strong>of</strong> non-Hodgkin’s<br />

lymphomas is reliable but requires epidemiological confirmation.<br />

Hodgkin’s disease is associated with genetic predisposition; however, environmental<br />

factors seem to have influence on it as well. In Europe and North America, epidemiological<br />

studies point to a relationship between the incidence <strong>of</strong> Hodgkin’s disease and low socio-<br />

economic status. A significant role in the disease is attributed to Epstein-Barr virus, as well as<br />

HIV. Occupational hazards may stimulate the disease (woodcutter) [1, 5].<br />

3.3.3. Symptoms<br />

In most cases patients seek initial medical help due to asymmetrical enlargement <strong>of</strong> lymph<br />

nodes, usually cervical or supraclavicular. Raised temperature or recurrent fever, weakness,<br />

drenching night sweats, and severe itching <strong>of</strong> the skin may appear. The disease causes body<br />

weight losses. Physical examination reveals lymph node enlargement, also liver or spleen<br />

enlargement may occur. If the central nervous system is involved, dysaesthesia, palsies and<br />

pains are observed [4, 5].<br />

3.3.4. Histopathological picture<br />

A generally acceptable and commonly used division distinguishes between two types <strong>of</strong><br />

lymphomas: Hodgkin’s disease and all other non-Hodgkin’s lymphomas.<br />

Hodgkin’s disease is a lymphoma that is histologically unique for the presence <strong>of</strong> Reed-<br />

Sternberg cells, which arise probably from the monoclonal line, most frequently from B<br />

lymphocytes. Non-Hodgkin’s lymphomas are the other neoplastic proliferations from B or T-<br />

lymphocytes.<br />

3.3.4.1 Hodgkin’s disease<br />

Typical histopathological symptoms include giant Reed-Sternberg cells arising from single<br />

nuclear Hodgkin cells, and cellular polymorphism, i.e. an accumulation <strong>of</strong> varied cells <strong>of</strong> the<br />

lymphatic system. Four histological types <strong>of</strong> Hodgkin lymphoma are distinguished (Rye’s<br />

classification) [4, 5, 6]:<br />

1. nodular sclerosis type (82%): the most frequent type, with best prognosis,<br />

characterized by the presence <strong>of</strong> nodes built <strong>of</strong> pathologic cells;<br />

2. mixed cellularity type (14%): varied cells in various proportions present in infiltrations,<br />

accompanied by slight sclerosis;<br />

3. lymphocyte-rich classical (3%): the lymph nodes tissue is composed largely <strong>of</strong><br />

lymphocytes, rarely other cells;<br />

47


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

48<br />

4. lymphocyte depleted type (1%) – the lymph nodes are characterized by a small<br />

number <strong>of</strong> lymphocytes and distinct sclerosis.<br />

3.3.4.2 Non-Hodgkin’s lymphomas<br />

Non-Hodgkin’s lymphomas are divided in the Killonian classification based on their<br />

morphological and immunohistochemic features. Table 4 presents the classification [4].<br />

Table 4.<br />

Killonian classification <strong>of</strong> non-Hodgkin’s lymphomas<br />

Low grade lymphomas<br />

B- cell T - cell<br />

Lymphocyte lymphomas Lymphocyte lymphomas<br />

Chronic lymphatic leukaemia and prolymphocytic<br />

leukaemia<br />

Chronic lymphatic leukaemia and prolymphocytic<br />

leukaemia<br />

Hairy cell leukaemia Mycosis fungoides<br />

Lymphoplasmacytic lymphoma (immunocytoma) Sezary’s syndrome<br />

Plasmacytic lymphomas Angioimmunoblastic lymphoma<br />

Centroblastic-centrocytic lymphomas T-zone lymphoma<br />

Nodular diffused or non-diffused lymphomas Small cell pleomorphic lymphoma<br />

Inter mediate grade lymphomas<br />

Some types <strong>of</strong> lymphoplasmacytoidal lymphomas<br />

Pleomorphic lymphomas<br />

Centroblastic-centrocytic centroblast-rich lymphomas<br />

High grade lymphomas<br />

Centroblastic lymphomas Pleomorphic transitional and large cell lymphomas<br />

Immunoblastic lymphomas Immunoblastic lymphomas<br />

Anaplastic large cell lymphomas (Ki-1+) Anaplastic large cell lymphomas (Ki-1+)<br />

Burkitt’s lymphoma<br />

Lymphoblastic lymphoma Lymphoblastic lymphoma<br />

3.3.5. Staging<br />

Table 5 presents the Ann Arbor system <strong>of</strong> Hodgkin’s disease and non-Hodgkin’s<br />

lymphomas. This classification is commonly used for disease staging.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Table 5.<br />

Hodgkin’s disease and non-Hodgkin’s lymphomas staging according to Ann Arbor staging system<br />

Disease stage Characteristics<br />

I<br />

II<br />

III<br />

IV<br />

A<br />

B<br />

Involvement <strong>of</strong> a single lymph node group and/or a single organ or site other than the lymph<br />

node (IE)<br />

Involvement <strong>of</strong> two or more lymph node groups on the same side <strong>of</strong> the diaphragm (II) or<br />

limited involvement <strong>of</strong> one extranodal organ or site with the involvement <strong>of</strong> one or more<br />

group <strong>of</strong> lymph nodes on the same side <strong>of</strong> the diaphragm (IIE)<br />

Involvement <strong>of</strong> lymph nodes on both sides <strong>of</strong> the diaphragm (III), may be accompanied by<br />

limited involvement <strong>of</strong> an estranodal organ or site (IIIE), or involvement <strong>of</strong> spleen (IIIS) or both<br />

(IIIES)<br />

Diffused or disseminated involvement <strong>of</strong> one or more extranodal organs or tissues with or<br />

without the involvement <strong>of</strong> lymph nodes<br />

No general symptoms<br />

With general symptoms: weight loss <strong>of</strong> 10% or more within the last 6 months and/or fever<br />

above 38 C and/or drenching night sweats<br />

The International Lymphoma Study Group (ILSG) has proposed another classification <strong>of</strong><br />

lymphomas based on the American and European (Killonian) classification, called the<br />

Revised European-American Classification <strong>of</strong> Lymphoid Neoplasms (REAL). A combination <strong>of</strong><br />

morphological, immunophenotypical, genetic and clinical features was used in the<br />

classification [4].<br />

Table 6.<br />

REAL classification <strong>of</strong> lymphoid tumors<br />

B-cell tumors T and NK-cell tumors<br />

Benign and chronic (survival without treatment calculated in years)<br />

B-cell chronic lymphocytic leukaemia (CLL), small<br />

lymphocytic lymphoma (SLL), prolymphocytic leukaemia<br />

(PLL)<br />

Lymphoplasmacytic lymphoma<br />

Hairy cell leuekmia<br />

Multiple myeloma<br />

Splenic marginal zone lymphoma (splenic lymphoma<br />

with villous lymphocytes – SLVL)<br />

Extranodal marginal zone lymphoma<br />

Mucosa associated lymphatic tissue lymphoma – MALT<br />

lymphoma<br />

Nodal marginal zone lymphoma<br />

Follicular lymphoma-FH, follicle centre lymphoma – FCL<br />

Mantle-cell lymphoma – MCL<br />

Benign and diffused lymphomas and leukaemias<br />

Benign non-nodular lymphomas<br />

Benign nodular lymphomas<br />

T-cell CLL, PLL<br />

large granular lymphocyte leukaemia – LGL<br />

Mycosis fungoides<br />

Malignant lymphomas (survival without treatment counted in months)<br />

Diffused large B-cell lymphoma – DLCL<br />

Anaplastic large-cell lymphoma – ALCL<br />

Peripheral T-cell lymphomas (many types)<br />

Aggressive lymphomas (survival without treatment counted in weeks)<br />

49


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Lymphoblastic leukaemias and lymphomas \ from pre-B<br />

cells<br />

50<br />

Burkitt’s lymphoma<br />

lymphocyte predominance<br />

Hodgkin’s disease<br />

3.3.6. Diagnostics and treatment<br />

3.3.6.1 Additional tests<br />

Pre-T Lymphoblastic leukaemias and lymphomas<br />

Adult T-cell leukaemia and lymphoma (HTLV1)<br />

Hodgkin-like ALCL<br />

Classical Hodgkin's disease<br />

nodular sclerosis – NS<br />

mixed cellularity – MC<br />

lymphocyte depletion – LD<br />

lymphocyte-rich – LR<br />

As regards biochemical diagnostics, note needs to be taken <strong>of</strong> a high erythrocyte<br />

sedimentation rate (ESR, Biernacki’s test) where results have two- or three-digit values. The<br />

increased concentration <strong>of</strong> lactic dehydrogenase (LDH) is observed. As regards<br />

morphological blood examinations, a low number <strong>of</strong> lymphocytes and a high proportion <strong>of</strong><br />

eosynophils or monocytes are observed. At advanced stages, anemia and other<br />

disturbances in hematopoiesis may occur as a result <strong>of</strong> bone marrow involvement.<br />

3.3.6.2 Diagnostic imaging<br />

A chest radiogram may reveal involvement <strong>of</strong> mediastinal lymph nodes, apparent from a<br />

widening <strong>of</strong> the central shade. In such situations, <strong>CT</strong> <strong>of</strong> the chest is required to make diagnosis<br />

complete. In bones roentgenograms presence, osteolysis foci can be detected and<br />

ultrasound scanning <strong>of</strong> the abdomen may reveal enlarged lymph nodes [4].<br />

3.3.6.3 Cytological and histopathological study<br />

Material collected by bone marrow biopsy from a Hodgkin’s disease patient usually does<br />

not show any deviation from standard; in 10% <strong>of</strong> patients, involvement <strong>of</strong> bone marrow with<br />

Reed-Sternberg and Hodgkin cells present in it is visible. With non-Hodgkin’s lymphomas,<br />

apart from examination <strong>of</strong> histological bioptates <strong>of</strong> bone marrow, it is diagnostically critical to<br />

do cytochemical tests, which are aimed at identifying nuclear or cytoplasmic enzymes<br />

typical <strong>of</strong> specific cells types and stages <strong>of</strong> their development. A specimen <strong>of</strong> tumor tissue<br />

(e.g. from lymph nodes) need to be taken for a histopathological study to confirm the<br />

diagnosis <strong>of</strong> lymphoma.<br />

3.3.6.4 Molecular tests<br />

The method <strong>of</strong> immunophenotyping bone marrow cells in a flow cytometer to assess<br />

superficial antigens is helpful in lymphomas differentiation [4]. Superficial antigens<br />

examinations is also done using the immunohistochemical method. At present, the most<br />

important techniques in the diagnostics <strong>of</strong> lymphomas are genetic methods, such as


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

polymerase chain reaction or fluorescence in situ hybridization. They are helpful in detecting<br />

features such as monoclonality, chromosomal aberrations and genetic mutations [7].<br />

3.3.6.5 Treatment <strong>of</strong> Hodgkin’s disease<br />

Treatment depends on the disease stage. For stages I and II, radiotherapy with isotope<br />

60Co or megavolt X-ray therapy are used. It achieves total r<strong>emission</strong> in 80–90% <strong>of</strong> patients. For<br />

stage IIB, radiotherapy and chemotherapy bring better effects (in keeping with the MOPP<br />

protocol: mechloretamine, vincristine, procarbasine, prednisone). For stage III A, radiation or<br />

radiation with chemotherapy are used. For stages IIIB and IV, chemotherapy based on the<br />

protocol above or a combination <strong>of</strong> doxorubicin, bleomycin, vinblastin, dacarbasine (ABVD)<br />

are administered. Another protocol is BEACOPP chemotherapy.<br />

If disease recurs after chemotherapy, other drug regimens such as DHAP and EPOCH are<br />

implemented.<br />

For some patients, bone marrow auto transplant can be considered. Bone marrow cells<br />

are first taken and frozen, next high-dose chemotherapy and radiotherapy are implemented,<br />

and subsequently the bone marrow is cleaned and transplanted.<br />

Treatment is not neutral for the patient and has a number <strong>of</strong> serious side effects, mainly<br />

complications resulting from direct action <strong>of</strong> cytostatic drugs and radiotherapy, such as<br />

secondary infections, local radiation-induced changes. Distant consequences <strong>of</strong> the therapy<br />

include secondary tumors (leukemia, lymphomas, cancers), cardiovascular complications<br />

(pericarditis, cardiosclerosis), pulmonary complications (radiation-induced pneumonia,<br />

pulmonary fibrosis), radiation-induced disorders <strong>of</strong> the thyroid gland, reproductive<br />

disturbances (risk <strong>of</strong> infertility increases) [4].<br />

When therapy is completed, patients should be examined for complete/incomplete<br />

r<strong>emission</strong>. The necessary measures include repeated physical examination, monitoring <strong>of</strong><br />

general blood count with smear, and diagnostic re-imaging if abnormalities were detected<br />

before. In case <strong>of</strong> partial radiological response, it is advisable to evaluate disease activity by<br />

histopathological assessment <strong>of</strong> lesion biopsy, or, as minimum, repeated radiological studies.<br />

Whenever possible, a <strong>PET</strong> study should be done for patients with partial response to identify<br />

those with high risk.<br />

After treatment, follow-up check-ups and examination are recommended every 3 months<br />

in the first year, every 6 months in the following 3 years, and then once a year. Laboratory<br />

tests and chest radiogram are done after 6, 12 and 24 months after therapy is finished. <strong>CT</strong><br />

and other radiological studies are recommended to confirm r<strong>emission</strong>, but further<br />

observation is not recommended, with the exception <strong>of</strong> assessment <strong>of</strong> residual disease.<br />

Follow-up examination <strong>of</strong> the thyrotropine hormone is recommended one, two and five years<br />

after neck irradiated. In the case <strong>of</strong> chest radiation, women at premenopausal age should<br />

51


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

be especially observed for secondary breast tumors. mammography is indicated for women<br />

aged 40–50 [8].<br />

52<br />

3.3.6.6 Treatment <strong>of</strong> non-Hodgkin’s lymphomas<br />

Therapy is complex and depends, among others, the on grade <strong>of</strong> lymphoma malignancy,<br />

intensity <strong>of</strong> clinical symptoms, and location <strong>of</strong> changes. For giant cell lymphoma, which<br />

represent 30–50% <strong>of</strong> all non-Hodgkin’s lymphomas, the CHOP protocol in combination with<br />

rituximab is standard treatment. Stem cells transplantation is still an experimental method. If<br />

treatment fails for patients in good general condition, conventional treatment followed by<br />

high-dose therapy and stem cells administration is recommended. Other chemotherapy<br />

regimens used are DHAP, ESHAP, or EPOCH. Treatment can be supplemented with<br />

radiotherapy. If patients cannot take high-dose treatment, it is recommended that<br />

conventional doses <strong>of</strong> drugs and radiotherapy are implemented. Also, palliative methods are<br />

used, depending on recommendations. Follicular lymphoma is the second leading non-<br />

Hodgkin’s lymphoma. In a small proportion <strong>of</strong> patients with stage I disease, the first-choice<br />

therapy is radiotherapy. Systemic therapy is acceptable in patients with large tumors, as for<br />

higher disease stages. In the majority <strong>of</strong> patients with grade III and IV disease, fully effective<br />

treatment is not possible. Since spontaneous r<strong>emission</strong> occurs in 15–20% <strong>of</strong> patients,<br />

chemotherapy is launched if B-symptoms, disturbed hematopoiesis or disease progression are<br />

observed. When long-term r<strong>emission</strong> is obtained, administration <strong>of</strong> rituximab with other<br />

regimens such as COP, CHOP should be considered. An alternative therapy is fludarabin or<br />

chlorambucil. When contraindications to chemotherapy exist, monoclonal antibodies are an<br />

option. Research suggests that beta-interferon as supportive therapy can produce good<br />

effects. Bone marrow auto transplant is still an experimental method for this tumor type.<br />

The disease should be monitored after treatment every 3 months in the first 2 years, every 6<br />

months in the next 3-year period, and then once a year. Peripheral blood tests are done 3, 6,<br />

12 and 24 months after therapy, and whenever alarming symptoms are observed. Thyroid<br />

gland functions are assessed after neck radiation and should be performed after 1, 2 and 5<br />

years. Diagnostic imaging is repeated 3, 6, 12 and 24 months after treatment is completed.<br />

Patients who had their chest irradiated, especially people under 25 or before the<br />

postmenopausal age, should be under observation and take screening examinations for<br />

secondary breast cancer are necessary [9, 10, 11].<br />

3.3.7. Prognosis<br />

Prognosis in Hodgkin’s disease is relatively good: about 80% <strong>of</strong> patients survive more than 5<br />

years and 60% more than 10 years. Negative prognostic factors include massive nodular<br />

changes, progression <strong>of</strong> the disease during therapy or within a year after chemotherapy,


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

advanced disease (stage IV), general symptoms, histopathological LD type <strong>of</strong> Hodgkin’s<br />

disease, age over 40 years, male sex.<br />

Prognosis for non-Hodgkin’s lymphomas depends on their grade <strong>of</strong> malignancy. High-<br />

grade lymphomas represent more than 70% <strong>of</strong> all lymphomas. Centrocytic lymphoma and T-<br />

cell proliferations have a poor prognosis. Prognosis is adversely affected by general<br />

symptoms such as fever, sweats, or loss <strong>of</strong> body weight. Five-year survivals for non-Hodgkin’s<br />

lymphomas generally represent about 40–59%. [4, 12] 3 .<br />

3.4. Esophageal cancer<br />

3.4.1. Epidemiology<br />

It is estimated, that esophageal cancer is the eighth leading tumor in the world: in 2002,<br />

462,000 new cases (4.2%) were reported. At the same time it is the sixth leading cause <strong>of</strong><br />

death (5.7%, 386,000 deaths in 2002). Men suffer more <strong>of</strong>ten, almost exclusively older than 40.<br />

In 2002, the incidence in men was estimated at 315,000 globally, and in women – 146,000<br />

globally [1]. However, in Asian and African countries, the incidence in men and women is the<br />

same [1]. In Poland, about 1300 cases <strong>of</strong> esophageal cancer are diagnosed every year [2].<br />

The incidence and mortality rates for the Polish female and male populations are: 0.9 and 0.8<br />

per 100,000, and 6.0 and 4.7 per 100,000 respectively [3].<br />

Considerable geographical differentiation <strong>of</strong> incidence rates is observed. The highest<br />

incidence is reported in China, the lowest in western Africa. Other regions with relatively high<br />

rates in southern and eastern Africa, south-eastern Asia and Japan [1]. The geographical<br />

differentiation in esophageal cancer incidence proves that environmental factors are<br />

instrumental [1].<br />

3 1. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin 2005; 55 (2): pp 74–108.<br />

2. Herold G at all. Medycyna wewnętrzna. PZWL 2001.<br />

3. The Globocan 2002 database: http://www-dep.iarc.fr<br />

4. Skotnicki AB, Nowak WS. Podstawy hematologii. Kraków, Medycyna Praktyczna 1998.<br />

5. The Merck manual <strong>of</strong> diagnosis and therapy. http://www.merck.com<br />

6. Coran R, Kumar V, Ramzi T. Robbins pathologic basis <strong>of</strong> disease. Elsevier/Saunders, 1999, 6th edition.<br />

7. B7CSH guidelines on nodal non-Hodgkin’s lymphoma – draft 2, August 2002.<br />

8. ESMO minimum clinical recommendations for diagnosis, treatment and follow-up <strong>of</strong> Hodgkin disease. Ann Oncol<br />

2005; 16: pp 54–55.<br />

9. ESMO Minimum Clinical Recommendations for diagnosis, treatment and follow-up <strong>of</strong> newly diagnosed large cell<br />

on-Hodgkin’s lymphoma. Ann Oncol 2005; 16: pp 58–59.<br />

10. ESMO Minimum Clinical Recommendations for diagnosis,treatment and follow-up <strong>of</strong> relapsed large cell non-<br />

Hodgkin’s ’s lymphoma. Ann Oncol 2005; 16: pp 60–61.<br />

11. ESMO Minimum Clinical Recommendations for diagnosis, treatment and follow-up <strong>of</strong> newly diagnosed follicular<br />

lymphoma. Annals <strong>of</strong> Oncology 2005; 16: pp 56–57.<br />

12. Coleman MP I at all. EUROCARE-3 summary: cancer survival in Europe at the end <strong>of</strong> the 20th century. Annals <strong>of</strong><br />

Oncology 2003; 14: pp 128–149.<br />

53


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

54<br />

The incidence <strong>of</strong> squamous carcinoma, which is a major type <strong>of</strong> this tumor, has been<br />

stable recently. However, the increasing incidence <strong>of</strong> esophageal adenocarcinoma is<br />

observed [1, 4]. The most probable explanation <strong>of</strong> this phenomenon is the increasing<br />

occurrence <strong>of</strong> Barrett’s esophagus, or gastroesophageal reflux-based precancerous<br />

conditions, which is very likely to be associated with the increasing obesity in societies.<br />

3.4.2. Risk factors<br />

Special note is taken <strong>of</strong> environmental factors in the development <strong>of</strong> squamous cell<br />

carcinomas. It is believed that poor diet may predispose to this tumor type. What is more,<br />

vitamin deficit in diet (vitamins A, C, rib<strong>of</strong>lavin, thiamine, pyridoxine), trace elements, food<br />

contamination with mould or high content <strong>of</strong> nitrates are also pointed up. Other risk factors<br />

include alcohol abuse and cigarettes smoking. Chronic diseases <strong>of</strong> esophagus, such as<br />

esophagitis, achalasi or chronic celiac disease, as well as genetic factors may predispose to<br />

the disease. It is also a well known fact that black people get the disease more frequently [1,<br />

4].<br />

Esophageal sdenocarcinoma <strong>of</strong>ten develops from a change in the esophageal lining<br />

(mucosa) called Barrett’s esophagus. Typically, Barrett’s esophagus occurs when normal<br />

squamous lining cells <strong>of</strong> the esophagus are replaced by columnar cells in response to long-<br />

term exposure to damage from chronic gastroesophageal reflux [4]. Research on<br />

carcinogenic transformation confirms that one <strong>of</strong> the essential staged that directly precede<br />

invasive esophageal cancer is high-grade dysplasia [5].<br />

3.4.3. Symptoms<br />

Initially, the disease is asymptomatic. Ailments caused by the esophagus blocked by tumor<br />

mass occurs later on. Problems with swallowing (74%) are the key symptom: first the<br />

swallowing solid food is impaired, then liquid food become hard to swallow as well. Other<br />

symptoms include loss <strong>of</strong> body weight (57%) and painful swallowing (17%). Less frequent<br />

ailments are dyspnoea, cough, hoarse throat and chest pain [2].<br />

3.4.4. Histopathological picture<br />

The most frequently diagnosed are squamous cell cancers. Cancers <strong>of</strong> this type usually are<br />

well or moderately differentiated. Regardless <strong>of</strong> the degree <strong>of</strong> differentiation, when<br />

symptoms occur the disease is advanced and prognosis is poor. Adenocarcinomas, the<br />

incidence <strong>of</strong> which is growing, are usually located in lower esophagus and may infiltrate<br />

stomach. The majority <strong>of</strong> tumors <strong>of</strong> this type produce mucus and are histologicaly similar to<br />

gastric cancer. Sometimes, the visible texture resembles small intestine cells [4]. Also,


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

gastrointestinal stromal sarcomas are located in the esophagus, where they transform into<br />

benign growths [6].<br />

3.4.5. Staging<br />

Table 7 presents the TNM staging system for esophageal cancer, as well as tumor stages<br />

according to the American Joint Committee on Cancer (AJCC) [7].<br />

Table 7.<br />

TNM and AJCC staging system <strong>of</strong> esophageal cancer<br />

Primary tumor<br />

Tx Primary tumor cannot be assessed<br />

T0 No evidence <strong>of</strong> primary tumor<br />

Tis Carcinoma in situ<br />

T1 Tumor invades lamina propria or submucosa<br />

T2 Tumor invades muscular membrane<br />

T3 Tumor invades adventitia<br />

T4 Tumor invades adjacent structures<br />

Regional lymph nodes<br />

Nx Regional lymph nodes cannot be assessed<br />

N0 No regional lymph node metastasis<br />

N1 Regional lymph node metastasis<br />

Distant metastasis<br />

MX Distant metastasis cannot be assessed<br />

M0 No distant metastasis<br />

M1 Distant metastasis<br />

Tumors <strong>of</strong> the lower thoracic esophagus<br />

M1a Metastasis in celiac lymph nodes<br />

M1b Other distant metastasis<br />

Tumors <strong>of</strong> the midthoracic esophagus<br />

M1a Not applicable<br />

M1b Nonregional lymph nodes and/or other distant metastasis<br />

Tumors <strong>of</strong> the upper thoracic esophagus<br />

M1a Metastasis in cervical nodes<br />

M1b Other distant metastasis<br />

AJCC stage groupings<br />

Grade 0 Tis N0 M0<br />

Grade I T1 N0 M0<br />

Grade IIA T2 N0 M0<br />

T3 N0 M0<br />

Grade IIB T1 N1 M0<br />

T2 N1 M0<br />

Grade III T3 N1 M0<br />

T4 any N M0<br />

Grade IV Any T any N M1<br />

Grade IVA Any T any N M1a<br />

Grade IVB Any T any N M1b<br />

55


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

56<br />

3.4.6. Diagnostics and treatment<br />

3.4.6.1 Physical examination<br />

On physical examination, enlargement <strong>of</strong> lymph nodes (e.g. cervical) or liver can be<br />

found.<br />

3.4.6.2 Diagnostic imaging<br />

A contrast radiological study <strong>of</strong> esophagus is done as the first diagnostic step. It helps<br />

detects tumor in the lumen <strong>of</strong> the esophagus, which obstructs the flow <strong>of</strong> a contrast medium<br />

through the esophagus to the stomach.<br />

Precise imaging studies can be done by <strong>CT</strong>. It seems that <strong>PET</strong> diagnostics is more accurate<br />

than <strong>CT</strong> in disease staging. A fusion <strong>of</strong> these two methods, <strong>PET</strong> and <strong>CT</strong>, is a promising tool that<br />

may significantly improve the accuracy <strong>of</strong> pre-surgery diagnostics [8].<br />

3.4.6.3 Endoscopy<br />

It is the first-choice examination in esophageal tumor diagnostics. It detects lesions in the<br />

mucous membrane <strong>of</strong> the esophagus that are invisible in other diagnostic methods. It also<br />

helps to sample tissues from suspected areas to confirm cancer histopathologically.<br />

Endoscopically, esophageal cancer may have polypoid (60%), ulcerative (25%) or flat,<br />

intraparietally spreading form (15%) [4]. The newest diagnostic method is endosonography,<br />

which not only helps assess the mucous membrane, like conventional endoscopy, but also,<br />

through the use <strong>of</strong> ultrasound, allows a non-invasive evaluation <strong>of</strong> the depth <strong>of</strong> carcinogenic<br />

infiltration and size <strong>of</strong> regional lymph nodes [2].<br />

3.4.6.4 Cancer markers<br />

Identification <strong>of</strong> cancer markers is characterized by low sensitivity and specificity.<br />

Carcinoembryonic antigen (CEA) or other cancer antigens (CA 19-9, CA 125) tests can be<br />

helpful in monitoring disease recurrences.<br />

3.4.6.5 Histopathological study<br />

Histopathological examinations are indispensable for final diagnosis <strong>of</strong> esophageal tumor.<br />

3.4.6.6 Radical treatment<br />

Qualified to radical treatment are patients with no metastasis found. Operational excision<br />

<strong>of</strong> carcinogenic infiltration is used, ranging from mucous membrane ablation to esophagus<br />

resection. Another standard policy is pre-surgery chemoradiotherapy or radiochemotherapy<br />

if the patient is inoperable. The choice <strong>of</strong> method depends on the disease stage. 5-<br />

fluorouracyl and cisplatin are used in the main. For stage I, initial surgery or


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

radiochemotherapy is implemented. For grade T4 N1 M0, chemoradiotherapy is used, but for<br />

grade T2/3 N0/1 M0/1a, surgical treatment with support therapy, or chemoradiotherapy plus<br />

another surgery, or sole chemoradiotherapy be considered [2, 9].<br />

3.4.6.7 Palliative treatment<br />

The aim <strong>of</strong> palliative treatment is to reduce pain, and to improve nutrition and quality <strong>of</strong><br />

life. Various methods are used to reduce disorder <strong>of</strong> esophageal passage or to support<br />

esophageal patency. This function is performed by radiochemotherapy in some patients, in<br />

other endoscopic procedures or esophageal stenting is used. If total occlusion <strong>of</strong> the<br />

esophagus occurs, gastrostomy may be required to feed the patient.<br />

3.4.7. Prognosis<br />

Esophageal cancer has a very poor prognosis (5-year survivals < 20%). Estimated 5-years<br />

survivals in the Polish population are 5.6% in men and 4.1% in women [10, 11] 4 .<br />

3.5. Cancer <strong>of</strong> the female genitals<br />

3.5.1. Endometrial cancer<br />

3.5.1.1 Epidemiology<br />

The geographical distribution <strong>of</strong> uterine corpus cancer resembles the distribution <strong>of</strong><br />

ovarian cancer. However, prognosis for cervical tumor is definitely more optimistic than for<br />

ovarian cancer. In 2002, 199,000 cases <strong>of</strong> uterine cancer were reported, which represents<br />

3.9% <strong>of</strong> all tumors in women. The incidence <strong>of</strong> uterine cancer in the world is 6.5 for per 100,000<br />

[1]. Also that year, 50,000 deaths caused by this disease were reported, which represents 1.7%<br />

<strong>of</strong> all cancer-related deaths in women. The annual death toll <strong>of</strong> uterine cancer is 1.6 per<br />

100,000 women. This tumor is the most commonly diagnosed in women in their<br />

4 1. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin 2005; 55 (2): pp 74–108.<br />

2. Szczeklik A. i wsp. Choroby wewnętrzne. Kraków, Medycyna Praktyczna 2005.<br />

3. The Globocan 2002 database: http://www-dep.iarc.fr<br />

4. Coran R, Kumar V, Ramzi T. Robbins pathologic basis <strong>of</strong> disease. Elsevier/Saunders, 1999, 6th edition.<br />

5. Postępy w chirurgii przełyku w 2005 roku. Medycyna Praktyczna 2006; 01: pp 9–14.<br />

6. National Cancer Institute. Esophageal cancer. http://cancerweb.ncl.ac.uk/cancernet/100089.html<br />

7. AJCC cancer staging manual, sixth edition;<br />

http://www.cancerstaging.org/education/tnmschema/esophagus.ppt<br />

8. Kato H, Miyazaki T, Nakajima M, et al. The incremental effect <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography on diagnostic<br />

accuracy in the initial staging <strong>of</strong> esophageal carcinoma. Cancer 2005; 103: pp 148–156.<br />

9. ESMO Minimal Clinical Recommendations for diagnosis, treatment and follow-up <strong>of</strong> esophageal cancer. Ann<br />

Oncol 2005; 16: pp 26–27.<br />

10. Narodowy program zwalczania chorób nowotworowych. ZałoŜenia i cele operacyjne 2006–2015.<br />

http://www.mz.gov.pl/wwwfiles/ma_struktura/docs/zalozenia_ustawy_o_npzchn.pdf<br />

11. Coleman MO et al. Eurocare-3 summary: cancer survival in Europe at the end <strong>of</strong> 20th century. Annals <strong>of</strong><br />

Oncology 2003; 1: pp 128–149.<br />

57


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

postmenopausal years. Globally, 91% <strong>of</strong> new cases are diagnosed in women aged 50 or<br />

older [1]. The highest incidence is observed in Europe (11.8 to 12.5 per 100,000) and in North<br />

America (22 per 100,000). The lowest incidence is reported in southern and eastern Asia<br />

(including Japan) and in Africa (incidence below 3.5 per 100,000) [1]. Based on 2002<br />

epidemiological data, the annual incidence <strong>of</strong> uterine cancer in Poland is 13 per 100,000<br />

women. It is also the cause <strong>of</strong> death <strong>of</strong> 2.8 per 100,000 women (age-standardized values) [2].<br />

58<br />

3.5.1.2 Risk factors<br />

Endometrial cancer is more frequent in developed countries, where the consumption <strong>of</strong><br />

dietary fat is higher. The most important risk factor is obesity, which increases the risk 3–10<br />

times. Endometrial cancer occurs in case <strong>of</strong> imbalance between progesterone and<br />

estrogen, with estrogen dominance (hormone replacement therapy, obesity, polycystic<br />

ovarian syndrome, nulliparity, late menopause, estrogen-secreting tumors, and lack <strong>of</strong> or rare<br />

ovulations. Irradiation <strong>of</strong> the pelvis, breast or ovarian cancers in the family increase the risk <strong>of</strong><br />

endometrial cancer.<br />

Endometrial hyperplasia usually precedes endometrial cancer.<br />

3.5.1.3 Symptoms<br />

In over 90% <strong>of</strong> patients pathological bleedings from the genitals (postmenopausal<br />

bleedings, irregular intermenstrual bleedings) are observed. It is estimated, that about 30% <strong>of</strong><br />

women with abnormal postmenopausal bleedings have endometrial cancer.<br />

3.5.1.4 Histopathological picture<br />

In over 80% <strong>of</strong> cases, histopathological texture corresponds to adenocarcinoma; 60% <strong>of</strong><br />

these are well differentiated, and 20% are non-differentiated. Infiltration <strong>of</strong> deeper layers <strong>of</strong><br />

the wall, as well as the risk <strong>of</strong> distant metastasis depend on the stage <strong>of</strong> the tumor. Sarcomas<br />

represent 5% <strong>of</strong> all tumors <strong>of</strong> the uterine corpus, including leyomiosarcomas, mixed<br />

mesodermal tumors, and endometrial stromal sarcomas. Sarcomas have a poorer prognosis<br />

[3,4].<br />

3.5.1.5 Staging<br />

The tumor staging system takes into account the grade <strong>of</strong> histological differentiation (G<br />

grades), as well as features <strong>of</strong> clinical advancement as established during operation, among<br />

them depth <strong>of</strong> infiltration, involvement <strong>of</strong> the cervix, lymph node or peritoneal cavity<br />

metastasis [3,4].<br />

Table 8 presents the 1988 generally approved staging system <strong>of</strong> the International<br />

Federation <strong>of</strong> Gynecology and Obstetrics (FIGO).


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Table 8.<br />

FIGO staging for endometrial cancer<br />

Grade Description<br />

Stage 0 In situ cancer<br />

Stage I IA – tumor confined to the corpus uteri, length <strong>of</strong> the uterine cavity does not exceed 8 cm<br />

IB – tumor confined to the corpus uteri , length <strong>of</strong> the uterine cavity exceeds 8 cm<br />

Stage II Cancer involves the corpus and the cervix<br />

Stage III Cancer extends outside the uterus, but is confined to the true pelvis<br />

Stage IV Cancer extends outside the true pelvis or infiltrates bladder mucosa or rectum (confirmed by<br />

histological study <strong>of</strong> samples)<br />

IVA – invasion <strong>of</strong> adjacent organs<br />

IVB – metastasis to distant organs<br />

3.5.1.6 Diagnostics and treatment<br />

In some patients, diagnosis is done based on anomalies revealed in a cytological smear. A<br />

microscopic study <strong>of</strong> cervical canal and uterine cavity scrapings obtained by fractionated<br />

microabrasia is necessary for diagnosis. Also, gynecological per-vaginam and per-rectum<br />

examination <strong>of</strong> true pelvis, and a chest radiogram are necessary [3, 4].<br />

Prophylaxis <strong>of</strong> uterine cancer involves observation and proper treatment <strong>of</strong> women in the<br />

high risk group. Abnormal bleedings are indications for curettage <strong>of</strong> the uterine cavity. In<br />

young patients, anovulation cycles should receive treatment. When adenomatous<br />

hyperplasia <strong>of</strong> the endometrium is identified, hormonal therapy or simple uterine<br />

hysterectomy can be attempted. The basic procedure for uterine cancer is surgery,<br />

recommended for all patients with stage I–III disease and no contraindications to surgery.<br />

During the procedure, it is necessary to sample material from the peritoneal cavity for<br />

cytological examination, and to revise the peritoneal cavity carefully, taking specimens <strong>of</strong><br />

the suspected places. In high risk patients, pelvic and periaortic lymph nodes should be<br />

sampled for histopathological examination or excised completely whenever metastasis is<br />

suspected. Supplementary treatment involves 40–50 Gy radiation and is applied to patients<br />

with lymph node or adnexa metastasis, deep infiltration <strong>of</strong> the muscular layer, or surgical<br />

margin involvement. Local radiation through brachytherapy may be considered.<br />

Radiotherapy is the only procedure for non-operational stages III and IV. Hormonal therapy is<br />

the first-choice systemic treatment; its efficacy is 75% if hormonal receptor is detected.<br />

Usually, progestogens are used. The hormonal therapy is the first-choice therapy in advanced<br />

stages; however whether it should be part <strong>of</strong> supplementary treatment is open to discussion.<br />

Moreover, the administration <strong>of</strong> progestogens should be considered in patients with<br />

hypertension, obesity and diabetes, because the therapy may increase these disorders [3, 4].<br />

59


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

60<br />

3.5.1.7 Prognosis<br />

Uterine cancer has a high survival rate: 86% in the United States and 78% in Europe, which<br />

is higher than the survival expected for breast cancer, especially in developing countries.<br />

Therapeutic effects depend on the tumor stage: for stage grade I, the 5-year survival is 80–<br />

90%; for stage II: 30–70%; for stage III: 15–40%; and for stage IV: 0–3% [1, 5] 5 .<br />

3.5.2. Cervical cancer<br />

3.5.2.1 Epidemiology<br />

Cervical cancer is one <strong>of</strong> the most frequent malignant tumors in women, and the seventh<br />

leading malignant tumor in the world, but second leading in women. In 2002, 493,000 new<br />

cases <strong>of</strong> cervical cancer and 274,000 deaths were reported. Cervical cancer is more<br />

frequent in developing countries (83% <strong>of</strong> all cases), where it represents 15% <strong>of</strong> all tumors in<br />

women. In developed countries, this tumor represents only 3.8% <strong>of</strong> new cases. The highest<br />

incidence rate is reported in regions such as Africa, Latin America, southern Asia, the<br />

Caribbean Islands. In developed countries, the incidence <strong>of</strong> cervical cancer is 14.5 per<br />

100,000 (age-standardized values). However, the incidence in regions such as Europe and<br />

North America has changed remarkably after screening programs were implemented. A very<br />

low incidence is observed in China (6.8 per 100,000), Western Asia (5.8 per 100,000). Mortality<br />

is clearly lower compared to incidence. The mortality to incidence ratio is 55% globally [1].<br />

The screening examinations implemented in the past few years pushed down the incidence<br />

and mortality rates significantly in well-developed countries. In some developing countries<br />

these rates are going down too (e.g. China) [1]. In Poland, the 2002 incidence <strong>of</strong> cervical<br />

cancer was 18.4 per 100,000 women, and morbidity stood at 7.8 per 100,000 (age-<br />

standardized values) [2].<br />

3.5.2.2 Risk factors<br />

One <strong>of</strong> the main etiological factors is infection with the human papilloma virus (HPV).<br />

Other infections that increase the risk <strong>of</strong> cervical cancer include HIV and Chlamydia [3].<br />

Recent epidemiological studies carried out with the use <strong>of</strong> high-sensitivity HPV detection<br />

methods have showed a correlation between the incidence <strong>of</strong> virus detection and the<br />

incidence <strong>of</strong> cervical cancer. Other risk factors include multiparity, smoking and oral<br />

contraceptives [1]. Women who do not eat enough fruit and vegetables, or are overweight,<br />

5 1. Parkin DM, Bray F, Ferlay J, Pisani P. Global Cancer Statistics, 2002. CA Cancer J Clin 2005; 55; pp 74–108.<br />

2. Globocan 2002 database. http://www-dep.iarc.fr/<br />

3. The Merck manual <strong>of</strong> diagnosis and therapy. www.merck.com<br />

4. Pawlicki M I wsp. Leczenie nowotworów. Bielsko-Biała, Alfa Medica Press, 1996.<br />

5. Coleman MP, Gatta G, Verdecchia A, Estève J, Sant M, Storm H, Allemani C, Ciccolallo L, Santaquilani M, Berrino<br />

F and the EUROCARE Working Group. EUROCARE-3 summary: cancer survival in Europe at the end <strong>of</strong> the 20th<br />

century. Ann Oncol 2003; 14: pp 128–149.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

are at greater risk. Low socioeconomic status predisposes to cervical cancer. Family<br />

predisposition to cervical cancer is observed in some patients [3].<br />

3.5.2.3 Symptoms<br />

The earliest and <strong>of</strong>ten the first symptom is abnormal bleeding from the genitals. it is <strong>of</strong>ten<br />

ignored by doctors and patients as incidental. Other symptoms include pain in the lumbar-<br />

sacral region, irregular menstruations. In 5–10% <strong>of</strong> cases no suspicious symptoms are found.<br />

Late symptoms are the reason why diagnoses <strong>of</strong> cervical cancer are late. The advanced<br />

stages may be dominated by dysuria, urgency, symptoms <strong>of</strong> renal insufficiency caused by<br />

pressure on the uterus from cancerous infiltration, or enlarged lymph nodes <strong>of</strong> the true pelvis<br />

[4].<br />

3.5.2.4 Histopathological picture<br />

Microscopic examination usually detects squamous cell carcinoma, which represents 95%<br />

<strong>of</strong> all malignant tumors <strong>of</strong> the cervix. Adenocarcinoma is less frequent (4–5%), and including<br />

varieties such as endometrioidal, mesonefroidal and adenosquamous cancer. Non-<br />

differentiated cancer, cylindroma, sarcoma, melanoma and lymphoma are very rare [4].<br />

3.5.2.5 Staging<br />

The system introduced by the International Federation <strong>of</strong> Gynecology and Obstetrics<br />

(FIGO) is generally used in staging. Table 9 presents the classification.<br />

61


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Table 9.<br />

FIGO staging for cervical cancer<br />

Stage Description<br />

Stage 0<br />

Stage I<br />

Stage II<br />

Stage III<br />

Stage IV<br />

62<br />

Carcinoma in situ<br />

Cervical carcinoma confined to uterus (extension to corpus should be disregarded)<br />

IA – invasive cancer, preclinical, diagnosed only by microscopy<br />

IA1 – minimal microscopical infiltration <strong>of</strong> the lining<br />

IA2 – invasion into lining less than 5 mm in depth, measured from the basement membrane, less<br />

than 7 mm in horizontal spread<br />

IB – invasion <strong>of</strong> cancer is greater (microscopically) than IA2, usually visible infiltration confined to<br />

the cervix only<br />

Cervical carcinoma invades beyond the uterus but not to the pelvic wall or to the lower third <strong>of</strong><br />

the vagina<br />

IIA – cancer invades vaginal fornix and its walls, but the lower third <strong>of</strong> the vagina<br />

IIB – cancer invades parametrium, but does not spread to the pelvic walls<br />

Carcinoma extends to the pelvic wall and/or involves the lower third <strong>of</strong> the vagina (in per rectum<br />

examination free space between cervical tumor and pelvis wall is not found). All cases <strong>of</strong><br />

hydronephrosis or nonfunctioning kidneys should be included.<br />

IIIA –cancer involves the lower third 3 <strong>of</strong> the vagina, without infiltrating parametrium to the bone<br />

IIIB – cancer invades parametrium to the bone or causes hydronephrosis or dysfunction <strong>of</strong> the<br />

kidney<br />

Cancer extends beyond the true pelvis or invades bladder mucosa and/or rectum bullous edema<br />

is not sufficient not classify a tumor as stage IV by cytoloscopy; only specimens from bladder or<br />

rectum tested by microscopy may qualify a tumor as stage IV (or a cytological study <strong>of</strong> urine)<br />

IVA – cancer invades bladder or rectal mucosa and/or extends beyond the true pelvis<br />

IVB – distant metastasis<br />

3.5.2.6 Diagnostics and treatment<br />

The key diagnostic phase is gynecological examination. Cytology is <strong>of</strong> paramount<br />

importance in early detection <strong>of</strong> cervical cancer in situ. The Papanicolau classification has<br />

been the most widely accepted in gynecological cytology, and is still in use in Poland. Grade<br />

I corresponds to normal picture, grade III requires more extensive diagnostics and treatment<br />

(features <strong>of</strong> strong inflammatory state and cellular dysplasia), group IV corresponds to<br />

squamous cell carcinoma in situ, and grade V is invasive cancer. Colposcopy helps locate<br />

abnormal lesions. Ultimate diagnosis can be made by histopathological examination <strong>of</strong><br />

material from cervix obtained by curettage <strong>of</strong> the cervical canal and uterine corpus.<br />

A number <strong>of</strong> other diagnostic studies are performed but unnecessary for diagnosis.<br />

Diagnostics is supplemented by imaging studies, predominantly <strong>CT</strong> <strong>of</strong> the pelvis or magnetic<br />

resonance imaging. <strong>PET</strong> diagnostics is used if cancerous dissemination is suspected. Venal<br />

urography helps expose urine flow disorders, which can be caused by tumor mass pressure<br />

[3].<br />

Basic treatment policies for invasive cervical cancer are surgery and radiotherapy. The<br />

choice <strong>of</strong> method depends on the tumor stage. Radiotherapy is a radical therapy method<br />

for all stages; however, surgical treatment is used with patients with stage I and, to some<br />

extent, stage IIA disease. The extent <strong>of</strong> surgery depends on the tumor stage. In non-invasive


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

states cryotherapy and laser therapy are used. Cone biopsy (excision <strong>of</strong> a wedge <strong>of</strong> the<br />

cervix uteri that includes the tumor zone through the vagina), is hardly ever the only<br />

treatment. The extent <strong>of</strong> surgery ranges from simple hysterectomy to surgical excision <strong>of</strong> all<br />

organs and tissues <strong>of</strong> the true pelvis. Outcomes <strong>of</strong> surgery and radiation are similar. Other<br />

approach is to combine radiotherapy and chemotherapy. At present multi-drug regimens<br />

are used that are based on cisplatin and contain combinations <strong>of</strong> many other cytostatic<br />

drugs. They attain about 50% response to treatments, but total regressions are rare and short-<br />

lived. Cisplatin, paclitaxel, topotecan, ifosphamide or fluorouracil are most frequently used [3,<br />

4].<br />

3.5.2.7 Prognosis<br />

Recently, the survival rate has improved for cervical cancer. However, that is not true for<br />

countries in Eastern Europe. It is estimated that the average 5-year survival in Europe is about<br />

60%, and differences between particular territories in Europe are relatively small. A survival<br />

rate higher than in other European countries is observed in the Czech Republic.<br />

Epidemiological data point to the crucial role <strong>of</strong> screening programs in improving survival<br />

rates in Western Europe. Screening programs comprise cytological examination <strong>of</strong> cervical<br />

smears [5] 6 .<br />

3.5.3. Ovarian cancer<br />

3.5.3.1 Epidemiology<br />

Ovarian cancer is the sixth leading tumor diagnosed in women and the seventh leading<br />

cause <strong>of</strong> death caused by malignant tumors in women. In 2002, 204,000 new cases <strong>of</strong> ovarian<br />

cancer and 125,000 deaths were reported globally. Incidence is higher in developed<br />

countries, where it is reaches 9 per 100,000, with the exception <strong>of</strong> Japan, where incidence is<br />

the lowest (6.4 per 100,000) [5]. The highest incidence rates are observed in the United States,<br />

Europe and Israel, while the lowest in Japan [3]. The incidence <strong>of</strong> ovarian cancer in Europe is<br />

18 per 100,000 people, and mortality is 12 per 100,000 people yearly [2]. In Eastern Europe,<br />

ovarian cancer is the sixth leading malignant tumor, but in Western Europe it is the third [1].<br />

The median age <strong>of</strong> women diagnosed with ovarian cancer 63. Morbidity increases with age,<br />

and peaks in the eighth decade <strong>of</strong> life [2]. In 2002 in Poland, the incidence <strong>of</strong> ovarian cancer<br />

in women was 12.5 per 100,000 and morbidity was 6.8 per 100,000.<br />

6 1. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin 2005; 55 (2): pp 74–108.<br />

2. The Globocan 2002 database: http://www-dep.iarc.fr<br />

3. American Cancer Society. Detailed guide: cervical cancer. http://www.cancer.org/<br />

4. Pawlicki M i wsp. Leczenie nowotworów. Bielsko Biała, Alfa Medica Press, 1996.<br />

5. Coleman MO et al. Eurocare-3 summary: cancer survival in Europe at the end <strong>of</strong> 20th century. Ann Oncol 2003; 1:<br />

pp 128–149.<br />

63


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

64<br />

Figure 3 presents the incidence and mortality rates for ovarian cancer in selected regions<br />

<strong>of</strong> the world.<br />

Figure 3.<br />

Incidence and mortality rates for ovarian cancer in selected geographic regions [7]<br />

3.5.3.2 Risk factors<br />

The disease occurs at any age, more frequently in patients above age 40. Also familiar<br />

incidence <strong>of</strong> disease is rare. The disease is more frequent in highly industrialized countries.<br />

3.5.3.3 Symptoms<br />

Ovarian cancer develops asymptomatically for a long time, and symptoms are<br />

uncharacteristic. As a result, the disease is usually diagnosed at advanced stages (FIGO<br />

stages III or IV) [4]. The most common symptoms are: pressure symptoms in the abdomen,<br />

gain <strong>of</strong> body weight, a feeling <strong>of</strong> fullness, bloating, belching. Bleeding from vagina and pain<br />

on one side <strong>of</strong> the hypogastrium may be present [4]. Acute symptoms are occasional and<br />

involve torsion <strong>of</strong> the ovarian peduncle and hemorrhage.<br />

3.5.3.4 Histopathological picture<br />

Histological classification is done based on the WHO division. Three types <strong>of</strong> ovarian tumors<br />

are distinguished: epithelial ovarian tumors, primary germ cell tumors and ovarian stromal


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

tumors. Occasionally, sarcomas or metastatic tumors (breast cancers, gastrointestinal<br />

cancers, cancers <strong>of</strong> other genital organs and lymphomas) can be found in this location. The<br />

most frequent are epithelial ovarian tumors. It is estimated that they represent over 85% <strong>of</strong> all<br />

ovarian tumors.<br />

3.5.3.5 Staging<br />

When no distant metastasis is found, staging requires laparotomy. If the disease seems<br />

limited, it is essential to proceed with laparotomy and assessment <strong>of</strong> diaphragm, intestines,<br />

peritoneum, regional lymph nodes, omentum or to sample washings <strong>of</strong> the peritoneal cavity<br />

for a histopathological study [4, 5].<br />

The International Federation <strong>of</strong> Gynecology and Obstetrics (FIGO) and the American Joint<br />

Committee on Cancer (AJCC) have proposed a staging system that is presented in Table 10.<br />

Table 10.<br />

AJCC and FIGO staging for ovarian cancer<br />

Stage Characteristics<br />

I<br />

II<br />

III<br />

IV<br />

Stage I: Growth limited to the ovaries.<br />

IA: Growth limited to one ovary; no ascites. No tumor on the external surface; capsule intact.<br />

IB: Growth limited to both ovaries: no ascites. No tumor on the external surfaces; capsules intact.<br />

IC: Tumor either stage Ia or Ib but with tumor on surface <strong>of</strong> one or both ovaries, or with capsule<br />

ruptured; or with ascites present containing malignant cells or with positive peritoneal washings.<br />

Stage II: Growth involving one or both ovaries with pelvic extension.<br />

IIa Extension and/or metastases to the uterus and/or fallopian tubes.<br />

IIb Extension to other pelvic tissues.<br />

IIc Tumor either stage IIa or IIb. but with tumor on surface <strong>of</strong> one or both ovaries: or with capsule(s)<br />

ruptured; or with ascites present containing malignant cells or with positive peritoneal washings<br />

Differences in inclusion criteria for stages IC or IIC have impact on diagnosis. It is critical to assess<br />

whether damage to capsule is a result <strong>of</strong> tumor invasion or surgical intervention, or whether cancer<br />

cells are present in peritoneal fluid, or in peritoneal washings.<br />

Stage III: Tumor involving one or both ovaries with positive peritoneal implants outside the pelvis<br />

and/or positive retroperitoneal or inguinale nodes. Superficial liver metastasis equals stage III. Tumor<br />

is limited to the true pelvis but with histologically proven malignant extension to small bowel or<br />

omentum.<br />

IIIa Tumor grossly limited to true pelvis with negative nodes but with histologically confirmed<br />

microscopic seeding <strong>of</strong> abdominal peritoneal surfaces.<br />

IIIb Tumor <strong>of</strong> one or both ovaries with histologically confirmed implants <strong>of</strong> abdominal peritoneal<br />

surfaces none exceeding 2 cm in diameter. Nodes are negative.<br />

IIIc Abdominal implants greater than 2 cm in diameter and/or positive retroperitoneal or inguinale<br />

nodes.<br />

Stage IV: Growth involving one or both ovaries, with distant metastases. If pleural effusion is present,<br />

there must be positive cytology. Parenchymal liver metastases equal stage IV<br />

65


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

66<br />

3.5.3.6 Diagnostics and treatment<br />

If growth is advanced, the abdomen perimeter increases, fluid is present in the abdomen,<br />

sometimes tumor is palpable on physical examination [4]. On gynecological examination,<br />

enlarged adnexa are palpable; sometimes tumor is palpable in the true pelvis. Enlargement<br />

<strong>of</strong> the ovaries, tumor-like or cysts-like lesions within adnexa or fluid in the abdominal cavity<br />

can be detected by ultrasonography <strong>of</strong> the abdominal cavity, or by transvaginal ultrasound.<br />

Ultrasound scanning is only tentative, and <strong>CT</strong> <strong>of</strong> the abdominal cavity is recommended.<br />

Rectoscopy and cystoscopy are done before surgery for initial staging and to plan<br />

therapeutic procedures.<br />

Ca-125 cancer marker is relatively specific and sensitive. However, its usefulness in initial<br />

diagnostics is limited, but is used to monitor the course <strong>of</strong> the disease [4]. High concentration<br />

<strong>of</strong> Ca-125 is associated with high odds <strong>of</strong> positive ovarian cancer diagnosis, but negative<br />

tests do not exclude the disease. Definite diagnosis <strong>of</strong> the ovarian cancer is based on a<br />

histopathological study <strong>of</strong> samples taken at surgery, <strong>of</strong>ten as part <strong>of</strong> laparotomy.<br />

Basic treatment is surgery, which can be sufficient only for stage IA. Other patients require<br />

systemic therapy, which is supplementary for stages I and II, and basic for stages III and IV<br />

following a cytoreduction procedure or exploratory laparotomy. Chemotherapy is<br />

implemented 4 weeks after surgery, with the regimen based on platinum derivatives<br />

(cisplatin, carboplatinum). Radiotherapy is not the first-choice therapy in ovarian cancer,<br />

however radiation is used in many centers as alternative supplementary treatment [7].<br />

3.5.3.7 Prognosis<br />

The 5-year survival rate in Europe is estimated at 37%, but remarkable differences between<br />

countries are observed [8]. The mortality rate for this tumor type has remained unchanged<br />

over the past few years [1]. The 10-year survival rate for stage IA disease may reach up to 86–<br />

92% 7 .<br />

7 1. Evidence-based cancer prevention: strategies for NGOs- a UICC handbook for Europe. http://www.uicc.org<br />

2. ESMO Minimum Clinical Recommendations for diagnosis, treatment and follow-up <strong>of</strong> epithelial ovarian<br />

carcinoma. Ann Oncol 2005; 16: pp 13–15.<br />

3. Coleman MP, Esteve J, Damiecki P, Arslan A, Renard H. Trends in cancer incidence and mortality. IARC Sci Publ.<br />

1993; 121: pp 1–806.<br />

4. Senn HJ I wsp. Kompendium onkologii. PZWL 1995.<br />

5. Parkin DM, Bray F, Ferlay J, Pisani P. Global Cancer Statistics, 2002. CA Cancer J Clin 2005; 55: pp 74–108.<br />

6. Globocan 2002 database. http://www-dep.iarc.fr/<br />

7. Pawlicki M i at all. Leczenie nowotworów. Bielsko Biała, Alfa Medica Press, 1996.<br />

8. Sant M, Aareleid T, Berrino F, Bielska Lasota M, Carlin P. M, Faivre J, Grosclaude P, Hédelin G, Matsuda T, Møller H,<br />

Möller T, Verdecchia A, Capocaccia R, Gatta G, Micheli A, Santaquilani M, Roazzi P, Lisi D and the EUROCARE<br />

Working Group. EUROCARE-3: survival <strong>of</strong> cancer patients diagnosed 1990–94 – results and commentary.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

3.6. Thyroid gland cancer<br />

3.6.1. Epidemiology<br />

In 2002, 141,000 cases <strong>of</strong> thyroid gland cancer were reported globally. It is one <strong>of</strong> the few<br />

tumor type that is more frequent in women than in men. The global incidence rate <strong>of</strong> thyroid<br />

cancer is 1.3 for men and 3.3 for women. This tumor represents 2.1% <strong>of</strong> all tumors in women [1,<br />

2]. Diagnostic practice seems to have impact on the frequency <strong>of</strong> positive thyroid cancer<br />

diagnosis (e.g. histological post-mortem assessment <strong>of</strong> goiter may have significant impact on<br />

the frequency <strong>of</strong> positive diagnosis). This may be the reason for the high incidence rate<br />

observed in the United States (incidence rates for women and men: 8.1 and 3.9) [1, 2]. The<br />

rates are also high in Australia/New Zealand, Japan and Central America [1]. In Poland. the<br />

incidence <strong>of</strong> thyroid cancer is 1.4 per 100,000 in men, and 4.1 per 100,000 in women. Disease-<br />

related mortality is 0.3 per 100,000 in men and 0.6 per 100,000 in women.<br />

3.6.2. Risk factors<br />

To estimate prognosis, four factors should be considered: age, sex, histological<br />

differentiation grade and tumor stage. Also, prognosis depends on whether or not treatment<br />

has been implemented. The age <strong>of</strong> the patient at initial positive diagnosis is important<br />

especially in the case <strong>of</strong> papillary and follicular thyroid gland cancers. The risk <strong>of</strong> death<br />

increases in patients over 40 years <strong>of</strong> age and is higher in children. It has been pointed out<br />

that male sex may affect prognosis negatively. Prognosis for papillary cancer is better than<br />

for follicular cancer; however, with regard to parameters such as sex and extension <strong>of</strong><br />

primary tumor, survival in both cases is similar. Poorly differentiated follicular cancers, as well<br />

as Hürtle cell cancer have a bad prognosis. The size <strong>of</strong> the tumor at the outset <strong>of</strong> treatment is<br />

a basic prognostic factor. Invasion <strong>of</strong> adjacent tissues, lymph node metastasis and distant<br />

metastasis significantly influence prognosis [4].<br />

3.6.3. Symptoms<br />

The disease is <strong>of</strong>ten asymptomatic. Symptoms can be divided into local, systemic and<br />

paraneoplastic. The most frequent symptom is a tumor in the neck. Also, enlarged cervical<br />

lymph nodes, swallowing problems, hoarseness or dyspnoea may appear. The presence <strong>of</strong><br />

generalized symptoms may be indicative <strong>of</strong> a higher stage or aggressiveness <strong>of</strong> thyroid<br />

cancer. Systemic symptoms include loss <strong>of</strong> body weight, and for medullary thyroid cancer -<br />

washy diarrheas. Paraneoplastic syndromes are grossly typical <strong>of</strong> medullary thyroid cancer,<br />

and are associated with imbalance <strong>of</strong> the adrenocorticotropic hormone and calcitonine [3,<br />

4].<br />

67


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

68<br />

3.6.4. Histopathological picture<br />

Five histopathological types <strong>of</strong> thyroid cancer are distinguished: papillary, follicular, Hürtle<br />

cell, medullary and anaplastic. Papillary (60–70%) and follicular cancer (10–15%) are the most<br />

common types. The other types represent 20% <strong>of</strong> cancer cases total [3].<br />

3.6.5. Staging<br />

Table 11, presents the TNM staging system designed by the American Joint Committee on<br />

Cancer (AJCC) [5].<br />

Table 11.<br />

TNM staging system by AJCC<br />

Primary tumor<br />

Tx Primary tumor cannot be assessed<br />

T0 No evidence <strong>of</strong> primary tumor<br />

T1 Tumor is ≤2 cm in greatest dimension , limited to thyroid gland<br />

T2 Tumor is >2 cm in greatest dimension, but ≤ 4 cm, limited to thyroid gland<br />

T3 Tumor is >4 cm in greatest dimension and tumor limited to thyroid gland or any tumor<br />

with minimal extrathyroid extension (extension to sternothyroid muscle s<strong>of</strong>t or<br />

perithyroid tissues)<br />

T4 T4a – tumor <strong>of</strong> any size extending beyond the thyroid capsule to invade subcutaneous<br />

s<strong>of</strong>t tissues, larynx, trachea, esophagus, or recurrent laryngeal nerve<br />

T4b – tumor invades paravertebral fascia or carotid artery, or mediastinal vessels<br />

Or anaplastic thyroid carcinomas are considered T4:<br />

T4a – intrathyroid anaplastic carcinoma, surgically resectable<br />

T4b – extrathyroid anaplastic carcinoma, surgically unresectable<br />

Regional lymph nodes (regional lymph nodes are cervical and upper mediastinal nodes)<br />

Nx Regional lymph nodes cannot be assessed<br />

N0 No regional lymph node metastasis<br />

N1 Regional lymph nodes metastasis<br />

N1a – metastasis to pretracheal, paratracheal, and prelaryngeal lymph nodes<br />

N1b – metastasis to cervical (unilateral, bilateral or superior mediastinal) lymph nodes<br />

Distant metastasis<br />

MX Distant metastasis cannot be assessed<br />

M0 No distant metastasis<br />

M1 Distant metastasis<br />

AJCC stage groupings for thyroid cancer<br />

Younger than 45<br />

Stage I Any T, any N, M0<br />

Stage II Any T, any N, M1<br />

Age 45 and over<br />

Stage I T1 N0 M0<br />

Stage II T2 N0 M0<br />

Stage III T3 N0 M0<br />

Follicular or papillary carcinoma


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

T1–T3 N1a M0<br />

Stage IV A T4a N0–N1a M0<br />

T1–T4a N1b M0<br />

Stage IV B T4b any N M0<br />

Stage IV C Any T, any N M1<br />

Stage I T1 N0 M0<br />

Stage II T2 N0 M0<br />

Stage III T3 N0 M0<br />

T1–T3 N1a M0<br />

Stage IVA T4a N0–N1a M0<br />

T1–T4a N1b M0<br />

Stage IVB T4b any N M0<br />

Stage IVC Any T, any N M1<br />

Medullary carcinoma<br />

Anaplastic carcinoma All anaplastic carcinomas are only stage IV<br />

IVA T4a any N M0<br />

IVB T4b any N M0<br />

IVC Any T, any N M1<br />

3.6.6. Diagnostics and treatment<br />

Physical examination detects nodules and enlargement <strong>of</strong> the thyroid gland. Precise<br />

assessment <strong>of</strong> regional cervical, supraclavicular lymph nodes is necessary. Before a patient is<br />

qualified to surgery, the vocal cords should be examined.<br />

When differentiating nodular lesions in the thyroid gland, the level <strong>of</strong> thyroid hormones<br />

should be measured. If no hormonal disturbances are found, this may be indicative <strong>of</strong> thyroid<br />

cancer. Patients with hypothyroidisms or hyperthyroidisms should be referred to an<br />

endocrinologist. Other recommended tests are tests <strong>of</strong> antibodies directed against thyroid<br />

antigens. Ultrasound-guided or unguided fine-needle biopsy <strong>of</strong> the lesion is necessary. No<br />

other studies are required for routine procedure. Ultrasound scanning <strong>of</strong> the thyroid gland is<br />

particularly helpful if same-time fine-needle aspiration biopsy is planned, or if enlarged lymph<br />

nodes or other additional nodules are palpable in physical examination. <strong>CT</strong> or MRI <strong>of</strong> the<br />

thyroid gland is helpful when other techniques are insufficient to determine the boundaries <strong>of</strong><br />

cancer infiltration, or when haemoptysis occurs. Measurements <strong>of</strong> calcitonine level in blood<br />

serum are recommended in medullary thyroid cancer. A chest radiogram and spirometry<br />

help evaluate the patient’s general condition and other accompanying diseases. Isotopic<br />

studies are diagnostically irrelevant in thyroid cancer, however they may have impact on<br />

preoperative and postoperative staging, just as ultrasonography or <strong>CT</strong> <strong>of</strong> the abdominal<br />

cavity have. With differentiated cancers, open biopsy <strong>of</strong> the thyroid gland is not used usually.<br />

However, it is sometimes necessary in the diagnostics <strong>of</strong> lymphomas [3, 4].<br />

The only radical treatment method for thyroid cancer is surgery. The extent <strong>of</strong> surgery<br />

depends on the stage and histological features <strong>of</strong> the tumor. The most <strong>of</strong>ten used<br />

supplementary treatment <strong>of</strong> highly differentiated tumors is iodine 131 radiotherapy, which is<br />

69


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

helpful in procedures against not only primary tumor but also distant metastasis. Sometimes<br />

external radiation <strong>of</strong> 5 000 Gy in fractions is used. Chemotherapy is not a routine procedure<br />

for thyroid cancer, but is used with recurrent disease, after partial tumor resection or lack <strong>of</strong><br />

response to the therapeutic methods described above. Doxorubicin or cisplatin are used, but<br />

partial responses do not exceed 20% for papillary cancer. Chemotherapy should be<br />

administered only in symptomatic patients with disease progression.<br />

70<br />

When response to treatment is positive, the average survival period may be expected to<br />

grow from 3–5 to 15–20 months [3, 4].<br />

3.6.7. Prognosis<br />

Prognosis in thyroid cancer is usually good, which is proved by a low ratio <strong>of</strong> the mortality<br />

to incidence rate (0.25), one <strong>of</strong> the reasons being good access <strong>of</strong> tumor location procedures<br />

[6]. Thyroid cancer is responsible for a small proportion <strong>of</strong> deaths compared to other tumors<br />

(0.5% <strong>of</strong> all tumor-related deaths) [1]. Based on 1985–1991 data, the 5-year relative survivals<br />

for papillary cancer range from 100% for stage I to 45.3% for stage IV. For follicular cancer,<br />

survivals are similar, and ranged depending on the stage from 100% to 47.1%. The value for<br />

medullary cancer is 100%–24.3%; and for anaplastic cancer it is only 9.1% [5] 8 .<br />

3.7. Head and neck cancer<br />

3.7.1. Classification<br />

Classifications used in oncology include the following under head and neck cancer: lip<br />

cancer, oral cavity cancer, pharyngeal cancer, laryngeal cancer, paranasal sinuses cancer<br />

and salivary glands cancer. The most frequently diagnosed cancer in this location is<br />

squamous cell laryngeal cancer, less frequently squamous cell tonsil and pharynx cancer.<br />

3.7.2. Epidemiology<br />

Every year, 500,000 people develop head and neck cancer [1]. The incidence <strong>of</strong> head<br />

and neck cancer is significantly higher in men than in women, with relatively small regional<br />

differences in incidence and survival [2, 3]. Based on 2002 data, the incidence <strong>of</strong> oral cavity<br />

8 1. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin 2005; 55 (2): pp 74–108.<br />

2. The Globocan 2002 database: http://www-dep.iarc.fr<br />

3. Pawlicki M et all. Leczenie nowotworów. Bielsko Biała, Alfa Medica Press, 1996.<br />

4. British Thyroid Association. Royal College <strong>of</strong> Physicians. Guidelines for the management <strong>of</strong> thyroid cancer in<br />

adults. http://www.rcplondon.ac.uk/pubs/books/thyroidcancer/thyroid_guidelines.pdf<br />

5. AJCC cancer staging manual, 6th edition. http://www.cancerstaging.org/education/tnmschema/thyroid.ppt<br />

6. Coleman MO et al. Eurocare-3 summary: cancer survival in Europe at the end <strong>of</strong> 20th century. Ann Oncol 2003;<br />

1: pp 128–149.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

cancers in Poland in men and women is 7.3 and 1.4 per 100,000 respectively; the incidence<br />

<strong>of</strong> nasopharyngeal cancer is 0.6 and 0.2 per 100,000 respectively; and the incidence <strong>of</strong> other<br />

pharyngeal cancers is 6 and 0.7 per 100,000 respectively [4].<br />

3.7.3. Risk factors<br />

Risk factors <strong>of</strong> laryngeal cancer are similar to those <strong>of</strong> lung cancer. In 85% <strong>of</strong> patients,<br />

smoking and alcohol abuse are identified as part <strong>of</strong> case history [3, 5]. Oral cavity cancer<br />

may be caused by poor oral hygiene, ill-fitting dentures or tobacco and betel chewing [5].<br />

Infection with the Epstein-Barr virus also have a role in developing head and neck cancers.<br />

Patients with a history <strong>of</strong> low-dose radiation have a predisposition to thyroid cancer and<br />

salivary gland cancer [5].<br />

3.7.4. Symptoms<br />

Precise clinical evaluation is a precondition for early diagnosis. Hard-healing wounds and<br />

ulcerations, dyskeratosis, leucoplakias (white stains on mucous membranes), and papillary or<br />

inflammatory changes should be monitored. These can be the first symptoms <strong>of</strong> tumor. Other<br />

symptoms include obstruction <strong>of</strong> the nasal ducts, pains <strong>of</strong> sinuses, hoarseness [1].<br />

3.7.5. Histopathological picture<br />

Squamous cell carcinomas represent the majority <strong>of</strong> tumors in this location. The small<br />

proportion <strong>of</strong> other types include lymphoepithelioma, which arises from lymphatic epithelium,<br />

and transitional cell carcinoma, which arises from transitional cells [1].<br />

3.7.6. Staging<br />

For many years, head and neck tumors are confined to head and neck. Local extensions<br />

to tissues lead to the involvement <strong>of</strong> regional lymph nodes. Metastasis via lymphatic vessels<br />

occur late, and dissemination via the circulatory system occurs mainly with big tumors or in<br />

immunocompromised patients [5]. A broadly used classification is the TNM staging system <strong>of</strong><br />

the American Joint Committee on Cancer. Stage T (T1–T4) is related to the location and size<br />

<strong>of</strong> a primary tumor; stage N (N0–N1) reflects the number and size <strong>of</strong> cervical lymph nodes<br />

involved, and stage M (M0–M1) is determined by distant metastasis. The TNM staging system is<br />

diversified and its criterion is tumor location [5, 6]. Stage I corresponds to a primary tumor<br />

below 2 cm in diameter, confined to one anatomical zone, with no metastasis to lymph<br />

nodes and no distant metastasis (T1 N0 M0). Stage II covers tumors below 4 cm in diameter or<br />

in two locations in one anatomical region (for example larynx) without regional or distant<br />

metastasis (T2 N0 M0). Stage III covers tumors over 4 cm in diameter or involving three<br />

71


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

adjacent areas in the head and neck and/or isolated regional metastasis below 3 cm in<br />

diameter (T3 N0 M0 or T1–T3 N1 M0). Stage IV is a massive tumor that invades bones,<br />

cartilages and/or spreads from the primary location to other region (for example from the<br />

oral cavity to the pharynx); metastasis to cervical lymph nodes greater than 3 cm, with<br />

numerous same-side, both-sides or opposite-side lymph nodes. Distant metastasis (T1–4 N1–3<br />

M0–1) is found. Clinical staging is supplemented with diagnostic imaging [5].<br />

72<br />

3.7.7. Diagnostics and treatment<br />

Enlarged cervical lymph nodes or neck tumor can be detected by physical examination.<br />

A lesions is definitely confirmed by a histopathological study <strong>of</strong> samples taken from it.<br />

Multifocal growth <strong>of</strong> a tumor, which is frequently observed, and superficial sampling may<br />

hinder histopathological verification. In about 10% <strong>of</strong> patients, initial diagnosis is based on a<br />

histopathological test <strong>of</strong> specimens from enlarged lymph nodes. Treatment <strong>of</strong> head and<br />

neck tumors requires a multidisciplinary approach and full cooperation between a<br />

laryngologist, surgeon, radiotherapist and chemotherapist. Response to treatment,<br />

chemosensitivity and radiosensitivity vary in this group [1]. The choice <strong>of</strong> treatment depends<br />

on the stage, location and microscopic texture <strong>of</strong> a primary tumor. Surgical excision is the<br />

first-choice therapy for lesions located in the lip or oral cavity, or metastasis to cervical lymph<br />

nodes. However, radiation is the basic procedure for pharyngeal and laryngeal tumors [1]. In<br />

many stage I tumors, response to surgical treatment and radiotherapy is similar regardless <strong>of</strong><br />

location. Radiation <strong>of</strong> primary tumor and cervical lymph nodes is used. Tumors > 2 cm in<br />

diameter and with extensions to regional lymph nodes require surgery. If metastasis to<br />

regional lymph nodes is detected, the region is radiated postoperatively. Another surgery is<br />

possible with recurrent tumors. With advanced tumors (most <strong>of</strong> stage II tumors and all stage III<br />

and IV tumor), combined therapy is implemented (surgery and radiotherapy). Surgery is more<br />

effective in monitoring large primary head and neck tumors that radiotherapy or<br />

chemotherapy, but radiotherapy is a better way to monitor peripheral tumors<br />

and micrometastasis inaccessible by palpation. Radiotherapy can be used before and after<br />

surgery but the latter method is preferred. It is not clear whether adjuvant chemotherapy (in<br />

combination with surgery or radiotherapy) increases the number <strong>of</strong> recoveries but it may<br />

prolong the period <strong>of</strong> no symptoms <strong>of</strong> disease. Drugs used for this purpose include cisplatin,<br />

bleomycin, fluorouracil, metotrexat are used [5].<br />

Whether tumor resection and response to treatment are complete is assessed by<br />

diagnostic imaging (<strong>PET</strong>, <strong>CT</strong> or MRI <strong>of</strong> the neck) [5].


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

3.7.8. Prognosis<br />

Unfortunately, despite a relatively good accessibility and self-assessment opportunities, the<br />

majority <strong>of</strong> patients consult a doctor as late as stage III or IV, and roughly 50% receive<br />

improper therapy for many months before they are diagnosed correctly [1].<br />

The survival rate for head and neck tumors is largely contingent on their location. Lip<br />

cancer has the best prognosis (94% <strong>of</strong> 5-year survivals) [6]. Depending on tumor location, 5-<br />

year survivals for head and neck tumors in Europe range from 62% for laryngeal cancer, to<br />

61% for salivary glands cancer, 43% for nasopharyngeal cancer, 45% for oral cavity cancer,<br />

40–50% for nasal cavity cancer, nearly 39% for tongue cancer, 32% for oropharyngeal<br />

cancer, 25% for laryngopharyngeal cancer, to only 10% for esophageal cancer [2, 7]. The<br />

survival rate is higher in women than in men, especially for oral cavity and pharyngeal cancer<br />

[2, 8].<br />

If tumor is managed properly, the 5-year survival rate for stage I disease is 90%. For more<br />

advanced stages, the expected 5-year survival rate is 75% for stage II; 45–75% for stage III;<br />

and < 35% for stage IV. The 5-year survival rate calculated for stage II and III total is 65%.<br />

When metastasis to lymph nodes is found, prognosis is much poorer: 5-year survival does not<br />

exceed 30%. Older patients survive longer and have longer asymptomatic periods after<br />

treatment than younger patients [5] 9 .<br />

3.8. Pancreatic cancer<br />

3.8.1. Epidemiology<br />

Every year, 227,000 people die <strong>of</strong> pancreatic cancer. This is the eighth leading cause <strong>of</strong><br />

death <strong>of</strong> all the tumors, and the thirteen with respect to incidence. Its high mortality relative<br />

to incidence proves its poor prognosis: the mortality to incidence ratio is 98%. The frequency<br />

9 1. Pawlicki M. Leczenie nowotworów. Bielsko-Biała, Alfa Medica Press, 1996.<br />

2. Sant M, Aareleid T, Berrino F, Bielska Lasota M, Carli PM, Faivre J, Grosclaude JP, Hédelin G, Matsuda T, Møller H,<br />

Möller T, Verdecchia A, Capocaccia R, Gatta G, Micheli A, Santaquilani M, Roazzi P, Lisi D and the EUROCARE<br />

Working Group. EUROCARE-3: survival <strong>of</strong> cancer patients diagnosed 1990–94 – results and commentary. Ann Oncol<br />

2003; 14: pp 61–118.<br />

3. Evidence-based Cancer Prevention: Strategies for NGOs - A UICC Handbook for Europe Evidence-based<br />

Cancer Prevention: Strategies for NGOs – A UICC Handbook for Europe. http://www.uicc.org<br />

4. Globocan 2002 database; http://www-dep.iarc.fr/<br />

5. The Merck manual <strong>of</strong> diagnosis and therapy. Merck and Co. 17 th edition; www.merck.com<br />

6. Snehal G, Patel and Jatin P. Shah. TNM Staging <strong>of</strong> Cancers <strong>of</strong> the Head and Neck: Striving for Uniformity Among<br />

Diversity. CA Cancer J Clin 2005; 55: pp 242–258.<br />

7. Coleman MP, Gatta G, Verdecchia A, Estève J, Sant M, Storm H, Allemani C, Ciccolallo L, Santaquilani M,<br />

Berrino F and the EUROCARE Working Group. EUROCARE-3 summary: cancer survival in Europe at the end <strong>of</strong> the<br />

20th century. Ann Oncol 2003; 14: pp 128–149.<br />

8. Berrino F, Gatta G. Variation in survival <strong>of</strong> patients with head and neck cancer in Europe by the site <strong>of</strong> origin <strong>of</strong><br />

the tumors. The EUROCARE Working Group. Eur J Cancer 1998; 34: pp 2154–2161.<br />

73


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

<strong>of</strong> pancreatic cancer in women and men is similar. Most cases are reported in developed<br />

countries (61%), where incidence and mortality in men are 7 and 9 per 100,000 <strong>of</strong> the<br />

population, in women – 4.5 and 6 per 100,000. It seems that lower incidence in developing<br />

countries is attributable to poorer diagnostic efficacy rather than to real differences. The<br />

highest incidence rates in developing countries is observed in Central and South America [1].<br />

In Poland, in 2002, the incidence <strong>of</strong> pancreatic cancer in men was 9.5 per 100,000, and<br />

morbidity was 8 per 100,000 (age-standardized values). In women, the rates were 6.1 per<br />

100,000 and 5.3 per 100,000 respectively [2].<br />

74<br />

3.8.2. Risk factors<br />

The etiology <strong>of</strong> the disease is unknown. Risk factors include age (the highest risk <strong>of</strong><br />

morbidity is in the 6th and 7th decades <strong>of</strong> life), sex (slightly more frequent in men), race<br />

(Polynesians, Hawaiians, Jews, American Indians), tobacco smoking (twice as frequent in<br />

people smoking at least 2 packs daily), diet (none <strong>of</strong> the factors analyzed was definitely<br />

identified as a risk factor: alcohol, c<strong>of</strong>fee, diet rich in fats), prior pancreas diseases, low<br />

socioeconomic status, occupational exposure (chemical, coke, metal, gas industry),<br />

participation <strong>of</strong> genetic factors were not found [3].<br />

3.8.3. Symptoms<br />

Early symptoms are occasional and uncharacteristic, which prevents early diagnosis. Pain<br />

in the mesogastrium may occur. Late symptoms include jaundice caused by tumor pressure<br />

on biliary ducts; or enlarged gall bladder detectable in physical examination (Courvoisier’s<br />

symptom), loss <strong>of</strong> body weight, or pain in the lumbar region. A frequent manifestation <strong>of</strong> the<br />

tumor is recurrent athero-thrombotic disease [4].<br />

3.8.4. Histopathological picture<br />

Histopathological diagnosis should be made in compliance with WHO criteria based on<br />

an examination <strong>of</strong> material from biopsy or fine-needle aspiration [5]. The most common type<br />

<strong>of</strong> tumors <strong>of</strong> the exocrine part <strong>of</strong> pancreas is ductal carcinoma (90%), which arises from the<br />

epithelium <strong>of</strong> the pancreatic ducts. Much less frequent is adenoid cystic cancer<br />

(cystadenocarcinoma) and acinar cell carcinoma. Relatively rare are oncocytic cancer,<br />

clear cell cancer, signet ring cell cancer, mucosal and squamous cell carcinoma. Extremely<br />

rare in the pancreas are tumors <strong>of</strong> other histopathological textures such as sarcomas,<br />

lymphomas or pancreatoblastoma, which is most frequent in children. About 5% <strong>of</strong> all<br />

cancerous hyperplasia <strong>of</strong> pancreas arises from cells <strong>of</strong> the exocrine system: they are so-<br />

called insulomas, benign or malignant, which overproduce pancreatic enzymes [6].


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

[7].<br />

3.8.5. Staging<br />

Table 12 presents the classification and stages <strong>of</strong> pancreatic cancer according to AJCC<br />

Table 12.<br />

AJCC staging system for exocrine pancreatic cancer<br />

Primary tumor<br />

Tx Primary tumor cannot be assessed<br />

T0 No evidence <strong>of</strong> primary tumor<br />

Tis Carcinoma in situ<br />

T1 Tumor limited to the pancreas, ≤2 cm in greatest dimension<br />

T2 Tumor limited to the pancreas, >2 cm in greatest dimension<br />

T3 Tumor invades the duodenum, common biliary duct, peripancreatic tissues<br />

T4 Tumor invades the stomach, spleen, large intestine, adjacent vessels<br />

Regional lymph nodes<br />

Nx Regional lymph nodes cannot be assessed<br />

N0 No regional lymph node metastasis<br />

N2 Regional lymph node metastasis<br />

Distant metastasis<br />

Mx Distant metastasis cannot be assessed<br />

M0 No distant metastasis<br />

M1 Distant metastasis<br />

Stage 0 Tis N0 M0<br />

Stage I T1–T2 N0 M0<br />

Stage II T3 N0 M0<br />

Stage III T1–T3 N1 M0<br />

Stage IVA T4, any N, M0<br />

Stage IVB Any T, any N, M1<br />

Stage groupings for pancreatic cancer<br />

3.8.6. Diagnostics and treatment<br />

Before a decision on therapy is made, apart from a precise case history and physical<br />

examination, peripheral blood morphology, liver enzyme levels and the level <strong>of</strong> glycaemia in<br />

blood serum should be tested. Other a radiogram <strong>of</strong> the chest, and abdomen imaging by<br />

ultrasonography and <strong>CT</strong> or magnetic resonance are required. Other diagnostic methods<br />

include endoscopic retrograde cholangiopancreatography (ERCP), endoscopic<br />

ultrasonography (EUS), laparoscopy with biopsy and laparoscopic ultrasonography [6].<br />

75


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

76<br />

Surgical treatment <strong>of</strong> pancreatic cancer is the only effective procedure. However, the 5-<br />

year survival rate after surgery is 10–20%, Pancreas surgery is done by:<br />

• radical resection (R0 – complete resection <strong>of</strong> tumor, no micro- or macroscopic<br />

involvement),<br />

• palliative resection (R1 – resection <strong>of</strong> tumor with residual microscopic involvement, R2 –<br />

resection <strong>of</strong> tumor with residual macroscopic involvement),<br />

• palliative procedures (bypass) [6].<br />

Pre- and postoperative chemotherapy, with or without radiotherapy, remain<br />

controversial.<br />

For disease with distant metastasis, treatment focuses in the main on symptoms. In some<br />

patients, radiotherapy may help to relieve pain. Patients may require the stenting <strong>of</strong> the<br />

biliary ducts due to jaundice that arises from tumor’s pressure on the biliary ducts; or it is<br />

necessary to unblock the ducts [4, 5]. An initial evaluation <strong>of</strong> response to therapy may be<br />

carried out by follow-up imaging. However, imaging is not required in properly managed<br />

patients. Response to treatment should be evaluated mostly based on clinical symptoms [5].<br />

Figure 4 presents an algorithm <strong>of</strong> a therapeutic procedure for pancreatic cancer [6].


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Figure 4.<br />

Algorithm <strong>of</strong> therapeutic procedure for pancreatic cancer; source: The Polish Oncology Union, Zalecenia<br />

postępowania diagnostyczno-terapeutycznego w nowotworach złośliwych u dorosłych, edited by M. Krzakowski [6]<br />

3.8.7. Prognosis<br />

Pancreatic cancer is among the tumors with the poorest prognosis. The average European<br />

5-year survival rate for pancreatic cancer is below 4% [8]. Prognosis depends on disease<br />

77


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

stage, but as epidemiological studies show, the patient’s age is a stronger factor [5]. Table 13<br />

presents the proportion <strong>of</strong> patients with 5 years’ survival by disease stage [5] 10 .<br />

Table 13.<br />

5-year survivals in pancreatic cancer by disease stage<br />

78<br />

Stage 0 -<br />

Stage I 5–35%<br />

Stage II 2–15%<br />

Stage III 2–15%<br />

Grade IVA 1–5%<br />

Grade IVB < 1%<br />

3.9. Gastrointestinal stromal tumor<br />

3.9.1. Epidemiology<br />

Sarcomas <strong>of</strong> s<strong>of</strong>t tissues are a group <strong>of</strong> heterogeneous tumors <strong>of</strong> mesenchymal origin. In<br />

Poland, a few hundred cases <strong>of</strong> sarcomas are diagnosed annually, most frequently in<br />

patients aged 60 plus [1]. One type is gastrointestinal stromal tumor (GIST). It is believed that<br />

GISTs represent about 0.1 to 0.3% <strong>of</strong> all tumors <strong>of</strong> the stomach and intestines [5].<br />

Epidemiological data on gastrointestinal stromal sarcomas published to date are not fully<br />

reliable [2]. The exact proportion <strong>of</strong> benign and malignant GIST types (sarcomas generating a<br />

potential risk <strong>of</strong> recurrence and/or inoperable dissemination) is not known either.<br />

Retrospective studies carried out in Sweden show that the mortality rate for GIST (benign and<br />

malignant) is 16 cases per year per million. For Poland, the incidence <strong>of</strong> about 600 new cases<br />

yearly is suggested. In the United States, the number <strong>of</strong> metastatic or inoperable GISTs is<br />

estimated at over 1000 new cases annually, which gives 3–4 cases per 1 million inhabitants.<br />

Other estimates imply that in Poland about 120–195 new cases can be expected every year.<br />

It seems that the incidence <strong>of</strong> this tumor type is similar in men and women [2].<br />

10 1. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin 2005; 55: pp 74–108.<br />

2. Globocan 2002 database. http://www-dep.iarc.fr/<br />

3. Pawlicki M et all, Leczenie nowotworów. Bielsko-Biała, Alfa Medica Press, 1996.<br />

4. Strum A, Largiader F, Wicki O. Kompendium onkologii. Warszawa, PZWL, 1995.<br />

5. ESMO Minimum Clinical Recommendations for diagnosis, treatment and follow-up <strong>of</strong> pancreatic cancer. Annals<br />

<strong>of</strong> Oncology 2005; 16: pp 124–125.<br />

6. Krzakowski M. et all, Zalecenia postępowania diagnostyczno-terapeutycznego w nowotworach złośliwych u<br />

dorosłych. http://www.puo.pl/ksiazka.php<br />

7. AJCC cancer staging manual, sixth edition.<br />

http://www.cancerstaging.org/education/tnmschema/exocrinepancreas.ppt<br />

8. Coleman MO et al. Eurocare-3 summary: cancer survival in Europe at the end <strong>of</strong> 20th century. Annals <strong>of</strong><br />

Oncology 2003; 1: pp 128–149.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

3.9.2. Symptoms<br />

Complaints are usually non-characteristic and may include abdominal pains, symptoms<br />

resembling occlusion <strong>of</strong>, or chronic bleeding from, the gastrointestinal tract; tumor palpable<br />

in the abdomen; or occasional acute abdomen symptoms. Small lesions may be long<br />

asymptomatic. The results we obtained as well as data from other authors suggest that the<br />

asymptomatic period preceding dissemination may be long, with the median exceeding 2<br />

years [2].<br />

3.9.3. Histopathological picture<br />

GIST-type sarcomas have been diagnosed more commonly for the past 4–5 years. Until<br />

recently, these tumors had been known and diagnosed under various names. Now the term<br />

“benign” is avoided with reference to GIST, and replaced with the descriptor “low-<br />

aggressiveness” because in some cases, generalization disease traits were found over longer<br />

periods <strong>of</strong> observation. GISTs derive most probably from precursors <strong>of</strong> pace-making<br />

neurological interstitial cells <strong>of</strong> Cajal. The most important prognostic factors are the size <strong>of</strong><br />

primary cancer focus, and the number <strong>of</strong> mitoses observed in 50 fields <strong>of</strong> vision in large<br />

magnification [2].<br />

3.9.4. Staging<br />

At present there is no widely acceptable staging system. A suggested classification takes<br />

into account aspects such as growth size, involvement <strong>of</strong> regional lymph nodes, distant<br />

metastasis and histopathological picture [5].<br />

3.9.5. Diagnostics and treatment<br />

Because GIST is a disease recently isolated based on its molecular and histological<br />

features, no standard diagnostic or therapeutic procedure exist.<br />

The most efficacious method in GIST treatment is radical surgery. The majority <strong>of</strong> patients<br />

(92%) are first operated without prior histopathological diagnosis <strong>of</strong> GIST (only in 8% have<br />

histological suspicions <strong>of</strong> GIST).<br />

In nearly 74% <strong>of</strong> patients, the first therapeutic surgery is possible by resecting stomach,<br />

small intestine or large intestine, or by excising a intraperitoneal/retroperitoneal tumor with<br />

macroscopic margin. Surgical treatment is highly efficacious mainly for low and intermediate<br />

degrees <strong>of</strong> aggressiveness. With stomach GIST, local excision <strong>of</strong> the tumor along with a<br />

section <strong>of</strong> the stomach wall is most frequent. Complete or incomplete resection is rarely<br />

performed. It seems, that the extent <strong>of</strong> stomach resection is neutral for recurrence.<br />

79


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Subsequent operations <strong>of</strong> recurrent tumors do not lead to recovery. In other locations,<br />

segmental resection <strong>of</strong> the small bowel or hemicolectomy are used.<br />

80<br />

Current scientific knowledge does not support the use <strong>of</strong> supplementary treatment <strong>of</strong> GIST.<br />

Inoperable or disseminated GIST is resistant to conventional chemotherapy and<br />

radiotherapy. Clinical researches focuses on treatment based on imatinib, a tyrosine kinase<br />

inhibitor.<br />

Recurrences after treatment are most frequently found in the abdominal cavity. Usually<br />

liver metastasis (54%), isolated metastasis (22%), or metastasis accompanied by<br />

intraperitoneal dissemination (32%) occur. Intraperitoneal dissemination without liver<br />

metastasis is observed in about 31% <strong>of</strong> patients. In a proportion <strong>of</strong> patients (15%), inoperable<br />

local recurrence is observed, which is a good pro<strong>of</strong> <strong>of</strong> the surgical potential <strong>of</strong> locally radical<br />

excision <strong>of</strong> the tumor [2, 3, 4].<br />

3.9.6. Prognosis<br />

Prognostic factors include the size <strong>of</strong> primary focus <strong>of</strong> the tumor and the number <strong>of</strong><br />

mitoses observed in 50 fields <strong>of</strong> vision under high magnification. About 20–30% <strong>of</strong> GIST tumors<br />

are malignant. Gastric cancer up to 5 cm in tumor size, and small bowel tumors up to 2 cm in<br />

size are benign as a rule. Following radical excision <strong>of</strong> the tumor, 35–65% 5-year survival is<br />

observed. Prognosis for patients with inoperable lesions or with metastasis is poor (median<br />

survival under 12 months or 8% 5-year survivals). The worst results are reported for patients with<br />

intraperitoneal dissemination caused by gastrointestinal tract obstruction [2, 4, 5] 11 .<br />

11 1. Pawlicki M et all, Leczenie nowotworów. Bielsko Biała, Alfa Medica Press, 1996.<br />

2. Ruka W, Rutkowski P, Nowecki Z, Nasierowska-Guttmejer A, Grzesiakowska U. Mięsaki podścieliskowe przewodu<br />

pokarmowego (GIST) – zalecenia postępowania diagnostyczno-terapeutycznego.<br />

http://www.coi.waw.pl/miesaki/index.htm<br />

3. Blay J-Y, Bonvalot S, Casali P, Choi H, Debiec-Richter M, Dei Tos AP, Emile J-F, Gronchi A, Hogendoorn PCW,<br />

Joensuu H, Le Cesne A, Mac Clure J, Maurel J , Nupponen N, Ray-Coquard I, Reichardt P, Sciot R, Stroobants S,<br />

van Glabbeke M, van Oosterom A, Demetri GD. Consensus meeting for the management <strong>of</strong> gastrointestinal<br />

stromal tumors. Report <strong>of</strong> the GIST Consensus Conference <strong>of</strong> 20–21 March 2004, under the auspices <strong>of</strong> ESMO. Ann<br />

Oncol 2005; 16: pp 566–578.<br />

4. DeMatteo RP, Lewis JJ, Leung D et all, Two hundred gastrointestinal stromal tumors. Recurrence patterns and<br />

prognostic factors for survival. Ann Surg 2000; 231: pp 51–58.<br />

5. Crosby JA, Catton CN, Davis A, Couture J, O’Sullivan B, Kandel R, Swallow CJ. Malignant Gastrointestinal Stromal<br />

Tumors <strong>of</strong> the Small Intestine: A Review <strong>of</strong> 50 Cases From a Prospective Database. Ann Surg Oncol 2001; 8 (1): pp<br />

50–59.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

3.10. Colon cancer<br />

3.10.1. Epidemiology<br />

In 2002, about million new cases <strong>of</strong> colon cancer were reported globally (9.4% <strong>of</strong> all<br />

tumors). No significant differences in incidence between men and woman are found for this<br />

cancer type. As regards incidence, it is the fourth leading tumor in men and the third leading<br />

tumor in women globally [4]. In 2002 in Poland, the standardized incidence rates for<br />

colorectal cancer were 31.9 per 100,000 men and 23.5 per 100,000 women. Mortality was<br />

18.2 per 100,000 and in women it was 11.4 per 100,000 [2]. The incidence <strong>of</strong> this tumor type is<br />

still growing [3], but mortality in Europe has decreased over the last decade [5].<br />

Approximately 25-time differences in the incidence <strong>of</strong> this tumor are observed in the world.<br />

The highest incidence rates are reported in North America, Australia, New Zealand, and<br />

Western Europe, and the lowest in Africa, Asia, South America. These differences may be<br />

attributable to different exposure to environmental impacts.<br />

In fast developing countries, the incidence <strong>of</strong> colorectal cancer is growing but in highly<br />

developed countries these rates tend to stabilize or even fall (North America) [4].<br />

Colorectal tumors are rare for people younger than 40. Above this age, risk increases [3].<br />

3.10.2. Risk factors<br />

It is believed, that genetic and environmental factors play an important role in developing<br />

colon cancer.<br />

Precancerous conditions that predispose to cancer include benign adenomas <strong>of</strong> the<br />

large intestine, polipomatosis syndromes and inflammatory bowel diseases. The majority <strong>of</strong><br />

occasional cancers arise from acquired mutations <strong>of</strong> suppressor genes such as p53, APC [3].<br />

It is believed that the risk factors <strong>of</strong> colon cancer may include improper diet (high intake <strong>of</strong><br />

meat, meals rich in animal fats). Epidemiological studies showed that no physical exercise,<br />

obesity, and central distribution <strong>of</strong> the fatty tissue are risk factors <strong>of</strong> colorectal cancer too [4].<br />

It seems that diet rich in fats and animal meat but calcium and selenium-deficient may<br />

adversely affect bacterial flora, trigger the synthesis <strong>of</strong> carcinogenic compounds, and<br />

increase the time <strong>of</strong> exposition <strong>of</strong> the bowel mucosa to these substances by slowing down<br />

their passage through the large intestine [3].<br />

A major portion <strong>of</strong> colon cancers arise from adenomas.<br />

81


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

82<br />

3.10.3. Symptoms<br />

The disease may be asymptomatic, or may cause symptoms initially ignored by the<br />

patient. The most frequent are latent bleeding and abdominal pain. Alterations <strong>of</strong> the normal<br />

defecation rhythm, diarrheas or constipations may occur. Hemorrhage, bowel ruptures or<br />

ileus are rare [3].<br />

3.10.4. Histopathological picture<br />

Almost all colorectal cancers arise from adenomatous polyps. In about 10% <strong>of</strong> cases,<br />

cancer cells are <strong>of</strong> endocrine type [11]. The most frequent type is adenocarcinoma, which<br />

represents 85% <strong>of</strong> all colon cancers. Mucous cancer represents about 10%. Other types are<br />

mucocellular carcinoma, squamous cell carcinoma, mixed and non-differentiated<br />

carcinomas [12].<br />

3.10.5. Staging<br />

Table 14 presents the TNM staging system for colorectal cancer [7].<br />

Table 14.<br />

TNM staging system and tumor classification for colon cancer<br />

Primary tumor<br />

Tx Primary tumor cannot be assessed<br />

T0 No evidence <strong>of</strong> primary tumor<br />

Tis Cancer in situ<br />

T1 Tumor invades submucosa<br />

T2 Tumor invades muscularis propria<br />

T3 Tumor invades through the muscularis propria into the subserosa<br />

T4 Tumor invades through the visceral peritoneum or invades adjacent structures<br />

Regional lymph nodes<br />

Nx Regional nodes cannot be assessed<br />

N0 No regional lymph node metastasis<br />

N1 Metastasis in 1–3 lymph nodes<br />

N2 Metastasis in 4 or more lymph nodes<br />

N3 Metastasis in mesenteric lymph nodes<br />

Distant metastasis<br />

M0 No distant metastasis<br />

M1 Distant metastasis<br />

TNM stage groupings<br />

Stage 0 Tis N0 M0<br />

Stage I T1 N0 M0<br />

T2 N0 M0<br />

Grade II T3 N0 M0


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

T4 N0 M0<br />

Stage III Any T N1 M0<br />

Any T N2, N3 M0<br />

Stage IV Any T any N M1<br />

There is a range <strong>of</strong> clinical pathological classifications <strong>of</strong> colon cancer. One <strong>of</strong> them a<br />

classification <strong>of</strong> colorectal cancers proposed by Astler-Coller [7]. This classification is<br />

presented in Table 15.<br />

Table 15.<br />

Astler-Coller classification <strong>of</strong> colorectal cancers<br />

Stage Histological features <strong>of</strong> tumor<br />

A Tumor limited to mucosa<br />

B1 Growth invades through muscularis propria, lymph nodes not involved<br />

B2 Tumor grows beyond muscularis propria, lymph nodes not involved<br />

C1 Growth invades through muscularis propria, lymph nodes not involved<br />

C2 Growth invades through muscularis propria, lymph nodes involved<br />

D Distant metastasis<br />

3.10.6. Diagnostics and treatment<br />

3.10.6.1 Screening examinations<br />

The key aim <strong>of</strong> screening examinations is to reduce the mortality rate <strong>of</strong> colon cancer. For<br />

this purpose fecal occult blood tests, sigmoidoscopy or full colonoscopy are performed [3].<br />

3.10.6.2 Physical examination<br />

Per rectum examination is essential. In some cases, a tumor is palpable during finger<br />

examination <strong>of</strong> the final section <strong>of</strong> the rectum.<br />

3.10.6.3 Laboratory tests<br />

Laboratory tests can detect iron deficiency anemia. Cancer diagnosis may be suggested<br />

by a high level <strong>of</strong> carcinoembryonic antigen (CEA) in blood serum. Fecal occult blood tests<br />

are positive.<br />

3.10.6.4 Endoscopy<br />

Colonoscopy is the most important diagnostic study. It helps evaluate the mucous<br />

membrane <strong>of</strong> the large intestine microscopically, and to sample lesions for histopathological<br />

examination. Recently, endosonography (fused ultrasonography and colonoscopy), has<br />

been used. It helps to estimate the depth <strong>of</strong> carcinogenic infiltration into the bronchial wall<br />

and to assess local lymph nodes and node metastasis in a non-invasive way.<br />

83


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

84<br />

3.10.6.5 Diagnostic imaging<br />

Ultrasound scanning, or the more precise <strong>CT</strong>, <strong>of</strong> the abdominal cavity provides grounds to<br />

suspect distant metastasis, for example to the liver.<br />

<strong>PET</strong> is used to evaluate disease recurrence when the patient has completed therapy.<br />

However, it is believed to be <strong>of</strong> little value for T-staging [3].<br />

3.10.6.6 Surgery<br />

Surgery is the basic therapy for colorectal cancer. The aim <strong>of</strong> the surgery is total resection<br />

<strong>of</strong> the tumor with regional lymph nodes. A traditional method is laparotomy; lately<br />

laparoscopic methods are used with similar distant results.<br />

3.10.6.7 Supplementary treatment<br />

Systemic chemotherapy with 5-fluorouracyl and levamisole, administered for one year<br />

following a surgery <strong>of</strong> stage C1, C2 cancer reduces the risk <strong>of</strong> local recurrences by 41% and<br />

<strong>of</strong> deaths by 33% compared to surgery-only therapy. A combination <strong>of</strong> 5-fluorouracyl<br />

with foline acid is also used for stage B2 [3]. Monoclonal antibodies against receptors for<br />

epithelial growth factor or endothelial growth factor are new drugs that can be used in<br />

combination with other chemotherapeutics. Second-line chemotherapy can be considered<br />

in patients with good general condition [6].<br />

3.10.6.8 Treatment <strong>of</strong> disseminated disease<br />

Inoperable tumors are treated with combined chemotherapy (5-fluorocytosine in<br />

combination with foline acid). Capecitabin or irinotecan with oxaliplatin are an alternative<br />

therapy. If intestinal passage is impaired due to tumor mass, its patency can be restored by<br />

surgery or using laser rays. Liver metastasis can be treated with transcutaneus alcohol<br />

injection or injection <strong>of</strong> cytostatic drugs to the hepatic artery [3]. Surgical excision <strong>of</strong> a single<br />

liver or lung metastasis should be considered [6].<br />

3.10.6.9 Evaluation <strong>of</strong> response to treatment<br />

Recommended are physical examination, measurement <strong>of</strong> CEA level, ultrasound<br />

scanning <strong>of</strong> the liver, and/or tomography <strong>of</strong> primary involvements 2–3 months after palliative<br />

therapy is completed.<br />

3.10.6.10 Follow-up<br />

There is no pro<strong>of</strong> that regular monitoring <strong>of</strong> the disease improves the efficacy <strong>of</strong> colon<br />

cancer management. Follow-up consultations in case <strong>of</strong> disease symptoms are<br />

recommended. Laboratory tests and imaging diagnostics should be used in patients with<br />

suspected disease progression if future palliative treatment is planned [6].


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

3.10.7. Prognosis<br />

Five-year survival rates vary depending on geographical location. In North America, the<br />

survivals are 65%, in Western Europe - 54%, but in Eastern Europe only 34%. A relatively good<br />

prognosis for this tumor means that the mortality rate is half the incidence rate [4].<br />

The findings <strong>of</strong> Eurocare-3 indicate that in the early 1990s, the 5-year survival rate for colon<br />

cancer patients in Poland was 26.3% in men and 28.7% in women. The five-year survival rate<br />

for colon cancer patients is about 50% lower in Poland compared to the survival rate for the<br />

whole Europe [9, 10].<br />

Prognosis in colon cancer depends on the tumor stage. The odds <strong>of</strong> 5-year survival for<br />

patients with stage A disease according to the Astler-Coller classification are 100%, for<br />

patients with stage B1 disease: 67%; for patients with stage B2 disease: 54%; for patients with<br />

stage C1 disease: 43%; and for patients with stage C2 disease: only 23% [11] 12 .<br />

12 1. Europe 1995, Estmates <strong>of</strong> cancer incidence and mortality in Europe in 1995.<br />

http://www.encr.com.fr/europe95.htm<br />

2. Globocan 2002: http://www-dep.iarc.fr/<br />

3. Szczeklik A et all, Choroby wewnętrzne. Kraków, Medycyna Praktyczna, 2005.<br />

4. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin 2005; 55 (2): pp 74–108.<br />

5. Europe’s cancer burden http://www.uicc.org/fileadmin/manual/5burden.pdf<br />

6. ESMO Minimum Clinical Recommendations for diagnosis, treatment and follow-up <strong>of</strong> advanced colorectal<br />

cancer. Ann Oncol 2005; 16: pp 18–19.<br />

7. Astler VB, Coller FA. The prognostic significance <strong>of</strong> direct extension <strong>of</strong> carcinoma <strong>of</strong> the colon and rectum. Ann<br />

Surg 1954; 139: p 846.<br />

8. AJCC cancer staging manual, sixth edition.<br />

http://www.cancerstaging.org/education/stagingmomentscolon06.ppt.<br />

9. Narodowy program zwalczania chorób nowotworowych. ZałoŜenia i cele operacyjne 2006–2015.<br />

http://www.mz.gov.pl/wwwfiles/ma_struktura/docs/zalozenia_ustawy_o_npzchn.pdf<br />

10. Coleman MO et al. Eurocare-3 summary: cancer survival in Europe at the end <strong>of</strong> 20th century. Ann Oncol 2003;<br />

1: pp 128–149.<br />

11. Coran R, Kumar V, Ramzi T. Robbins pathologic basis <strong>of</strong> disease. Elsevier/Saunders, 1999, wydanie 6.<br />

12. Pawlicki M et all, Leczenie nowotworów. Bielsko Biała, Alfa Medica Press, 1996.<br />

85


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

4. DESCRIPTION OF INTERVENTION<br />

86<br />

4.1. Description <strong>of</strong> method<br />

<strong>PET</strong> is a diagnostic method that produces images <strong>of</strong> metabolic processes at the molecular<br />

level. The <strong>PET</strong> technology has been developing since the early 1970s [1].<br />

<strong>CT</strong> is an accepted tool used in imaging diagnostics. Currently, multi-detector-row spiral<br />

tomography scanners prevail on the market.<br />

The first <strong>PET</strong>/<strong>CT</strong> fused scanning system was marketed in 2001 [1, 2]. <strong>PET</strong>/<strong>CT</strong> scanners are<br />

now manufactured by 4 companies: CPS Innovations, GE Medical Solutions, Philips Medical<br />

Systems and Siemens Medical Solutions [12]. In 2003, <strong>PET</strong>/<strong>CT</strong> systems represented 79% <strong>of</strong> total<br />

<strong>PET</strong> scanner sales. In 2004, the figure grew to 91% [13]. The first <strong>PET</strong>/<strong>CT</strong> unit in Poland was<br />

installed in Bydgoszcz in 2002.<br />

<strong>PET</strong> tomography uses the fact that metabolic processes have different dynamics in<br />

pathologically changed cells. These can be located using radionuclide-traced molecules,<br />

which are incorporated into metabolic cycles without no impact on physiological processes.<br />

Scintigraphic tracers are highly specific and a small amount <strong>of</strong> radionuclide is enough to<br />

produce diagnostic images.<br />

Intracellular processes can be examined by <strong>PET</strong> indirectly by locating <strong>positron</strong>-emitting<br />

tracers. A <strong>positron</strong> is a particle <strong>of</strong> the same mass as an electron but <strong>of</strong> the opposite charge.<br />

After leaving the where the deposited radionuclide disintegrates, it travels the distance <strong>of</strong><br />

about 4–5 mm, gradually loosing its kinetic energy in the surrounding tissue and then, when it<br />

collides with the nearest electron, annihilation occurs, resulting in the production <strong>of</strong> two<br />

gamma ray photons, each with the energy <strong>of</strong> 511 keV. The photons move in opposite<br />

directions and are registered by a system <strong>of</strong> detectors located in the ring [9] (Figure 5).<br />

Information on the number <strong>of</strong> gamma ray photons captured by the detectors is transmitted<br />

to the computer, which reconstructs, three-dimensionally, the distribution <strong>of</strong> radioactivity in<br />

the patient’s body.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Figure 5.<br />

Annihilation and detection <strong>of</strong> gamma rays<br />

The most frequently used in <strong>PET</strong> is radioisotope-traced glucose ( 18 F fluorodeoxyglucose, 18 F-<br />

FDG). The physical half-life <strong>of</strong> 18 F-FDG is 110 min. Other tracers are carbon 11 C (half-life <strong>of</strong> 20<br />

min), nitrogen 13 N (half-life <strong>of</strong> 10 min), oxygen 15 O (half-life <strong>of</strong> 122 s), rubidium 82 Rb (half-life <strong>of</strong><br />

75 s).<br />

4.2. Examination protocol, absorbed dose<br />

Before the examination, the patient should not eat for minimum 4 hours. After the<br />

radiopharmaceutical is given intravenously, the patient needs to lie down for an hour and<br />

drink a liter <strong>of</strong> water. Then <strong>CT</strong> and <strong>PET</strong> scanning is performed. Depending on indications,<br />

whole-body or selected region are scanned. After the image is reconstructed and processed<br />

the findings help not only determine the type <strong>of</strong> lesion, but also its exact localization [3]<br />

(Figure 6).<br />

87


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Figure 6.<br />

Dissemination <strong>of</strong> tumor: metastatic lymph nodes in the left groin<br />

a) <strong>CT</strong> mage; b) <strong>PET</strong>/<strong>CT</strong> image; c) <strong>PET</strong> image<br />

88<br />

For standard whole-body scanning with 18 F-FDG, the activity administered is about 370<br />

MBq (10 mCi), but the effective dose absorbed by the patient is about 10mSv [10], and only<br />

about 3 times higher than natural background radiation [2]. Obviously, this dose is received in<br />

a single test, while radiation from natural sources is taken in gradually over the whole year.<br />

Radiation effects are known to depend not only on the dose, but also on distribution in time.<br />

What is more, the dose absorbed in <strong>PET</strong> should be added to the dose absorbed from <strong>CT</strong><br />

scanning, which is about 5–10mSv, depending on the test protocol.<br />

4.3. Limitations <strong>of</strong> method<br />

The resolution <strong>of</strong> a <strong>PET</strong> scanner is currently 3–5 mm [1, 10], and is limited by the total<br />

distance traveled by the <strong>positron</strong> after annihilation, as well as by the fact, that the angular<br />

distribution is not exactly 180° (the acceptable difference is ±0.5°) [9, 10]. The efficiency and<br />

resolution <strong>of</strong> the method are also contingent on the distance between detectors and their<br />

sizes: the smaller the detectors and closer to each other and to the source <strong>of</strong> radiation, the<br />

higher resolution can be achieved. On the other hand, the smaller the scanner’s ring in<br />

diameter, the higher the probability <strong>of</strong> dispersion and coincidences [9]. Currently, scintillator<br />

detectors with Bi4Ge3O12 (BGO) crystals are the most frequently used. Contemporary <strong>PET</strong><br />

scanners are used for 3D visualization [10].<br />

Due to low spatial resolution multi-modal examination is used: a <strong>PET</strong> image is superimposed<br />

by rotation, shift and calibration on a high-resolution anatomical picture from <strong>CT</strong> or MRI [10].<br />

A <strong>CT</strong> scanner fused with a <strong>PET</strong> scanner should have at least 4 rows <strong>of</strong> detectors [13].


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Another problem is short half-life <strong>of</strong> the tracer, therefore miniature cyclotrons need to be<br />

installed close to the <strong>PET</strong>/<strong>CT</strong> scanner. The synthesis <strong>of</strong> the radiopharmaceutical must occur<br />

within a short time between manufacture and administration [9].<br />

4.4. Uses <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

<strong>PET</strong>/<strong>CT</strong> is particularly useful in oncology, neurology and cardiology.<br />

4.4.1. Oncology<br />

In cancer diagnostics, 18 F-FDG is used in the first place. Oncological indications for <strong>PET</strong><br />

scanning include:<br />

1. Assessment <strong>of</strong> nodular lesions in pulmonary parenchyma.<br />

For malignant lesions, <strong>PET</strong> sensitivity is 97% and <strong>PET</strong> specificity is 78%, but for benign lesions,<br />

the values are 89% (sensitivity) and 100% (specificity) [4, 5]. The standardized uptake value<br />

(SUV) is used to assess the type <strong>of</strong> lesion. The SUV tells us how intense the uptake <strong>of</strong> 18 F-FDG is<br />

[1, 6]. A nodule with the SUV below 1.5 has low odds <strong>of</strong> malignancy [1].<br />

2. Staging<br />

Currently, <strong>PET</strong> has an important role in the diagnostics <strong>of</strong> non-small cell lung cancer, colon<br />

and esophageal cancer, thyroid cancer, breast cancer, melanoma, lymphomas and<br />

malignant head and neck tumors [1, 2]. <strong>PET</strong> helps to determine boundaries <strong>of</strong> carcinogenic<br />

lesions in a primary growth, and to distinguish it from an oedema zone, atelectasis or<br />

inflammatory changes. In the case <strong>of</strong> lung tumors, <strong>PET</strong> helps to recognize the character <strong>of</strong><br />

the fluid in the pleura: it distinguishes benign exudations from fluid containing cancer cells<br />

with 92% precision [7]. Also, <strong>PET</strong> helps assess lymph nodes, especially those below 1 cm: for<br />

mediastinal lymph nodes its sensitivity is 80–90%, and specificity is 85–100% [8]. Note that the<br />

inflammatory condition <strong>of</strong> lymph nodes reduces <strong>PET</strong> sensitivity by giving false positive results<br />

[1, 6]. <strong>PET</strong> is also helpful in assessing distant metastasis. Often, these lesions are not revealed<br />

by any other imaging technologies. Detecting metastatic foci in brain causes difficulties,<br />

because the cortex has the highest physiological level <strong>of</strong> FDG accumulation in the human<br />

body [1].<br />

3. Follow-up examination to determine the risk <strong>of</strong> recurrence after therapy is completed.<br />

<strong>PET</strong> is more sensitive to recurrent carcinogenic processes, i.e. in esophageal cancer the<br />

sensitivity to local recurrences is 100%, but specificity is 57% due to false positive results<br />

produced by inflammatory changes [1]. Also, <strong>PET</strong> helps tell postoperative or post-<br />

radiotherapy cicatrix from recurrent tumor [2].<br />

4. Radiotherapy planning<br />

89


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

90<br />

<strong>PET</strong> helps detect additional disease foci that are not apparent in <strong>CT</strong>, which results in<br />

greater radiation areas. <strong>PET</strong> detects the most metabolically active regions within a tumor,<br />

based on which doses can be adjusted [1].<br />

4.4.2. Neurology<br />

In imaging <strong>of</strong> neurological diseases with <strong>PET</strong>, the radiotracers used include 18 F-FDG, 18 F-<br />

fluorodopa, 11 C- methionine, 11 C-flumazenil, 15 O-water. Neurological indications for <strong>PET</strong><br />

scanning include [11]:<br />

1. Brain tumor staging (neurooncology).<br />

<strong>PET</strong> is a non-invasive, preoperational method to stage tumors, especially gliomas, based<br />

on the activity <strong>of</strong> glucose uptake in white matter. Highly malignant gliomas display a higher<br />

use <strong>of</strong> glucose than low-grade tumors. Post-surgery <strong>PET</strong> helps detect residual tumor mass and<br />

to recurrence from radionecrosis.<br />

2. Detecting and assessing epileptic foci.<br />

<strong>PET</strong> is helpful especially in evaluating temporal epilepsy. During epileptic seizures, the<br />

glucose intake level <strong>of</strong> the epileptic focus grows, but drops between seizures. <strong>PET</strong> is also useful<br />

in evaluating drug-resistant childhood epilepsy.<br />

3. Evaluation <strong>of</strong> dementia-type disorders<br />

In Alzheimer’s disease patients, low glucose metabolism, and low regional brain blood flow<br />

and regional oxygen consumption are observed in frontal, parietal and temporal lobes. In<br />

multi-infarct dementia, the disorders above are anatomically associated with the regions <strong>of</strong><br />

pathological changes <strong>of</strong> arterial vascularization.<br />

4. Motoric dysfunction<br />

In Parkinson’s disease patients, low uptake <strong>of</strong> 18 F-fluorodopa in the putamen is apparent. In<br />

Huntington’s disease, decrease in glucose metabolism is evident in the striatum, at later<br />

stages also in the frontal lobes. Other motoric dysfunction syndromes assessable by <strong>PET</strong><br />

include multisystemic atrophy, corticobasal degeneration, progressive supranuclear palsy,<br />

and Wilson’s disease.<br />

5. Assessment <strong>of</strong> brain tissue ischemia.<br />

<strong>PET</strong> is helpful in assessing cortical strokes, where it is used to measure changes in regional<br />

brain blood flow before and after therapeutic intervention. It also allows for an early<br />

identification <strong>of</strong> low regional brain blood flow in patients with subarachnoidal hemorrhage<br />

symptoms.<br />

6. Neuroactivation.<br />

15 O-water <strong>PET</strong> is used to evaluate neuronal activity for the purposes <strong>of</strong> locating specific<br />

brain functions such as motoric behaviors or cognitive functions.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

4.4.3. Cardiology<br />

<strong>PET</strong> scanning <strong>of</strong> the heart uses 82 Rb-chloride, 13 N-ammoniac, 18 F-FDG and 11 C- palmitic<br />

acid. Cardiologic indications for <strong>PET</strong> studies include [14]:<br />

1. Diagnostics <strong>of</strong> coronary disease.<br />

<strong>PET</strong> is used to determine the extent and intensiveness <strong>of</strong> heart muscle damage arising from<br />

ischemic disease, by indicating loss <strong>of</strong> perfusion in the heart muscle.<br />

2. Assessment heart muscle vitality<br />

18 F-FDG examination is essential before the revascularisation procedure as it differentiates<br />

regions <strong>of</strong> “frozen” myocardium from postinfarct scars.<br />

4.5. Results<br />

<strong>PET</strong>/<strong>CT</strong> fusion in diagnostic imaging allows more precise classification <strong>of</strong> lesions visible in<br />

<strong>PET</strong> studies as it narrows down their anatomical location. These days, <strong>PET</strong>/<strong>CT</strong> is replacing <strong>PET</strong><br />

and is the fastest developing imaging method. Modern <strong>PET</strong> scanners and multi-detector-row<br />

<strong>CT</strong> scanners reduce the testing time remarkably, and faster scanning makes the use <strong>of</strong><br />

radiopharmaceuticsals more efficient 13 .<br />

13 1. Rohren EM, Turkington TG, Coleman RE. Clinical applications <strong>of</strong> <strong>PET</strong> in oncology. Radiology 2004; 231: pp 305–<br />

332.<br />

2. Pruszyński B. Osiągnięcia diagnostyki obrazowej na przełomie roku 2003/2004. www.schering.pl<br />

3. Hany TF, Steinert HC, Goerres GW et al. <strong>PET</strong> diagnostic accuracy: improvement with in-line <strong>PET</strong>-<strong>CT</strong> system; initial<br />

results. Radiology 2002; 225: pp 575–581.<br />

4. Gould MK, Maclean CC, Kuscher WG et al. Accuracy <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography for diagnosis <strong>of</strong><br />

pulmonary nodules and mass lesions; a meta <strong>analysis</strong>. JAMA 2001; 285: pp 914–924.<br />

5. Patz EF, Lowe VJ, H<strong>of</strong>fman JM et al. Focal pulmonary abnormalities: evaluation with F-18 fluorodeoxyglucose <strong>PET</strong><br />

scanning. Radiology 1993; 188: pp 487–490.<br />

6. Shim SS, Lee KS, Kim BT et al. Focal parenchymal lung lesions showing a potential <strong>of</strong> false positive and falsenegative<br />

interpretations on integrated <strong>PET</strong>/<strong>CT</strong>. AJR 2006; 186: pp 639–648.<br />

7. Erasmus JJ, McAdams HP, Rossi SE et al. FDG <strong>PET</strong> <strong>of</strong> pleural effusions in patients with non- small lung cancer. AJR<br />

2000; 175: pp 245–249.<br />

8. Patz EF, Lowe VJ, Goodman PC et al. Thoracic nodal staging with <strong>PET</strong> imaging with 18-FDG in patients with<br />

bronchogenic carcinoma. Chest 1995; 108: pp 1617–1621.<br />

9. Jednoróg S, Mazur G, Janiak KM et all, KrótkoŜyciowe izotopy pierwiastków lekkich jako znaczniki tomografii<br />

pozytronowej (<strong>PET</strong>). Problemy Medycyny Nuklearnej 1998; 12 (24): pp 191–200.<br />

10. Kochanowicz E, Kulka J. Detekcja i rekonstrukcja obrazu w <strong>PET</strong>. Problemy Medycyny Nuklearnej 2002; 16 (31):<br />

pp 123-132.<br />

11. Chmielowski K. Emisyjna tomografia pozytronowa (<strong>PET</strong>) w neurologii. Problemy Medycyny Nuklearnej 2002; 16<br />

(31): pp 143–150.<br />

12. <strong>PET</strong>-<strong>CT</strong> Consensus Conference: SNMTS; American Society <strong>of</strong> Radiologic Technologist (ASRT). Fusion imaging:<br />

a new type <strong>of</strong> technologist for a new type <strong>of</strong> technology. J Nucl Med Technol 2002; 30: pp 201–204.<br />

13. Brink JA. <strong>PET</strong>/<strong>CT</strong> unplugged: the merging technologies <strong>of</strong> <strong>PET</strong> and <strong>CT</strong> imaging. AJR 2005; 184: pp S135–S137.<br />

14. Thrall JH, Ziessman HA. Nuclear Medicine. Mosby 2001 St. Louis.<br />

91


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

5. METHODOLOGY<br />

92<br />

5.1. Purpose <strong>of</strong> study<br />

The purpose <strong>of</strong> this report is a comparative cost <strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with<br />

diagnostic technologies financed in Poland from public sources in oncological<br />

diagnostics. This part <strong>of</strong> the report provides a clinical and epidemiological <strong>analysis</strong>.<br />

The study was ordered by the Agency for Health Technology Assessment in Poland.<br />

5.2. Method <strong>of</strong> clinical efficacy assessment<br />

Clinical questions were prepared consistent with the subject <strong>of</strong> the <strong>analysis</strong>;<br />

A search strategy for scientific studies was designed;<br />

The world’s key medical databases and clinical studies registers were searched for<br />

primary studies;<br />

Primary studies were selected based on pre-defined inclusion criteria;<br />

Trial results and their statistical and clinical significance were reviewed;<br />

A meta-<strong>analysis</strong> <strong>of</strong> the results <strong>of</strong> the trials included was executed;<br />

The comparative <strong>analysis</strong> results were interpreted and conclusions formulated.<br />

The search strategy for primary studies, the assessment <strong>of</strong> studies reliability, as well as data<br />

extraction, statistical <strong>analysis</strong> and results interpretation methods were designed based on<br />

guidelines <strong>of</strong> the Medical Services Advisory Committee [„Guidelines for the assessment <strong>of</strong><br />

diagnostic technologies” August 2005, Australia].<br />

5.3. Search strategy for primary studies<br />

As the first step <strong>of</strong> searching for trials a general search strategy was adopted without<br />

further specifying the target population, in order to locate a group <strong>of</strong> trials concerning the<br />

use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in oncological diagnostics. Additionally, for 12 indications individual searches<br />

were conducted with targeted population (breast cancer, lung cancer, colorectal<br />

cancer, ovarian cancer, cervical cancer, endometrial cancer, lymphoma, gastric and<br />

esophageal cancer, melanoma, glioma, sarcoma, head and neck cancer). In the process<br />

<strong>of</strong> searching for reports on the technology under assessment, results <strong>of</strong> a high specificity


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

strategy that identified only those publications where the hybrid term “<strong>PET</strong>-<strong>CT</strong>” or its<br />

equivalents appeared (#9) – on the basis <strong>of</strong> search strategy presented in „Revision<br />

Sistematica Sobre la Efectividad E Indicationes del Sistema Hibrido <strong>PET</strong>-TAC.” Unidad de<br />

Evaluacion de Tecnologias Sanitarias 2006. Agencia Lain Entralgo, were combined with<br />

results <strong>of</strong> a high sensitivity strategy that identified all publications describing the diagnostic<br />

method under discussion (search for publications with “<strong>PET</strong>” and “<strong>CT</strong>” titles, abstracts or<br />

keywords - #8).<br />

Table 16.<br />

Search strategy<br />

ID<br />

Search strategy<br />

#1 „Positron-Emission Tomography” [MeSH] OR „Tomography, Emission-Computed” [MeSH]<br />

#2 <strong>PET</strong> OR <strong>positron</strong> <strong>emission</strong> tomography<br />

#3 #1 OR #2<br />

#4 („Tomography, Spiral Computed” [MeSH] OR „Tomography, X-Ray Computed” [MeSH])<br />

#5 <strong>CT</strong> OR computed tomography<br />

#6 #4 OR #5<br />

#7<br />

dual OR integral OR integration OR combination OR combined OR fusion OR fused OR hybrid OR<br />

coincidental OR combining OR coincidence OR coregistered<br />

#8 #3 AND #6<br />

#9 #3 AND #7<br />

#10 #8 OR #9<br />

#11 „Neoplasms” [MeSH]<br />

#12 „Medical Oncology” [MeSH]<br />

#13 #11 OR #12<br />

#14 #10 AND #13<br />

#15 #10 AND #13 Limits: English, Polish, Publication Date from 1998/01/01, Humans<br />

Last search date: 20-03-2006.<br />

To design the search strategy, a database <strong>of</strong> medical terms was used (Medical Subject<br />

Headings – MeSH). The database includes thematic entries with synonyms and terms<br />

close in meaning assigned to them. Linking the database search with the broad terms <strong>of</strong><br />

the MeSH database allows to avoid ignoring publications with non-standard terminology.<br />

Detailed search results including described search strategies are presented in appendix.<br />

Reviews and meta<strong>analysis</strong> were searched on the grounds <strong>of</strong> search strategy given in table<br />

16 with replacement R<strong>CT</strong> filter with “review” or “meta<strong>analysis</strong>” limitations.<br />

5.4. Medical database search<br />

The above search strategy was applied to the following databases:<br />

Medline by Pubmed,<br />

93


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

94<br />

Cochrane Library (The Cochrane Database <strong>of</strong> Systematic Reviews, The Cochrane<br />

Con- trolled Trials Register),<br />

EmBase,<br />

and medical web sites:<br />

NICE (National Institute for Clinical Excellence),<br />

SBU (Statens beredning för medicinsk utvärdering),<br />

NCCHTA (The National Coordinating Centre for Health Technology Assessment),<br />

CADTH (The Canadian Agency for Drugs and Technologies in Health),<br />

INAHTA (International Network <strong>of</strong> Agencies for Health Technology Assessment),<br />

MSAC (Medical Services Advisory Committee),<br />

CRD (Centre for Reviews and Dissemination):<br />

DARE (Database <strong>of</strong> Abstracts <strong>of</strong> Reviews <strong>of</strong> Effects),<br />

NHS EED (NHS Economic Evaluation Database),<br />

Health Technology Assessment Database,<br />

Ongoing Reviews Database<br />

Also, the bibliographies <strong>of</strong> the primary studies were searched. Secondary studies were<br />

reviewed (object articles, systematic reviews, meta-analyses, agency reviews) for possible<br />

additional primary studies. Moreover, to make the list <strong>of</strong> publications complete,<br />

manufacturers <strong>of</strong> diagnostic equipment were requested to provide all their studies,<br />

especially those concerning the pre-marketing period, as well as marketing information<br />

on <strong>PET</strong>-<strong>CT</strong> scanners.<br />

The search was conducted independently by two researchers. Next, trial abstracts were<br />

reviewed for inclusion into the <strong>analysis</strong>, also by two independent researchers.<br />

5.5. Criteria for the inclusion <strong>of</strong> primary studies in the <strong>analysis</strong><br />

Initially, titles and abstracts in the publications found were analyzed. If information on the<br />

use <strong>of</strong> <strong>PET</strong> (including <strong>PET</strong>-<strong>CT</strong>) imaging or <strong>CT</strong> in a trial was found, it was qualified for further<br />

verification depending on how the full text was evaluated.<br />

As the second stage, full texts in an electronic version were analyzed with a special focus<br />

on trial methodology, in order to identify information on the use <strong>of</strong> hybrid <strong>PET</strong>-<strong>CT</strong> scanners.<br />

Reports where the use <strong>of</strong> the technique under discussion was confirmed were further verified.<br />

Based on the guidelines <strong>of</strong> the Medical Services Advisory Committee, the verification process<br />

focused on the selection <strong>of</strong> trials, where two strategies were assessed at the same time: <strong>PET</strong>-<br />

<strong>CT</strong> and another strategy financed in Poland. On this basis a decision was made whether to<br />

include or exclude a trial, stating the reasons.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

In case <strong>of</strong> primary studies, there were no restrictions on the period <strong>of</strong> follow-up or size <strong>of</strong><br />

population. Trials done on humans, for which full texts <strong>of</strong> reports are accessible in Polish<br />

libraries in English or Polish were included. Restrictions were defined for the date <strong>of</strong><br />

publication, which narrowed down the scope <strong>of</strong> search to studies published after 1 Jan 1998.<br />

This criterion was accommodated based on the results <strong>of</strong> database searches (publication <strong>of</strong><br />

first reports on the hybrid <strong>PET</strong>-<strong>CT</strong> scanner) and the date <strong>of</strong> launching the technology under<br />

assessment on the diagnostic procedures market.<br />

5.5.1. Population<br />

The population were patients diagnosed for oncological indications.<br />

5.5.2. Intervention<br />

The <strong>analysis</strong> included primary prospective or retrospective clinical trials, where <strong>PET</strong>-<strong>CT</strong> results<br />

were assessed (regardless <strong>of</strong> the type <strong>of</strong> radiopharmaceutical used). The essential criterion<br />

for the inclusion <strong>of</strong> a publication into the <strong>analysis</strong> was whether the diagnostic techniques<br />

compared were verified through an independent reference test (gold standard).<br />

5.5.3. Technologies compared (comparators)<br />

Diagnostic technologies financed in Poland from public sources and used in oncological<br />

diagnostics: computed tomography, magnetic resonance image, endoscopic ultrasoundith<br />

fine needle aspiration, scintigraphy I131, and the others.<br />

5.5.4. End points<br />

• Diagnostic efficacy <strong>of</strong> testing methods in comparison to the respective reference<br />

method or clinical observation;<br />

• Change <strong>of</strong> therapeutic decision;<br />

• Impact on clinical end points;<br />

• Safety.<br />

5.6. Trial quality assessment<br />

The adopted scale <strong>of</strong> trial quality helps evaluate publications with regard to whether they<br />

are properly designed and conducted, and ensures reliability <strong>of</strong> results.<br />

95


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

96<br />

In compliance with EBM requirements (Evidence Based Medicine), the process <strong>of</strong> trial<br />

quality assessment used the QUADAS 14 form, which consisted <strong>of</strong> 14 questions. The first four<br />

questions check whether the choice <strong>of</strong> population was right; questions 5-9 assess whether the<br />

measurements and the reference test were executed correctly, the remaining questions<br />

rated the interpretation and presentations <strong>of</strong> the results. Questions 3-7 were acknowledged<br />

as critical, becouse answer “yes” was a requirement for study inclusion into <strong>analysis</strong>. Residual<br />

questions provided additional study quality assessment.<br />

In order to standardize the assessment procedure, a single form was prepared to verify all<br />

the publications included in the <strong>analysis</strong>.<br />

Each study was rated independently by two analysts. In case <strong>of</strong> discrepancies, the final<br />

decision was made through formal consensus.<br />

Each question was evaluated “yes”, “no”, or “unclear”. Checklists <strong>of</strong> trial quality assessment<br />

for each study are given in an appendix hereto.<br />

5.7. Statistical <strong>analysis</strong><br />

The calculation <strong>of</strong> the basic parameters <strong>of</strong> the diagnostic efficacy <strong>of</strong> a screening<br />

method is based on the data scheme in table 17, which indicate the number <strong>of</strong> patients<br />

testing positive (T+) and negative (T-) in experimental tests administered to patients with<br />

(D+) or without (D-) a disease as established based on reference (index) test (also called<br />

the gold standard).<br />

Table 17.<br />

Measurement results <strong>of</strong> assessing diagnostic tests<br />

Reference test<br />

Test under assessment<br />

Positive result <strong>of</strong> test under assessment (T+)<br />

Negative result <strong>of</strong> test under assessment<br />

(T-)<br />

Disease (D+)<br />

No disease (D+)<br />

a (TP) b (FP)<br />

c (FN) d (TN)<br />

a - number <strong>of</strong> patients testing positive (i.e. number <strong>of</strong> true positives - TP)<br />

B - number <strong>of</strong> patients without disease but testing positive (i.e. number <strong>of</strong> false<br />

positives - FP)<br />

c - number <strong>of</strong> patients testing negative (i.e. number <strong>of</strong> false negatives - FN)<br />

14 Whiting P, Rutjes AWS, Reitsma JB, Bossuyt PMM, Kleijen J. The development <strong>of</strong> QUADAS: a tool for the quality<br />

assessment <strong>of</strong> studiem <strong>of</strong> diagnostic accuracy included in systematic reviews. BMC Medical Research Methodology<br />

2003; 3 (25).


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

d - number <strong>of</strong> patients without disease but testing negative (i.e. number <strong>of</strong> true<br />

negatives - TN)<br />

Table 18 gives the definitions <strong>of</strong> the basic parameters <strong>of</strong> diagnostic efficacy applied in<br />

the report.<br />

Table 18.<br />

Characteristics <strong>of</strong> parameters <strong>of</strong> diagnostic efficacy<br />

Parameter<br />

<strong>of</strong> diagnostic efficacy<br />

Sensitivity<br />

Specificity<br />

Accuracy<br />

Positive likelihood ratio<br />

Negative likelihood ratio<br />

Diagnostic odds ratio<br />

Positive predictive value<br />

Negative predictive value<br />

Interpretation<br />

Mathematical formula<br />

Se =<br />

Proportion <strong>of</strong> patients testing positive;<br />

reflect the test’s ability to detect disease a + c<br />

Proportion <strong>of</strong> patients testing negative;<br />

describes the test’s ability to confirm<br />

absence <strong>of</strong> disease<br />

Proportion <strong>of</strong> patients correctly<br />

diagnosed by the test<br />

Likelihood quotient for positive test results<br />

in patients with disease and for the same<br />

result in patients not diagnosed with<br />

disease<br />

Likelihood quotient for negative test results<br />

in patients with disease and for the same<br />

result in patients not diagnosed with<br />

disease<br />

Odds ratio <strong>of</strong> testing positive by patients<br />

with disease and the odds <strong>of</strong> obtaining<br />

the same test result for patients without<br />

disease; alternative interpretation: the<br />

odds <strong>of</strong> disease developing in patients<br />

testing positive and <strong>of</strong> disease<br />

developing in patients testing negative<br />

The likelihood <strong>of</strong> the disease tested<br />

actually developing in patients testing<br />

positive<br />

a<br />

d<br />

Sp =<br />

b + d<br />

a + d<br />

Acc =<br />

a + b + c + d<br />

a / a + c Se<br />

LR+<br />

= =<br />

b / b + d 1−<br />

Sp<br />

c / a + c 1−<br />

Se<br />

LR−<br />

= =<br />

d / b + d Sp<br />

a / c<br />

DOR =<br />

b / d<br />

=<br />

a<br />

PPV =<br />

a + b<br />

NPV<br />

The likelihood <strong>of</strong> absence <strong>of</strong> the disease<br />

tested in patients testing negative c + d<br />

d<br />

LR +<br />

LR −<br />

Included in the <strong>analysis</strong> are trials that sought information on the results <strong>of</strong> the diagnostic<br />

methods under assessment in comparison with a reference method. The results were grouped<br />

according to the parameters <strong>of</strong> a four-field table: TP, TN, FP and FN.<br />

Next, in order to assess the diagnostic efficacy, parameters such as sensitivity (Se),<br />

specificity (Sp), accuracy (Acc), positive likelihood ratio (LR+), negative likelihood ratio (LR-)<br />

and diagnostic odds ratio (DOR) were calculated with a 95% confidence interval. Whenever<br />

=<br />

97


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

possible, a meta-<strong>analysis</strong> <strong>of</strong> results <strong>of</strong> studies was also conducted. To evaluate differences in<br />

diagnostic efficacy <strong>of</strong> trials, McNemar’s test was done, provided data were available (test <strong>of</strong><br />

proportions with matched data array).<br />

98<br />

For a comparative rating <strong>of</strong> odds <strong>of</strong> accurate diagnosis (consistent with the reference<br />

standard) for the diagnostic methods under assessment, the OR parameters statistical<br />

significance was rated based on EBM guidelines. For statistically significant results, the NNT<br />

parameter was additionally calculated, specifying confidence intervals.<br />

When no statistically significant discrepancies <strong>of</strong> study results were identified, a meta-<br />

<strong>analysis</strong> <strong>of</strong> the results was conducted using the fixed effect model. For end points that were<br />

statistically significant in a heterogeneity test, the random effect model was used. The<br />

statistical s<strong>of</strong>tware MetaDiSc v.1.3 was used to calculate these parameters and to carry out a<br />

meta-analyses <strong>of</strong> sensitivity and specificity. The meta-<strong>analysis</strong> <strong>of</strong> accuracy, OR and NNT, and<br />

graphs representing this results were done in Stats Direct v. 2.5.2.<br />

5.8. Conflict <strong>of</strong> interest<br />

None <strong>of</strong> the authors <strong>of</strong> the study reported any conflict <strong>of</strong> interest.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

6. SEARCH RESULTS<br />

Based on the results <strong>of</strong> a search for publications that describe the diagnostic efficacy<br />

<strong>of</strong> the hybrid <strong>PET</strong>-<strong>CT</strong> scanner, which was executed in keeping with the search strategy<br />

presented above, a list <strong>of</strong> trials that tentatively met the inclusion criteria was compiled.<br />

Next, the full texts <strong>of</strong> these trials were reviewed, and consequently a final list <strong>of</strong> trials<br />

included was established. The results <strong>of</strong> this review are given in Table 19.<br />

Table 19.<br />

List <strong>of</strong> the results <strong>of</strong> a search targeted at selected patient populations.<br />

Name <strong>of</strong> cancer<br />

Number <strong>of</strong> abstracts read<br />

Number <strong>of</strong> full texts<br />

received from libraries<br />

Number <strong>of</strong> trials included<br />

in <strong>analysis</strong><br />

1. Lung cancer 1520 250 4<br />

2. Ovarian cancer 138 96 2<br />

3. Cervical cancer 248 86 0<br />

4. Sarcoma 210 111 1<br />

5. Glioma 269 192 0<br />

6. Lymphoma 641 72 2<br />

7. Head and neck cancer 932 331 4<br />

8. Esophageal and gastric<br />

cancer<br />

393 117 2<br />

9. Colorectal cancer 422 190 1<br />

10. Breast cancer 545 86 0<br />

11. Melanoma 213 113 0<br />

12. Endometrial cancer 32 24 0<br />

Total 5563 1754 16<br />

5563 publications describing clinical trials in form <strong>of</strong> abstracts were found as a result <strong>of</strong><br />

searching databases according to the established strategy extended with population<br />

selection. Those abstracts were initially reviewed with respect to the diagnostic<br />

technology applied. The full texts <strong>of</strong> the items selected (1754) were next reviewed taking<br />

into account all inclusion criteria. Finally, 16 trials, that met the inclusion criteria were<br />

selected and reviewed in detail for the results reported. Detailed general search results<br />

without aiming at patient population are given in section “18 Appendix”. As a result <strong>of</strong><br />

that search strategy 10 clinical trials were included additionally: 3 trials for thyroid cancer,<br />

1 for gynecological malignancies, 1 for pancreatic cancer and 5 for various oncological<br />

diseases. Finally, 26 clinical trials were incuded into <strong>analysis</strong>.<br />

Three HTA reports concerning <strong>PET</strong>-<strong>CT</strong> were found:<br />

99


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

100<br />

• „<strong>CT</strong>-<strong>PET</strong> scanners - systematic review, expert panel”. Paris: Comite d'Evaluation et<br />

de Diffusion des Innovations Technologiques (CEDIT), 2002 – only abstract in english;<br />

• „Revision Sistematica Sobre la Efectividad E Indicationes del Sistema Hibrido <strong>PET</strong>-<br />

TAC.” Unidad de Evaluacion de Tecnologias Sanitarias 2006. Agencia Lain<br />

Entralgo. – available in spanish only;<br />

• „Combined <strong>positron</strong> <strong>emission</strong> tomography (<strong>PET</strong>-<strong>CT</strong>)”. Cigna Healthcare Coverage<br />

Position 2005 – available only with high fee.


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

7. COMPARATIVE ANALYSIS OF THE EFFICACY OF <strong>PET</strong>-<strong>CT</strong> VS<br />

<strong>CT</strong> IN LUNG CANCER STAGING<br />

7.1. Results <strong>of</strong> trial search<br />

As a result <strong>of</strong> searching medical databases 4 primary prospective trials were identified:<br />

Antoch 2003 (tab. 135, app. 18.1), Lardinois 2003 (tab. 136, app. 18.1), Cerfolio 2005 (tab.<br />

137, app. 18.1) and Shim 2005 (tab. 138, app. 18.81), where <strong>PET</strong>-<strong>CT</strong> was used in lung<br />

cancer staging.<br />

In each <strong>of</strong> the trials, a hybrid <strong>PET</strong>-<strong>CT</strong> scanner was compared with <strong>CT</strong>. Both methods<br />

were verified based on a histopathological index test.<br />

7.2. Population characteristics<br />

In all the four studies, patients with non-small cell lung cancer (NSCLC) took part.<br />

Pre-surgery staging was executed in three <strong>of</strong> the trials (Lardinois 2003, Cerfolio 2005, Shim<br />

2005). In Antoch 2003, staging was executed for 19 out <strong>of</strong> 27 patients (70%), and restaging<br />

following neoadjuvant chemotherapy was executed for 8 out <strong>of</strong> 27 patients (30%).<br />

Cerfolio 2005 and Shim 2005 excluded patients receiving pre-surgery chemotherapy or<br />

radiotherapy. Cerfolio 2005 additionally excluded patients with type I diabetes. The authors<br />

<strong>of</strong> the report in Shim 2005 reported that 33% patients had had tuberculosis confirmed<br />

clinically or by imaging.<br />

Table 20 lists the initial characteristics <strong>of</strong> patients for each <strong>of</strong> the trials.<br />

Table 20.<br />

Initial characteristics <strong>of</strong> patients by trial<br />

Parameter<br />

Size <strong>of</strong> population<br />

Age [years]<br />

Male proportion<br />

Histology<br />

adenocarcinoma<br />

squamous cell<br />

carcinoma<br />

(proportion <strong>of</strong><br />

patients)<br />

large cell carcinoma<br />

bronchoalveolar<br />

carcinoma<br />

Antoch<br />

2003<br />

27<br />

mean: 56<br />

85%<br />

no data<br />

no data<br />

no data<br />

no data<br />

Lardinois<br />

2003<br />

50<br />

mean: 62<br />

57%<br />

57%<br />

27%<br />

16%<br />

0%<br />

Cerfolio<br />

2005<br />

383<br />

mean: 68<br />

59%<br />

38%<br />

44%<br />

0%<br />

3%<br />

Shim 2005 Total<br />

110<br />

mean: 56<br />

74%<br />

51%<br />

41%<br />

4%<br />

3%<br />

570<br />

mean: 58*<br />

63%<br />

42%<br />

42%<br />

2%<br />

3%<br />

101


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

102<br />

neuroendocrinal<br />

carcinoma<br />

carcinoid<br />

pleomorphic cancer<br />

no data<br />

no data<br />

no data<br />

other NSCLC types,<br />

mixed types or no data 0%<br />

unidentified type<br />

*average weighted by the number <strong>of</strong> patient participating in the three trials<br />

In Antoch 2003, 27 patients diagnosed with NSCLC were included. In Lardinois 2003, 50<br />

patients with documented or suspected NSCLC were included, but ultimately 49<br />

participated (one patient was diagnosed with lymphoma). In Cerfolio 2005, patients with<br />

confirmed non-small cell lung cancer or a lung nodule were been included. Ultimately 383<br />

patients were included.<br />

110 patients with documented NSCLC were included in Shim 2005; surgeries and<br />

histopathological tests were done for 106 patients and the results were reviewed.<br />

The total <strong>of</strong> 570 patients aged 56-68 on the average, with the male population<br />

representing 57–85% took part in the study. The proportion <strong>of</strong> patients with<br />

adenocarcinoma identified histologically ranged from 38% to 57%,, and the proportion <strong>of</strong><br />

patients with squamous cell carcinoma ranged from 27% to 44%. Other histological<br />

cancer types represented a small proportion.<br />

7.3. Description <strong>of</strong> Intervention<br />

7.3.1. <strong>PET</strong>-<strong>CT</strong> imaging<br />

In three trials included in the study (Lardinois 2003, Cerfolio 2005 and Shim 2005)<br />

the Discovery LS system manufactured by General Electric Medical Systems (Milwaukee,<br />

USA) was used for staging, and in Antoch 2003 the system was Biograph by Siemens<br />

Medical Solutions (H<strong>of</strong>fman Estates, USA). The authors <strong>of</strong> Lardinois 2003 and Shim 2005<br />

noted that the <strong>PET</strong>-<strong>CT</strong> system consisted <strong>of</strong> a <strong>PET</strong> scanner (Advance NXi, GE Medical<br />

Systems) and a <strong>CT</strong> scanner (LightSpeed Plus, GE Medical Systems).<br />

In all the trials included, FDG (fluoro-deoxy-glucose) was used as radiopharmaceutical, its<br />

activity ranging from 350 MBq in Antoch 2003, to 350 - 400 MBq in Lardinois 2003, 370 MBq in<br />

Shim 2005, and 555 MBq in Cerfolio 2005. The radiopharmaceutical was administered 50<br />

and 60 minutes before a <strong>PET</strong>-<strong>CT</strong> scan was taken in Lardinois 2003 and Antoch 2003,<br />

respectively. The authors <strong>of</strong> the other two trials give no information on FDG.<br />

Table 21 contains a detailed description <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning<br />

0%<br />

0%<br />

0%<br />

2%<br />

3%<br />

0%<br />

11%<br />

0%<br />

0%<br />

2%<br />

0%<br />

1%<br />

2%<br />

0,4%<br />

8%


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Table 21.<br />

Description <strong>of</strong> intervention<br />

Trial<br />

Antoch 2003<br />

Lardinois<br />

2003<br />

Cerfolio<br />

2005<br />

Shim<br />

2005<br />

<strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

type<br />

Biograph<br />

Discovery<br />

LS<br />

Discovery<br />

LS<br />

Discovery<br />

LS<br />

<strong>PET</strong>-<strong>CT</strong> scanner<br />

manufacturer<br />

Siemens Medical<br />

Solutions<br />

(H<strong>of</strong>fman Estates, Ill)<br />

GE Medical Systems<br />

(Milwaukee, WI)<br />

GE Medical Systems<br />

(Milwaukee, WI)<br />

GE Medical Systems<br />

(Milwaukee, WI)<br />

Radiopharmace<br />

utical type and<br />

administration<br />

method<br />

FDG intravenous<br />

FDG intravenous<br />

FDG intravenous<br />

FDG intravenous<br />

Trial range<br />

whole body<br />

whole body<br />

whole body<br />

whole body<br />

Radiomarker<br />

activity<br />

[MBq]<br />

350<br />

350–400<br />

In Cerfolio 2005 and Shim 2005, patients had not eaten 4 and 6 hours prior to the test<br />

respectively. This information is unavailable for the other studies.<br />

In Antoch 2003, the image obtained from <strong>PET</strong>-<strong>CT</strong> scanning was evaluated by a team<br />

made up <strong>of</strong> a radiologist and nuclear medicine expert, each with 1.5 years’ experience<br />

with <strong>PET</strong>-<strong>CT</strong>. They had had access to the patient’s case history (so did the <strong>CT</strong> team).<br />

For Lardinois 2003, two medical boards were created, each composed <strong>of</strong> a nuclear<br />

radiology expert and thoracic surgeon. One <strong>of</strong> the boards first evaluated the result <strong>of</strong> <strong>PET</strong><br />

scanning and next <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning. The board knew no clinical data or other diagnostic<br />

imaging results. In Cerfolio 2005, a <strong>PET</strong>-<strong>CT</strong> image was analyzed by a nuclear medicine<br />

consultant. In Shim 2005, the assessment was done by one <strong>of</strong> two nuclear medicine<br />

consultants.<br />

7.3.2. Diagnostic test compared<br />

In all the trials included in this <strong>analysis</strong>, <strong>PET</strong>-<strong>CT</strong> tests were compared with <strong>CT</strong>.<br />

In Antoch 2003, <strong>CT</strong> was executed using the Somaton Emotion spiral technique<br />

(Siemens Medical Solutions, Erlangen, Germany); in Lardinois 2003, the Light Speed Plus<br />

scanner was used; in Shim 2005, the HighLight or Light Speed Ultra 16 spiral technique<br />

(GE Medical Systems) was used; in Cerfolio 2005, the authors give no information on the<br />

<strong>CT</strong> scanner used.<br />

Whole-body scans were taken in Antoch 2003, while in Lardinois 2003 the <strong>CT</strong> image<br />

covered the thorax and pelvis. In Shim 2005, the area scanned extended from the<br />

supraclavicular area halfway through the kidneys, while for Cerfolio 2005 this information<br />

is unavailable.<br />

Intravenous and oral contrast medium was used in Antoch 2003; intravenous contrast<br />

555<br />

370<br />

103


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

104<br />

medium was used in Shim 2005 and Lardinois 2003; the authors <strong>of</strong> Cerfolio 2005 provide<br />

no information on the contrast medium use.<br />

Table 22 provides detailed descriptions <strong>of</strong> <strong>CT</strong> for each <strong>of</strong> the trials.<br />

Table 22.<br />

Description <strong>of</strong> diagnostic tests compared<br />

Trial<br />

Antoch 2003<br />

Lardinois 2003<br />

Cerfolio 2005<br />

Shim 2005<br />

<strong>CT</strong> scanner type<br />

Somaton Emotion<br />

Light Speed Plus<br />

no data<br />

HighLight or Light<br />

Speed Ultra 16<br />

<strong>CT</strong> scanner producer<br />

Siemens Medical<br />

Solutions<br />

GE Medical Systems<br />

no data<br />

GE Medical Systems<br />

Contrast medium<br />

type and method<br />

intravenous and oral<br />

contrast medium<br />

intravenous contrast<br />

medium<br />

no data<br />

intravenous contrast<br />

medium<br />

Study range<br />

whole body<br />

from head to<br />

pelvis<br />

no data<br />

from supraclavicular to<br />

half the height <strong>of</strong><br />

kidneys<br />

In Antoch 2003, diagnostic images were evaluated by two radiologists with 8 and 11<br />

years’ experience. In Lardinois 2003, the other board analyzed <strong>CT</strong> scans, followed by <strong>PET</strong><br />

scans, and next compared the images. In Cerfolio 2005 and Shim 2005, the results were<br />

interpreted by an experienced thoracic radiologist, who in Shim 2005 was not familiar<br />

with the results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning, pathological tests, or the clinical condition, save for<br />

the information on the presence <strong>of</strong> lung cancer.<br />

7.3.3. Reference test<br />

Histopathological examination was a reference test for all the trials included in this<br />

<strong>analysis</strong>. In two clinical trials metastases were confirmed also by clinical follow-up<br />

(Lardinois 2003) or MRI (Cerfolio 2005), morover in the second trial ultrasonography was<br />

used in nodal staging.<br />

In Antoch 2003, mediastinal endoscopy and resection <strong>of</strong> lymph nodes were executed<br />

by an experienced thoracic surgeon. Mediastinal endoscopy was recognized sufficient<br />

as long as it helped remove, and complete Histopathological examination <strong>of</strong><br />

paratracheal, tracheobronchial and subcranial lymph nodes. A precise dissection <strong>of</strong><br />

lymph nodes was executed as part <strong>of</strong> thoracotomy, but supraclavicular lymph nodes<br />

were spared in both mediastinal endoscopy and thoracotomy. Tumor and lymph nodes<br />

were removed in a surgical procedure in 16 patients, and only for this group was T-staging<br />

possible. Mediastinoscopy was done for 11 patients. No other treatment was applied in


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

between the imaging and surgery.<br />

In Lardinois 2003, histopathological tests were done following lung resection with<br />

dissection <strong>of</strong> mediastinal lymph nodes. 82% <strong>of</strong> patients were given a surgery: 71% had<br />

mediastinal lymph nodes removed, 6% underwent exploratory thoracotomy, and 4%<br />

underwent sphenoid resection. Surgery was not possible in 9 patients due to metastases<br />

outside thorax (8 patients) and the presence <strong>of</strong> carcinomous cells in the pleural fluid (1<br />

patient). A limitation <strong>of</strong> lung functions was observed during a sphenoid resection in 2<br />

patients, which prevented lobectomy. Lymph nodes were not removed in these patients<br />

either as the size <strong>of</strong> mediastinal lymph nodes was less than 5 mm. Out <strong>of</strong> the three patients<br />

who had an exploratory thoracotomy, one was diagnosed with dissemination <strong>of</strong><br />

carcinoma within lungs, and one with aorta infiltration. Therefore full staging was possible<br />

with 35 patients only. Lesions outside the chest were graded on clinical observation and<br />

biopsy.<br />

In Lardinois 2003, the disease was staged with respect to lymph nodes metastasis by<br />

averaging the score ranging from 0 to 3, where 0 meant incorrect staging, 1 – ambiguous<br />

but incorrect staging, 2 – correct but ambiguous staging, and 3 - correct staging. Scores<br />

<strong>of</strong> 0 and 1 were considered jointly in the results analyses as both implied incorrect staging.<br />

In Cerfolio 2005, histopathological examination <strong>of</strong> cancer was executed following the<br />

resection and biopsy <strong>of</strong> all spots suspected <strong>of</strong> metastases (N2, N3, M1). Mediastinal<br />

endoscopy was done in order to perform a biopsy <strong>of</strong> paratracheal areas, while<br />

transesophageal endoscopic ultrasonography was used to perform biopsy <strong>of</strong> the nodes in<br />

the back <strong>of</strong> the aorto-pulmonary window, subcranial nodes, peri-esophageal nodes and<br />

lung ligament nodes suspected <strong>of</strong> metastasis. The nodes were initially scanned using<br />

ultrasound, and the examiner was blinded to the results <strong>of</strong> other diagnostic imaging. For<br />

M-staging <strong>of</strong> liver, adrenal glands or the opposite lung, biopsy was performed, and lesions in<br />

bones and brain were scanned using MRI. Next, the lesions were removed along with lymph<br />

nodes in N2 negative patients.<br />

In Shim 2005, tumor resection <strong>of</strong> mediastinal lymph nodes dissection were performed by<br />

one <strong>of</strong> a pair <strong>of</strong> experienced thoracic surgeons, who had reviewed the imaging tests results.<br />

All palpable lymph nodes were removed. Pneumonectomy, including lobectomy and<br />

removal <strong>of</strong> part <strong>of</strong> a lung with mediastinal lymph node was performed in 106 patients. In 83%,<br />

lobectomy only was possible because the primary tumor was limited to the lobe only; in 8.5%<br />

only bilobectomy was performed because the tumor had disseminated onto another lobe or<br />

bronchi, and in 8.5% part <strong>of</strong> a lung was removed. In this trial, the tumor (histopathological<br />

grade, size, involvement <strong>of</strong> adjacent organs, necrosis, distance to the edge <strong>of</strong> resection)<br />

and the lymph nodes were described by a pathologist with 10 years' experience. Lymph<br />

105


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

nodes were numbered according to the map proposed by Mountain and Dresler.<br />

Ultimately, 393 groups <strong>of</strong> lymph nodes in 106 patients were staged. It is worth mentioning<br />

that in Shim 2005 node lesions instead <strong>of</strong> patients number were used as the basis <strong>of</strong> N-<br />

staging.<br />

106<br />

7.4. Findings<br />

7.4.1. T-staging<br />

T-staging was performed in three <strong>of</strong> the trials included in this part <strong>of</strong> the <strong>analysis</strong> (Antoch<br />

2003, Lardinois 2003 and Shim 2005).<br />

In each <strong>of</strong> the trials the T-staging was based on the TNM system created by the AJCC<br />

(American Joint Committee on Cancer) 15 . The results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> scanning were<br />

verified histopathologically.<br />

Table 23 presents the consistency between the reference test and the results <strong>of</strong> T-<br />

staging done using <strong>of</strong> the methods compared, as quoted by the authors <strong>of</strong> each trial.<br />

Table 23.<br />

Consistency between T-staging and reference test, by <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong> trial.<br />

trial<br />

Antoch<br />

2003<br />

Lardinois<br />

2003<br />

N*<br />

16<br />

40<br />

Shim 2005 106<br />

Correct staging<br />

(accuracy)<br />

n (%)<br />

15 (94%)<br />

35 (88%)<br />

91 (86%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Ambiguous<br />

staging<br />

n (%)<br />

0**<br />

4 (10%)<br />

0**<br />

Incorrect staging<br />

n (%)<br />

Overstag<br />

ed<br />

1 (6%)<br />

4 (4%)<br />

*Number <strong>of</strong> patients verified using reference test<br />

**Ambiguous results were not taken into account in the trial<br />

1 (2%)<br />

Understag<br />

ed<br />

0 (0%)<br />

11<br />

(10%)<br />

Correct<br />

staging<br />

(accuracy)<br />

n (%)<br />

12 (75%)<br />

23 (58%)<br />

84 (79%)<br />

<strong>CT</strong><br />

Ambiguous<br />

staging<br />

n (%) Overstag<br />

ed<br />

0**<br />

8 (20%)<br />

0**<br />

Incorrect staging<br />

n (%)<br />

Understag<br />

ed<br />

3 (19%) 1 (6%)<br />

13<br />

(12%)<br />

9 (22%)<br />

The data presented above show that in all these trials, the proportion <strong>of</strong> correctly<br />

classified patients was higher for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>. The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 86 and 94%,<br />

and the accuracy <strong>of</strong> <strong>CT</strong> is 58 and 79%. The percentage <strong>of</strong> overstaged and understaged<br />

cases is 4 and 6% and 0 and 10% respectively for <strong>PET</strong>-<strong>CT</strong> and 12 and 19% and 6 and 8.5%<br />

for <strong>CT</strong> respectively.<br />

In Antoch 2003, the authors described <strong>PET</strong>-<strong>CT</strong> as more accurate than <strong>CT</strong> alone. The<br />

15 AJCC cancer staging manual. 6th ed. New York, NY: Springer, 2002: 165-177<br />

9<br />

(8,5%)


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

difference between the groups is not statistically significant. In Lardinois 2003, the<br />

difference between the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> was statistically significant<br />

(p = 0,001, a paired sign test) in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>. In Shim 2005, the authors did not report<br />

statistically significant differences between tests in this respect (p=0.25).<br />

The results <strong>of</strong> meta-<strong>analysis</strong> <strong>of</strong> diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in T-staging are presented in<br />

graph 1.<br />

107


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Graph 1.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in T-staging<br />

The diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in T-staging as calculated by meta-<strong>analysis</strong> <strong>of</strong> three trials is<br />

86% (95% CI: 81; 91).<br />

108<br />

Antoch 2003 0,94 (0,70, 1,00)<br />

Lardinois 2003 0,88 (0,73, 0,96)<br />

Shim 2005 0,86 (0,78, 0,92)<br />

Result <strong>of</strong> meta<strong>analysis</strong> 0,86 (0,81, 0,91)<br />

0,6 0,7 0,8 0,9 1,0<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>CT</strong> in T-staging is presented in graph 2. As<br />

there was significant heterogeneity between trials (p = 0.0372) meta-<strong>analysis</strong> was<br />

performed using the random effects method.<br />

Graph 2.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>CT</strong> results in T-staging<br />

Antoch 2003 0,75 (0,48, 0,93)<br />

Lardinois 2003 0,58 (0,41, 0,73)<br />

Shim 2005 0,79 (0,70, 0,87)<br />

Meta-<strong>analysis</strong> result 0,71 (0,55, 0,84)<br />

Diagnostic accuracy (95% confidence interval)<br />

0,4 0,6 0,8 1,0 1,2<br />

Diagnostic accuracy (95% confidence interval)


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

The diagnostic accuracy <strong>of</strong> <strong>CT</strong> as calculated by meta-<strong>analysis</strong> <strong>of</strong> three trials is 71% (95%<br />

CI: 55; 84).<br />

Meta-<strong>analysis</strong> <strong>of</strong> the proportion <strong>of</strong> patients for whom T-staging with the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> or<br />

<strong>CT</strong> was consistent with histopathological examination results is presented in graph 3.<br />

Graph 3.<br />

Odds ratio for the consistency between the reference test and T-staging results, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>.<br />

.<br />

Antoch 2003 5,00 (0,40, 262,30)<br />

Lardinois 2003 5,17 (1,52, 20,08)<br />

Shim 2005 1,59 (0,73, 3,52)<br />

Meta-<strong>analysis</strong> result [fixed] 2,42 (1,36, 4,29)<br />

0,2 0,5 1 2 5 10 100 1000<br />

Odds ratio (95% confidence<br />

interval)<br />

Calculated by meta-<strong>analysis</strong>, the odds ratio is 2.42 (95% CI: 1.36; 4.29). It means that the<br />

odds ratio <strong>of</strong> T-staging consistent with the reference test is 2.42 times higher for <strong>PET</strong>-<strong>CT</strong> than<br />

for <strong>CT</strong>. The result is statistically significant.<br />

In order to obtain one additional case <strong>of</strong> correct T-staging, <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> <strong>CT</strong> scanning<br />

needs to be done with 8 patients with non-small cell lung carcinoma; NNT = 8 (95% CI: 5;<br />

20).<br />

7.4.2. Assessment <strong>of</strong> lymph node involvement (N feature)<br />

In three <strong>of</strong> the trials, the disease was assessed with respect to the presence <strong>of</strong> metastases<br />

to lymph nodes.<br />

In two studies (Antoch 2003 and Lardinois 2003), the disease was staged based on the<br />

TNM lung cancer classification system designed by the AJCC (American Joint Committee<br />

on Cancer). In Shim 2005, metastasis to lymph nodes was assessed and located in 10<br />

groups, according to the definition <strong>of</strong> lymph nodes map for lung cancer staging proposed<br />

109


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

by Mountain and Dresler.<br />

110<br />

The N-status was verified by in the course <strong>of</strong> tumor resection coupled with mediastinal<br />

lymph nodes dissection (Antoch 2003, Lardinois 2003, Shim 2005) or mediastinal endoscopy<br />

(Antoch 2003).<br />

The number true positives (TP), false positives (FP), false negatives (FN) and true<br />

negatives (TN) for <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> scanning is presented in table 24.<br />

Table 24.<br />

Number <strong>of</strong> TP, FP, FN and TN patients in N-staging using <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong>.<br />

Study<br />

Antoch 2003<br />

Shim 2005**<br />

TP<br />

8*<br />

28<br />

FP<br />

1*<br />

58<br />

<strong>PET</strong>-<strong>CT</strong><br />

*Calculated based on data available<br />

**The values reflect the number <strong>of</strong> lymph nodes, not the number <strong>of</strong> patients as for the other trials.<br />

FN<br />

1*<br />

5<br />

In Antoch 2003, the difference between the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in<br />

N-staging is statistically significance in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> (p = 0.004). Lardinois 2003 provides<br />

no information on the statistical significance <strong>of</strong> the differences between the two<br />

diagnostic methods. In Shim 2005, the difference between <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in detecting<br />

malignant tumors in lymph node groups were not statistically significant (p = 0.249).<br />

<strong>PET</strong>-<strong>CT</strong> helped diagnose 23 out <strong>of</strong> 33 lymph node groups with malignant tumor (85%),<br />

and for <strong>CT</strong> the value was 70%. At the same time, the differences in specificity and<br />

accuracy between the two methods were statistically significant (p < 0.001).<br />

Table 25 presents the following diagnostic parameters: sensitivity (Se), specificity (Sp),<br />

accuracy (Acc), positive likelihood ratio (LR+), negative likelihood ratio (LR-), and<br />

diagnostic odds ratio (DOR) calculated for the methods under discussion (<strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong>).<br />

No meta-<strong>analysis</strong> <strong>of</strong> the diagnostic efficacy could be performed as the data presented<br />

by the authors were not sufficient and the results presented differed.<br />

TN<br />

17*<br />

302<br />

TP<br />

7*<br />

23<br />

FP<br />

7*<br />

112<br />

<strong>CT</strong><br />

FN<br />

3*<br />

10<br />

TN<br />

10*<br />

248


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Table 25.<br />

Evaluation <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in N-staging.<br />

Shim 2005<br />

Lardinois 2003<br />

Antoch 2003<br />

Parameter<br />

Statistical<br />

significance<br />

(<strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong>)<br />

<strong>CT</strong><br />

<strong>PET</strong>-<strong>CT</strong><br />

Statistical<br />

significance<br />

(<strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong>)<br />

<strong>CT</strong><br />

<strong>PET</strong>-<strong>CT</strong><br />

Statistical<br />

significance<br />

(<strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong>)<br />

<strong>CT</strong><br />

<strong>PET</strong>-<strong>CT</strong><br />

no data<br />

70%<br />

(51; 84)*<br />

85%<br />

(68; 95)*<br />

no data<br />

no data<br />

no data<br />

no data<br />

70%<br />

(35; 93)*<br />

89%<br />

(52; 100)*<br />

Se<br />

(95% CI)<br />

p < 0,001<br />

69%<br />

(64; 74)*<br />

84%<br />

(80; 88)*<br />

no data<br />

no data<br />

no data<br />

no data<br />

59%<br />

(33; 82)*<br />

94%<br />

(73; 100)*<br />

Sp<br />

(95% CI)<br />

no data<br />

2,24<br />

(1,71; 2,94)*<br />

5,27<br />

(4,00; 6,94)*<br />

no data<br />

no data<br />

no data<br />

no data<br />

1,70<br />

(0,85; 3,42)*<br />

16,00<br />

(2,35; 108,99)*<br />

LR+<br />

(95% CI)<br />

no data<br />

0,44<br />

(0,26; 0,74)*<br />

0,18<br />

(0,08; 0,41)*<br />

no data<br />

no data<br />

no data<br />

no data<br />

0,51<br />

(0,18; 1,42)*<br />

0,12<br />

(0,02; 0,75)*<br />

LR-<br />

(95% CI)<br />

p < 0,001<br />

69%<br />

(64; 74)*<br />

271/393**<br />

84%<br />

(80; 87)*<br />

330/393**<br />

p = 0,12<br />

59%<br />

(42; 75)*<br />

22/37<br />

81%<br />

(65; 92)*<br />

30/37<br />

p = 0,004<br />

63%<br />

(34; 86)*<br />

17/27<br />

93%<br />

(66; 100)*<br />

25/27<br />

Acc<br />

(95% CI)<br />

n/N<br />

no data<br />

5,09<br />

(2,35; 11,06)*<br />

29,16<br />

(10,81; 78,64)*<br />

no data<br />

no data<br />

no data<br />

no data<br />

3,33<br />

(0,63; 17,57)*<br />

136,00<br />

(7,51; 2462,80)*<br />

DOR<br />

(95% CI)<br />

*Calculated based on data available<br />

**The values reflect the number <strong>of</strong> lymph nodes, not the number <strong>of</strong> patients, as in the other studies.<br />

111


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

112<br />

Based on the data above <strong>PET</strong>-<strong>CT</strong> sensitivity is 0.85 and 0.89, and <strong>CT</strong> sensitivity is 0.70, which<br />

means that the probability <strong>of</strong> patients with metastasis to lymph nodes testing positive is 85–<br />

89% for <strong>PET</strong>-<strong>CT</strong> and 70% for <strong>CT</strong>.<br />

The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is within the range 0.84-0.94, so the probability <strong>of</strong> negative results<br />

in patients without metastasis to lymph nodes is 84-94%. The specificity <strong>of</strong> <strong>CT</strong> is 0.59 and 0.69,<br />

which means that the probability <strong>of</strong> negative result is 59-69% for patients who tested<br />

negative for metastasis to lymph nodes in the reference test.<br />

The positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 5.27–16.00, therefore the probability <strong>of</strong> positive<br />

results in patients with metastasis to lymph nodes is 5.27-16 times higher than for patients<br />

without metastasis. The positive result likelihood ratio for <strong>CT</strong> is 1.70-2.24. Therefore the<br />

probability <strong>of</strong> positive results in patients with metastasis to lymph nodes is 1.70-2.24 <strong>of</strong> that<br />

for patients diagnosed negatively for metastasis in histopathological tests.<br />

The negative likelihood ratio for <strong>PET</strong>-<strong>CT</strong> study, is 0.12-0.18, so the probability <strong>of</strong> negative<br />

results in patients with metastasis to lymph nodes is 0.12-0.18 <strong>of</strong> that for patients without<br />

metastasis. The negative likelihood ratio for <strong>CT</strong> is 0.44-0.51. Therefore the probability <strong>of</strong><br />

negative results in patients with metastasis to lymph nodes is 0.44-0.51 <strong>of</strong> that for patients<br />

without metastasis.<br />

Graph 4 illustrates the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in N-staging as calculated by meta-<br />

<strong>analysis</strong> <strong>of</strong> two trials.<br />

Graph 4.<br />

Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostic accuracy in N-staging<br />

Antoch 2003 0,93 (0,76, 0,99)<br />

Lardinois 2003 0,81 (0,65, 0,92)<br />

Meta-<strong>analysis</strong> result 0,85 (0,76, 0,93)<br />

0,6 0,7 0,8 0,9 1,0<br />

Diagnostic accuracy (95% confidence interval)


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

The diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> established by meta-<strong>analysis</strong> is 85% (95% CI: 76; 93).<br />

Graph 5 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>CT</strong> for patients with<br />

non-small cellular lung carcinoma.<br />

Graph 5.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>CT</strong> in N-staging<br />

Antoch 2003 0,63 (0,42, 0,81)<br />

Lardinois 2003 0,59 (0,42, 0,75)<br />

Meta-<strong>analysis</strong> result 0,61 (0,49, 0,72)<br />

0,4 0,6 0,8 1,0<br />

Diagnostic accuracy (95% confidence interval)<br />

The diagnostic accuracy <strong>of</strong> <strong>CT</strong> as calculated by meta-<strong>analysis</strong> <strong>of</strong> two trials is 61% (95%<br />

CI: 49; 72).<br />

The diagnostic odds ratio calculated for <strong>PET</strong>-<strong>CT</strong> is 29.16 and 136.00. The probability <strong>of</strong><br />

positive results is 29-136 times higher for patients with metastasis to lymph nodes than for<br />

patients with no metastasis detected by the reference test. The diagnostic odds ratio for <strong>CT</strong> is<br />

3.33–5.09, so the probability <strong>of</strong> positive results in patients with metastasis detected by the<br />

reference test is 3.33–5.09 <strong>of</strong> the probability for patients without metastasis to lymph nodes.<br />

Graph 6 presents the odds ratio for N-staging consistent with the reference test as<br />

calculated by meta-<strong>analysis</strong> <strong>of</strong> two trials. Shim 2005 was not taken into account in the<br />

meta-<strong>analysis</strong> since its authors provided the number <strong>of</strong> correctly staged lymph nodes, not<br />

the number <strong>of</strong> correctly diagnosed patients, as in the other trials.<br />

113


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Graph 6.<br />

Odds ratio for the consistency <strong>of</strong> N-staging with the reference test, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>.<br />

114<br />

Antoch 2003 7,35 (1,28, 74,58)<br />

Lardinois 2003 2,92 (0,92, 9,86)<br />

Meta-<strong>analysis</strong> result [fixed] 3,95 (1,65, 9,44)<br />

0,5 1 2 5 10 100<br />

Odds ratio (95% confidence<br />

interval)<br />

The odds ratio is 3,95 (95% CI: 1,65; 9,44) so the probability <strong>of</strong> proper assessment <strong>of</strong><br />

lymph node involvement is almost four times higher for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>. The result is<br />

statistically significant.<br />

In order to have one additional case <strong>of</strong> correct N-staging in patients with non-small cell<br />

lung carcinoma it is necessary to use <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> <strong>CT</strong> with 4 patients, NNT = 4 (95% CI: 3;<br />

10).<br />

7.4.3. Assessment <strong>of</strong> distant metastases (M feature)<br />

Cancer staging based on the TNM system with a special attention to the presence <strong>of</strong><br />

distant metastases was executed for two trials included in the <strong>analysis</strong>: Antoch 2003 and<br />

Lardinois 2003.<br />

In Antoch 2003, the presence <strong>of</strong> distant metastases was verified based on clinical follow-<br />

up for two patients, and on biopsy for another two. In Lardinois 2003, the authors verified the<br />

presence <strong>of</strong> extrathoracic metastasis based on biopsy or other imaging techniques.<br />

In Antoch 2003, 17 distant metastases were detected with 4 patients using <strong>PET</strong>-<strong>CT</strong>, while<br />

when <strong>CT</strong> scanning was used only 14 metastases were detected with 4 patients. For both<br />

methods compared, no false positive results were detected. In 1 patient metastasis was<br />

detected 8 months later (false negative result).


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Lardinois 2003 reported only that metastasis to pelvis was identified in 2 patients.<br />

7.4.4. TNM staging system<br />

In all the trials included in this <strong>analysis</strong> (Antoch 2003, Lardinois 2003, Cerfolio 2005, Shim<br />

2005), disease staging was done based on the TNM lung cancer classification system<br />

designed by the AJCC (American Joint Committee on Carcinoma).<br />

Detailed results provided by the authors <strong>of</strong> each study are given in table 26.<br />

115


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

Table 26.<br />

Lung cancer staging results for <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> by trial<br />

Shim 2005<br />

Cerfolio 2005<br />

Antoch 2003<br />

Statistical<br />

significance<br />

<strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong><br />

Disease<br />

stage<br />

Parameter<br />

<strong>CT</strong><br />

n/N (%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

n/N (%)<br />

Statistical<br />

significance<br />

<strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong><br />

<strong>CT</strong><br />

n/N (%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

n/N (%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

n/N (%)<br />

no data no data no data no data<br />

Statistical<br />

<strong>CT</strong><br />

significance<br />

n/N (%) <strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong><br />

no data no data<br />

49%<br />

44%<br />

no data<br />

I<br />

no data no data no data no data<br />

no data no data<br />

46%<br />

34%<br />

no data<br />

II<br />

no data no data no data no data<br />

no data no data<br />

60%<br />

66%<br />

no data<br />

III<br />

Se<br />

no data no data no data no data<br />

no data no data<br />

67%<br />

83%<br />

no data<br />

IV<br />

no data no data no data no data no data no data no data no data<br />

no data<br />

total<br />

no data no data no data no data<br />

no data no data<br />

79%<br />

76%<br />

no data<br />

I<br />

no data no data no data no data<br />

no data no data<br />

93%<br />

92%<br />

no data<br />

II<br />

no data no data no data no data<br />

no data no data<br />

75%<br />

79%<br />

no data<br />

III<br />

Sp<br />

no data no data no data no data<br />

no data no data<br />

92%<br />

93%<br />

no data<br />

IV<br />

no data no data no data no data no data no data no data no data<br />

no data<br />

total<br />

no data<br />

no data<br />

no data no data<br />

47/71 (66%)<br />

63/71 (89%)<br />

66%<br />

68%<br />

no data<br />

I<br />

no data<br />

no data<br />

no data no data<br />

14/ 18 (78%)<br />

17/18 (94%)<br />

82%<br />

84%<br />

no data<br />

II<br />

no data<br />

no data<br />

no data no data<br />

9/17 (53%)<br />

12/17 (71%)<br />

69%<br />

74%<br />

no data no<br />

data<br />

III<br />

Acc<br />

no data no data no data no data<br />

no data no data<br />

92%<br />

93%<br />

no data<br />

IV<br />

p = 0,001<br />

no data no data no data<br />

70/106 (66%)<br />

92/106 (87%)<br />

p = 0,008<br />

19/27 (70%*)<br />

26/27 (96%*)<br />

total<br />

116


<strong>Cost</strong>-<strong>effectiveness</strong> <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> <strong>positron</strong> <strong>emission</strong> tomography In oncological diagnostics<br />

*Value calculated based on data availabl<br />

117


Antoch 2003 notes that the difference in accuracy <strong>of</strong> cancer staging is statistically significant in<br />

favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning (p = 0.008). Lardinois 2003 reports that in the staging <strong>of</strong> the disease,<br />

<strong>PET</strong>-<strong>CT</strong> testing proved more accurate compared with <strong>CT</strong> alone.<br />

118<br />

In Shim 2005, <strong>PET</strong>-<strong>CT</strong> was statistically more accurate in disease staging compared to <strong>CT</strong> (p<br />

= 0,001). Cerfolio 2005 provided no additional data.<br />

Graph 7 shows the diagnostic accuracy <strong>of</strong> <strong>PET</strong> /<strong>CT</strong> in cancer staging as established by<br />

meta-<strong>analysis</strong> <strong>of</strong> two trials.<br />

Graph 7.<br />

Diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in cancer staging (TNM)<br />

Antoch 2003 0,96 (0,81, 1,00)<br />

Shim 2005 0,87 (0,79, 0,93)<br />

Results <strong>of</strong> meta-<strong>analysis</strong> 0,88 (0,83, 0,93)<br />

0,700 0,775 0,850 0,925 1,000<br />

The diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> tests in the staging <strong>of</strong> non-small cellular lung cancer is<br />

88% (95% CI: 83; 93).<br />

Graph 8 illustrates the meta-<strong>analysis</strong> <strong>of</strong> two trials that evaluated the diagnostic accuracy<br />

<strong>of</strong> <strong>CT</strong> in cancer staging.<br />

Diagnostic Accuracy (95% confidence interval)


Graph 8.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>CT</strong> in cancer staging (TNM)<br />

Antoch 2003 0,70 (0,50, 0,86)<br />

Shim 2005 0,66 (0,56, 0,75)<br />

Results <strong>of</strong> meta-<strong>analysis</strong> 0,67 (0,59, 0,74)<br />

The diagnostic accuracy <strong>of</strong> <strong>CT</strong> as calculated by meta-<strong>analysis</strong> is<br />

67% (95% CI: 59; 74).<br />

0,4 0,6 0,8 1,0<br />

Diagnostic accuracy (95% confidence interval)<br />

Graph 9 shows the results <strong>of</strong> meta-<strong>analysis</strong> for the proportion <strong>of</strong> patients in whom the<br />

disease was staged correctly using <strong>PET</strong>-<strong>CT</strong> as compared to <strong>CT</strong> alone.<br />

Graph 9.<br />

Odds ratio for the consistency between the reference test and total cancer staging results, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong><br />

Antoch 2003 10,95 (1,24, 503,96)<br />

Shim 2005 3,38 (1,62, 7,29)<br />

Meta-<strong>analysis</strong> result [fixed] 3,91 (2,04, 7,50)<br />

1 2 5 10 100 1000<br />

Odds ratio (95% confidence<br />

The odds ratio is 3.91 (95% CI: 2,04; 7,50), so the probability <strong>of</strong> correct cancer staging for<br />

119


patients with non-small cell lung carcinoma is 3.91 times higher for <strong>PET</strong>-<strong>CT</strong> scanning than for<br />

<strong>CT</strong>. The difference is statistically significant.<br />

120<br />

In order to obtain one additional case <strong>of</strong> correct staging <strong>PET</strong>-<strong>CT</strong> scanning needs to be<br />

performed instead <strong>of</strong> <strong>CT</strong> for 5 patients with non-small cell lung cancer NNT = 5 (95% CI: 4;<br />

9).<br />

7.4.5. Impact on therapy<br />

Information on the impact <strong>of</strong> the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> standard imaging methods (<strong>CT</strong>)<br />

on the therapeutic procedure is given only in one <strong>of</strong> the trials analyzed (Antoch 2003).<br />

Staging with the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> allowed for correct tumor classification to a higher stage<br />

in 1 out <strong>of</strong> 27 patients (4%), and to a lower stage in 7 patients (26%), which led to new<br />

recommendations for further treatment in case <strong>of</strong> 5 patients (18.5%). For 1 out <strong>of</strong> the 5<br />

patients plans <strong>of</strong> surgery, which had been made based on <strong>CT</strong> scanning, were<br />

abandoned, while 4 patients were scheduled for surgeries.<br />

7.4.6. Safety<br />

None <strong>of</strong> the trials reported on the safety <strong>of</strong> the diagnostic procedures used.<br />

7.5. Results<br />

As a result <strong>of</strong> searching medical databases 4 primary prospective trials were identified<br />

(Antoch 2003, Lardinois 2003, Cerfolio 2005 and Shim 2005), where the <strong>PET</strong>-<strong>CT</strong> method was<br />

compared to <strong>CT</strong> in patients suffering from non-small cell lung cancer (N=570). All the trials<br />

were verified histopathologically. Indications for scanning were based on results <strong>of</strong> T-<br />

staging.<br />

In the assessment <strong>of</strong> T-status in all the trials under assessment, the proportion <strong>of</strong> correctly<br />

diagnosed patients is higher when <strong>PET</strong>-<strong>CT</strong> is adopted in comparison to <strong>CT</strong> only. The diagnostic<br />

accuracy calculated by meta-<strong>analysis</strong> is 86% (95% CI: 81; 91) for <strong>PET</strong>-<strong>CT</strong> and 71% (95% CI: 55;<br />

84) for <strong>CT</strong>.<br />

Calculated by meta-<strong>analysis</strong> <strong>of</strong> three studies, the odds ratio <strong>of</strong> consistency between the<br />

reference test and T-staging results for <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong> is 2.42 (95% CI: 1.36; 4.29). The value is<br />

statistically significant; NNT = 8 (95% CI: 5; 20).<br />

Based on the analyses <strong>of</strong> all the diagnostic parameters established (sensitivity,<br />

specificity, accuracy), <strong>PET</strong>-<strong>CT</strong> is more efficacious than <strong>CT</strong> in rating the involvement <strong>of</strong> lymph<br />

nodes (N-status). The diagnostic accuracy calculated by meta-<strong>analysis</strong> is 85% (95% CI: 76;


93) for <strong>PET</strong>-<strong>CT</strong> and 61% (95% CI: 49; 72) for <strong>CT</strong>. Sensitivity is 85-89%, for <strong>PET</strong>-<strong>CT</strong> and 70% for <strong>CT</strong>,<br />

and specificity is 84-94% for <strong>PET</strong>-<strong>CT</strong> and 59-69% for <strong>CT</strong>.<br />

The odds <strong>of</strong> N-staging results being consistent with the reference test is nearly 4 times<br />

higher for <strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong> alone, and the value is statistically significant: OR=3.95 (95% CI:<br />

1.65; 9.44); NNT = 4 (95% CI: 3; 10).<br />

The accuracy <strong>of</strong> the TNM staging system evaluated by meta-<strong>analysis</strong> is 88% (95% CI: 83; 93)<br />

for <strong>PET</strong>-<strong>CT</strong> and 67% (95% CI: 59; 74) for <strong>CT</strong>.<br />

The odds <strong>of</strong> obtaining small cell lung cancer TNM staging results consistent with the<br />

reference test is higher for <strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong> alone, and the differences observed are<br />

statistically significant; OR = 3.91 (95% CI: 2.04; 7.50); NNT = 5 (95% CI: 4; 9).<br />

In summary, the <strong>PET</strong>-<strong>CT</strong> method has a higher diagnostic efficacy than <strong>CT</strong> in the staging <strong>of</strong><br />

clinical non-small cell lung cancer (NSCLC).<br />

121


8. COMPARATIVE ANALYSIS OF THE EFFICACY OF <strong>PET</strong>-<strong>CT</strong> VS<br />

<strong>CT</strong> IN LYMPHOMA STAGING<br />

122<br />

8.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching medical databases, two primary studies were found<br />

(Freudenberg 2003; tab. 144, app. 18.1 and Schaefer 2004; tab. 145, app. 18.1) that<br />

met the inclusion criteria for this <strong>analysis</strong>, and compared directly <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong><br />

diagnostic efficacy in lymphoma staging.<br />

8.2. Population characteristics<br />

The target population were patients with lymphoma confirmed histologically or<br />

cytologically. Both studies were retrospective.<br />

Freudenberg 2003 included patients who had taken lymphoma therapy for the<br />

purposes <strong>of</strong> restaging.<br />

In Schaefer 2004, the key objective was to evaluate diagnostic efficacy in staging and<br />

restaging. The inclusion criterion in this study was <strong>PET</strong>-<strong>CT</strong> and contrast <strong>CT</strong> scanning <strong>of</strong><br />

patients, assuming time intervals <strong>of</strong> up to 24 days between tests, and assuming no other<br />

treatment was implemented in the meantime.<br />

Table 27 contains the initial characteristics <strong>of</strong> the patients.<br />

Table 27.<br />

Initial characteristics <strong>of</strong> lymphoma patients<br />

Hodgkin<br />

disease<br />

Parameter Freudenberg 2003 Schaefer 2004<br />

Number <strong>of</strong> patients included in study 27 60<br />

Number <strong>of</strong> patients (men/women) 18<br />

Disease stage according to Ann Arbor classification no data<br />

Number <strong>of</strong> patients (men/women) 9<br />

Non-<br />

Hodgkin<br />

lymphoma Initial disease stage according to Ann Arbor classification no data<br />

42<br />

(15/27)<br />

I – 9<br />

II – 24<br />

III – 7<br />

IV – 2<br />

18<br />

(8/10)<br />

I – 8<br />

II – 5<br />

III – 0<br />

IV – 5<br />

Women 11 23<br />

Men 16 37<br />

Age (years)<br />

median age:<br />

46 (17–70)<br />

median age:<br />

39,6±17,1


8.3. Description <strong>of</strong> intervention<br />

8.3.1. <strong>PET</strong>-<strong>CT</strong> imaging<br />

In both studies, imaging diagnostics was executed using a <strong>PET</strong>-<strong>CT</strong> scanner with a hybrid<br />

head. Also for both studies, the radiopharmaceutical used for <strong>PET</strong> images was 18-FDG (18-<br />

fluoro-deoxy-glucose).<br />

In Freudenberg 2003, Biograph scanning system manufactured by Siemens Medical was<br />

used. <strong>PET</strong> scans were done approx. 1 hour after 360 ± 20 MBq 18-FDG was administered. The<br />

tests were interpreted as follows: <strong>PET</strong>-<strong>CT</strong> by two nuclear medicine consultants independently,<br />

and <strong>CT</strong> by two experienced radiologists. The teams were mutually blinded to the results <strong>of</strong><br />

scans they interpreted. Next, combined FDG-<strong>PET</strong> and <strong>CT</strong> images were assessed by one<br />

doctor.<br />

In Schaefer 2004, Discovery LS manufactured by GE Medical Systems (Wukesha, Wis.)<br />

was used. 370 MBq <strong>of</strong> fluoro-deoxy-glucose was administered. The authors provide no<br />

information on the way <strong>PET</strong>-<strong>CT</strong> scans were interpreted.<br />

Table 28 provides a detailed description <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong> tests.<br />

Table 28.<br />

Description <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> imaging for both studies<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong><br />

Scanner<br />

type<br />

Freudenberg 2003 Biograph<br />

Schaefer 2004<br />

Discovery<br />

LS<br />

<strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

manufacturer<br />

Siemens<br />

Medical<br />

Solutions<br />

(H<strong>of</strong>fman<br />

Estates, III)<br />

Ge Medical<br />

Systems<br />

(Milwaukee)<br />

Radiomarker<br />

type<br />

FDG-<strong>PET</strong><br />

intravenously<br />

FDG-<strong>PET</strong><br />

intravenously<br />

8.3.2. Diagnostic technology compared<br />

Scanning<br />

scope<br />

Radiomarker<br />

activity<br />

(MBq)<br />

Whole body 360± 20<br />

intravenously 370<br />

In both studies included, <strong>PET</strong>-<strong>CT</strong> was compared with <strong>CT</strong> with contrast medium.<br />

Spiral <strong>CT</strong> was performed in several hospitals for Schaefer 2004. Contrast medium was<br />

administered intravenously in all tests (approx. 120 to 220 ml contrast medium). The test was<br />

done within 24 days following <strong>PET</strong>-<strong>CT</strong> scanning (9.1±7.0 days on average).<br />

In Freudenberg 2003, a standard protocol was used. Oral or intravenous contrast<br />

medium was used for scanning.<br />

123


124<br />

8.3.3. Reference test<br />

Biopsy <strong>of</strong> lymph node with suspected cancer was the reference (index) test or gold<br />

standard confirming the disease for lymphoma; for cancers located outside nodes the<br />

reference test was biopsy <strong>of</strong> the organ with suspected lesions.<br />

Not all patients were tested with the reference test in both studies. In addition to<br />

biopsy and clinical observation, the authors mention the following as reference tests:<br />

• conventional disease staging by clinical examination, laboratory diagnostic<br />

tests, USG, MRI, scintigraphy,<br />

• clinical observation.<br />

A list <strong>of</strong> reference methods used is given in table 29.<br />

Table 29.<br />

List <strong>of</strong> diagnosis reference tests<br />

Study<br />

Reference test<br />

8.4. Findings<br />

Freudenberg 2003<br />

1. Biopsy: in 7 patients<br />

2. Other imaging studies: 8/27<br />

patients<br />

3. Clinical observation<br />

4. USG: 23/27<br />

5. <strong>PET</strong><br />

8.4.1. Diagnosis efficacy in disease staging<br />

Schaefer 2004<br />

1. Biopsy; no data on patients with biopsy<br />

performed<br />

2. Clinical observation<br />

3. Other diagnosis methods, such as MRI,<br />

scintigraphy, laboratory tests<br />

Disease staging based on the Ann Arbor classification was done for all tests included<br />

in this <strong>analysis</strong>.<br />

In Schaefer 2004, the efficacy in detecting lymphomas in and outside <strong>of</strong> lymph nodes<br />

was evaluated.<br />

8.4.1.1 In nodes<br />

Study results concerning the diagnostic efficacy in detecting cancer in node locations<br />

is given in table 30.


Table 30.<br />

Test diagnostic efficacy in detecting cancer in node locations<br />

Results /<br />

parameters<br />

TP true positive<br />

dodatni<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

Staging Restaging Staging Restaging<br />

19 11 19 9<br />

FP false positive 0 0 0 4<br />

TN true negative 0 28 0 24<br />

FN false negative 0 2 0 4<br />

SE sensitivity<br />

100%<br />

(82; 100)<br />

SP specificity -<br />

LR+ -<br />

LR- -<br />

ACC<br />

100%<br />

(82; 100)<br />

DOR -<br />

85%<br />

(55; 98)<br />

100%<br />

(88; 100)<br />

47,64<br />

(3,02; 751,73)<br />

0,18<br />

(0,06; 0,56)<br />

95%<br />

(83; 99)<br />

262,20<br />

(11,66; 5 894,2)<br />

100%<br />

(82; 100)<br />

-<br />

-<br />

-<br />

100%<br />

(82; 100)<br />

In case <strong>of</strong> staging, both sensitivity and accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> were 100%.<br />

-<br />

69%<br />

(39; 91)<br />

86%<br />

(67; 96)<br />

4,85<br />

(1,82; 12,87)<br />

0,36<br />

(0,16; 0,82)<br />

80%<br />

(65; 91)<br />

13,50<br />

(2,77; 65,78)<br />

In restaging, <strong>PET</strong>-<strong>CT</strong> sensitivity was 85% (95% CI: 55; 98), representing 11 out <strong>of</strong> 13<br />

patients; <strong>PET</strong>-<strong>CT</strong> specificity was 100% (95% CI: 88; 100) representing 28 out <strong>of</strong> 28 patients.<br />

Both these parameters had higher values than with <strong>CT</strong>, where sensitivity was 69% (95% CI:<br />

39; 91), and specificity was 86% (95% CI: 67; 96).<br />

In restaging, test accuracy was 95% (95% CI: 83; 99) for <strong>PET</strong>-<strong>CT</strong>, and 80% (95% CI: 65; 91)<br />

for <strong>CT</strong>.<br />

125


126<br />

8.4.1.2 Locations outside <strong>of</strong> nodes<br />

Table 31.<br />

Test diagnostic efficacy in detecting cancer locations outside <strong>of</strong> nodes<br />

Results /<br />

parameters<br />

TP true<br />

positive<br />

FP false<br />

negative<br />

TN true<br />

negative<br />

FN false<br />

negative<br />

SE sensitivity<br />

SP specificity<br />

LR+<br />

LR-<br />

ACC<br />

DOR<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

Staging Restaging Staging Restaging<br />

3 4 1 3<br />

0 0 0 5<br />

15 37 15 32<br />

1 0 3 1<br />

75%<br />

(19; 99)<br />

100%<br />

(78; 100)<br />

22,40<br />

(95% CI: 1,38;<br />

363,91)<br />

0,31<br />

(95% CI: 0,08;<br />

1,19)<br />

95%<br />

(95% CI: 74; 100)<br />

72,33<br />

(95% CI: 2,40;<br />

2176,8)<br />

100%<br />

(40; 100)<br />

100%<br />

(91; 100)<br />

68,40<br />

(95% CI: 4,29; 1090,5)<br />

0,10<br />

(95% CI: 0,01; 1,41)<br />

100%<br />

(95% CI: 90; 100)<br />

675,0<br />

(95% CI: 11,88;<br />

38359,8)<br />

25%<br />

(1; 81)<br />

100%<br />

(78; 100)<br />

9,60<br />

(95% CI: 0,46; 200,49)<br />

0,72<br />

(95% CI: 0,40; 1,29)<br />

84%<br />

(95% CI: 60; 97)<br />

13,29<br />

(95% CI: 0,44; 399,82)<br />

75%<br />

(19; 99)<br />

86%<br />

(71; 95)<br />

5,55<br />

(95% CI: 2,06; 14,97)<br />

0,29<br />

(95% CI: 0,05; 1,59)<br />

85%<br />

(95% CI: 71; 94)<br />

19,20<br />

(95% CI: 1,65; 222,85)<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity and specificity <strong>of</strong> lesion detection in location outside <strong>of</strong> nodes 75% (3<br />

out <strong>of</strong> 4 patients) and 100% (15 out <strong>of</strong> 15 patients) respectively for staging, and 100% (4 out<br />

<strong>of</strong> 4 patients) and 100% (37 out <strong>of</strong> 37 patients) for restaging. For <strong>CT</strong> the results were: 25%<br />

sensitivity (1 out <strong>of</strong> 4 patients) and 100% specificity (15 out <strong>of</strong> 15 patients) for staging, and<br />

75% sensitivity (3 out <strong>of</strong> 4 patients) and 86% specificity (32 out <strong>of</strong> 37 patients) for restaging.<br />

For both staging and restaging, <strong>PET</strong>-<strong>CT</strong> demonstrated higher accuracy (95% for staging<br />

and 100% for restaging). The accuracy <strong>of</strong> <strong>CT</strong> was 84% for staging and 85% for restaging.<br />

McNemar’s test executed for staging and restaging showed statistically significant<br />

difference in specificity, in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> (p = 0.004), and no significant differences were<br />

found for sensitivity (p = 0.11).<br />

8.4.1.3 Total results<br />

Total results for the staging and restaging <strong>of</strong> disease in lymphatic and extra-lymphatic,<br />

locations in Schaefer 2004 and Freudenberg 2003, are given in table 32.


Table 32.<br />

Diagnostic efficacy in lymphomas detection in locations in and outside <strong>of</strong> nodes.<br />

Study<br />

Schaefer 2004<br />

Freudenberg<br />

2003<br />

TP<br />

37<br />

13<br />

8.4.1.3.1 Sensitivity<br />

FP<br />

0<br />

0<br />

<strong>PET</strong>-<strong>CT</strong><br />

Graph 10 illustrates <strong>PET</strong>-<strong>CT</strong> sensitivity calculated by meta-<strong>analysis</strong>.<br />

Graph 10.<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity<br />

Schaeffer 2004<br />

Meta-<strong>analysis</strong> results<br />

TN<br />

80<br />

13<br />

<strong>PET</strong>-<strong>CT</strong> scanning sensitivity was 93% (95% CI: 80; 98) in Schaefer 2004, and 93% (95% CI:<br />

66; 100) in Freudenberg 2003. Total sensitivity for <strong>PET</strong>-<strong>CT</strong> as calculated by meta-<strong>analysis</strong> was<br />

93% (95% CI: 82; 98).<br />

0,5 1<br />

FN<br />

Sensitivity (95% confidence interval)<br />

<strong>CT</strong> imaging sensitivity as calculated by meta-<strong>analysis</strong> is presented in graph 11.<br />

3<br />

1<br />

TP<br />

32<br />

11<br />

FP<br />

9<br />

6<br />

<strong>CT</strong><br />

TN<br />

71<br />

7<br />

0,93 (0,80, 0,98)<br />

Freudenberg 2003 0,93 (0,66, 1,00)<br />

0,93 (0,82, 0,98)<br />

FN<br />

8<br />

3<br />

127


Graph 11.<br />

<strong>CT</strong> sensitivity<br />

128<br />

Schaeffer 2004<br />

Freudenberg 2003 0,79 (0,49, 0,95)<br />

Meta-<strong>analysis</strong> results 0,80 (0,66, 0,89)<br />

0,2 0,5 1<br />

<strong>CT</strong> imaging sensitivity was 80% (95% CI: 64; 91) in Schaefer 2004, and 79% (95% CI: 49; 95)<br />

in Freudenberg 2003. Total sensitivity for <strong>CT</strong> as calculated by meta-<strong>analysis</strong> was 80% (95%<br />

CI: 66; 89). Heterogeneity was not affirmed.<br />

8.4.1.3.2 Specificity<br />

Sensitivity (95% confidence<br />

interval)<br />

<strong>PET</strong>-<strong>CT</strong> specificity as calculated by meta-<strong>analysis</strong> is presented in graph 12.<br />

0,80 (0,64, 0,91)


Graph 12.<br />

<strong>PET</strong>-<strong>CT</strong> specificity<br />

Schaeffer 2004 1,00 (0,95, 1,00)<br />

Freudenberg 2003<br />

Meta-<strong>analysis</strong> result 1,00 (0,96, 1,00)<br />

0,5 1<br />

Specificity (95% confidence interval)<br />

<strong>PET</strong>-<strong>CT</strong> specificity was 100% (95% CI: 95; 100) in Schaefer 2004, and 100% (95% CI: 75; 100)<br />

in Freudenberg 2003. <strong>PET</strong>-<strong>CT</strong> specificity as calculated by meta-<strong>analysis</strong> was 100% (95% CI: 96;<br />

100). No heterogeneity was found in the meta-<strong>analysis</strong>.<br />

<strong>CT</strong> sensitivity as calculated by meta-<strong>analysis</strong> is presented in graph 13.<br />

1,00 (0,75, 1,00)<br />

129


Graph 13.<br />

<strong>CT</strong> specificity<br />

130<br />

Schaeffer 2004 0,89 (0,80, 0,95)<br />

Freudenberg 2003<br />

Meta-<strong>analysis</strong> result<br />

0,0 0,2 0,4 0,6 1,0<br />

<strong>CT</strong> specificity was 89% (95% CI: 80; 95) in Schaefer 2004, and 54% (95% CI: 25; 81) in<br />

Freudenberg 2003. Total <strong>CT</strong> specificity as calculated by meta-<strong>analysis</strong> was 84% (95% CI: 75;<br />

91). Significant heterogeneity <strong>of</strong> the results was observed (p = 0,0048).<br />

8.4.1.3.3 Positive likelihood ratio<br />

Specificity (95% confidence interval)<br />

The positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> scanning is presented in graph 14.<br />

0,54 (0,25, 0,81)<br />

0,84 (0,75, 0,91)


Graph 14.<br />

Positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> scanning<br />

Schaeffer 2004 148,17 (9,33, 2352,41)<br />

Freudenberg 2003 25,20 (1,65, 385,27)<br />

Result <strong>of</strong> meta-<strong>analysis</strong> 60,37 (8,66, 420,73)<br />

The positive likelihood ratio (LR+) as calculated by meta-<strong>analysis</strong> was 60.37 (95% CI:<br />

8.66; 420.73), which means that the probability <strong>of</strong> positive results with <strong>PET</strong>-<strong>CT</strong> scanning is<br />

almost 60 times higher for patients with lesions in and outside <strong>of</strong> lymph nodes than for<br />

patients without such lesions.<br />

1 2 5 10 100 1000 1,00E+05<br />

Positive likelihood ratio (95%<br />

confidence interval)<br />

The positive likelihood ratio for <strong>CT</strong> study is presented in graph 15.<br />

131


Graph 15.<br />

Positive likelihood ratio for <strong>CT</strong> study.<br />

132<br />

Schaefer 2004 7,11 (3,77, 13,41)<br />

Freudenberg 2003 1,70 (0,89, 3,25)<br />

Meta-<strong>analysis</strong> result 3,48 (0,81, 15,01)<br />

The positive likelihood ratio (LR+) as calculated by meta-<strong>analysis</strong> was 3,48 (95% CI:<br />

0,81; 15,01), which means that the probability <strong>of</strong> positive results with <strong>CT</strong> scanning <strong>of</strong> patients<br />

with lesions in and outside <strong>of</strong> lymph nodes was 348% <strong>of</strong> the probability for patients without<br />

such lesions.<br />

0,5 1 2 5 10 100<br />

8.4.1.3.4 Negative likelihood ratio<br />

Positive result likelihood ratio (95% confidence)<br />

Graph 16 illustrates the negative likelihood ratio for <strong>PET</strong>-<strong>CT</strong>.


Graph 16.<br />

Negative likelihood ratio for <strong>PET</strong>-<strong>CT</strong> study<br />

Schaefer 2004<br />

Freudenberg 2003<br />

Meta-<strong>analysis</strong> result 0,09 (0,04, 0,21)<br />

The negative likelihood ratio (LR-) as calculated by meta-<strong>analysis</strong> was 0,09 (95% CI:<br />

0,04; 0,21), which means that the probability <strong>of</strong> negative results with <strong>PET</strong>-<strong>CT</strong> scanning <strong>of</strong><br />

patients with lesions in and outside <strong>of</strong> lymph nodes was 9% <strong>of</strong> the probability for patients<br />

without such lesions.<br />

0,01 0,1 0,2 0,5<br />

Negative likelihood ratio (95% confidence<br />

Graph 17 illustrates the negative likelihood ratio for <strong>CT</strong>.<br />

0,09 (0,03, 0,23)<br />

0,10 (0,02, 0,47)<br />

133


Graph 17.<br />

Negative likelihood ratio for <strong>CT</strong> study<br />

134<br />

Schaefer 2004<br />

Freudenberg 2003<br />

Meta-<strong>analysis</strong> result<br />

The negative likelihood ratio (LR-) as calculated by meta-<strong>analysis</strong> is 0.26 (95% CI: 0,15;<br />

0,44), which means that the probability <strong>of</strong> negative results with <strong>CT</strong> scanning for patients with<br />

lesions in and outside <strong>of</strong> lymph nodes is 26% <strong>of</strong> the probability for patients without such<br />

lesions.<br />

0,1 0,2 0,5 1 2<br />

8.4.1.3.5 Diagnostic odds ratio<br />

Negative likelihood ratio (95% confidence<br />

Graph 18 illustrates the diagnostic odds ratio for <strong>PET</strong>-<strong>CT</strong>.<br />

0,23 (0,12, 0,42)<br />

0,40 (0,13, 1,22)<br />

0,26 (0,15, 0,44)


Graph 18.<br />

Diagnostic odds ratio for <strong>PET</strong>-<strong>CT</strong><br />

Schaefer 2004 1725,00 (86,88, 34248,86)<br />

Freudenberg 2003 234,00 (9,07, 6511,88)<br />

Meta-<strong>analysis</strong> result 710,84 (77,86, 6490,00)<br />

5 10 100 1000 1,00E+05<br />

Diagnostic odds ratio (95% confidence interval)<br />

The odds ratio as calculated by meta-<strong>analysis</strong> is 710.84 (95% CI: 77,86; 6490,00), which<br />

means that the probability <strong>of</strong> positive results with <strong>CT</strong> scanning <strong>of</strong> patients with lesions in or<br />

outside <strong>of</strong> lymph nodes is approx. 710 times higher than for patients without such lesions.<br />

Graph 19 illustrates the diagnostic odds ratio for <strong>CT</strong>.<br />

135


Graph 19.<br />

Diagnostic odds ratio for <strong>CT</strong><br />

136<br />

Schaefer 2004 31,56 (11,16, 89,26)<br />

Freudenberg 2003 4,28 (0,80, 22,93)<br />

Meta+analzsis result 13,01 (1,85, 91,34)<br />

0,5 1 2 5 10 100<br />

The odds ratio as calculated by meta-<strong>analysis</strong> is 13.01 (95% CI: 1,85; 91,34), which<br />

means that the probability <strong>of</strong> positive results with <strong>CT</strong> scanning <strong>of</strong> patients with lesions in and<br />

outside <strong>of</strong> lymph nodes is more than 13 times higher than for patients without such lesions.<br />

8.4.1.3.6 Diagnostic accuracy<br />

Diagnostic odds ratio (95% confidence interval)<br />

Graph 20 illustrates the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning.


Graph 20.<br />

<strong>PET</strong>-<strong>CT</strong> accuracy<br />

Schaefer 2004<br />

Freudenberg 2003<br />

Meta-<strong>analysis</strong> result<br />

<strong>PET</strong>-<strong>CT</strong> accuracy as calculated by meta-<strong>analysis</strong> is 97% (95% CI: 93; 99).<br />

Graph 21 illustrates the diagnostic accuracy <strong>of</strong> <strong>CT</strong> scanning.<br />

Graph 21.<br />

<strong>CT</strong> accuracy<br />

Schaefer 2004<br />

Meta-<strong>analysis</strong> result<br />

0,5 1<br />

Meta-<strong>analysis</strong> <strong>of</strong> accuracy (95% confidence interval<br />

0,2 0,5 1<br />

Accuracy meta-<strong>analysis</strong> (95% confidence interval)<br />

<strong>CT</strong> accuracy as calculated by meta-<strong>analysis</strong> is 78% (95% CI: 57; 93).<br />

0,98 (0,93, 0,99)<br />

0,96 (0,81, 1,00)<br />

0,97 (0,93, 0,99)<br />

0,86 (0,78, 0,92)<br />

Freudenberg 2003 0,67 (0,46, 0,83)<br />

0,78 (0,57, 0,93)<br />

137


138<br />

8.4.1.3.7 Results<br />

Table 33.<br />

Diagnostic accuracy in detecting lymphomas<br />

Se[%]<br />

Sp[%]<br />

LR-<br />

LR+<br />

DOR<br />

Acc<br />

<strong>PET</strong>-<strong>CT</strong><br />

93%<br />

(95% CI: 80; 98)<br />

100%<br />

(95% CI: 95;<br />

100)<br />

0,09<br />

(95% CI: 0,03;<br />

0,23)<br />

148,17<br />

(95% CI: 9,33;<br />

2352,41)<br />

1725,00<br />

(95% CI: 86,88;<br />

34248,86)<br />

98%<br />

(95% CI: 93; 99)<br />

Schaefer 2004<br />

<strong>CT</strong><br />

80%<br />

(95% CI: 64; 91)<br />

89%<br />

(95% CI: 80; 95)<br />

0,23<br />

(95% CI: 0,12;<br />

0,42)<br />

7,11<br />

(95% CI: 3,77;<br />

13,41)<br />

31,56<br />

(95% CI: 11,16;<br />

89,26)<br />

86%<br />

(95% CI: 78; 92)<br />

Freudenberg 2003<br />

<strong>PET</strong>-<strong>CT</strong><br />

93%<br />

(95% CI: 66;<br />

100)<br />

100%<br />

(95% CI: 75;<br />

100)<br />

0,10<br />

(95% CI: 0,02;<br />

0,47)<br />

25,20<br />

(95% CI: 1,65;<br />

385,27)<br />

234,00<br />

(95% CI: 9,07;<br />

6511,88)<br />

96%<br />

(95% CI: 81;<br />

100)<br />

<strong>CT</strong><br />

79%<br />

(95% CI: 49; 95)<br />

54%<br />

(95% CI: 25; 81)<br />

0,40<br />

(95% CI: 0,13;<br />

1,22)<br />

1,70<br />

(95% CI: 0,89;<br />

3,25)<br />

4,28<br />

(95% CI: 0,80;<br />

22,93)<br />

67%<br />

(95% CI: 46; 83)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Total result<br />

93%<br />

(95% CI: 82; 98)<br />

100%<br />

(95% CI: 96;<br />

100)<br />

0,09<br />

(95% CI: 0,04;<br />

0,21)<br />

60,37<br />

(95% CI: 8,66;<br />

420,73)<br />

710,84<br />

(95% CI: 77,86;<br />

6490,0)<br />

97%<br />

(95% CI: 93; 99)<br />

8.4.2. Accuracy <strong>of</strong> staging based on Ann Arbor staging system<br />

Freudenberg 2003 compared the usefulness <strong>of</strong> diagnostic tests in disease staging<br />

according to the Ann Arbor staging system.<br />

<strong>CT</strong><br />

80%<br />

(95% CI: 66; 89)<br />

84%<br />

(95% CI: 75; 91)<br />

0,26<br />

(95% CI: 0,15;<br />

0,44)<br />

3,48<br />

(95% CI: 0,81;<br />

15,01)<br />

13.01<br />

(95% CI: 1,85;<br />

91,34)<br />

78%<br />

(95% CI: 57; 93)<br />

In Freudenberg 2003, staging based on <strong>CT</strong> scans was correct for 13 patients (48%), and<br />

staging based on <strong>PET</strong>-<strong>CT</strong>scans was correct for 26 patients (96%). The difference between<br />

<strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> is statistically significant (p = 0.002).<br />

<strong>PET</strong>-<strong>CT</strong> scanning allowed to classify correctly disease at a higher stage than identified by<br />

<strong>CT</strong> in 6 patients (26%), and to classify correctly disease at a lower stage in 7 patients (26%).<br />

Table 34 provides results <strong>of</strong> lymphoma staging using <strong>PET</strong>-<strong>CT</strong> as compared to <strong>CT</strong> in<br />

Freudenberg 2003.<br />

Table 34.<br />

Results <strong>of</strong> lymphoma staging by <strong>PET</strong>-<strong>CT</strong> as compared to <strong>CT</strong>.<br />

Freudenberg 2003<br />

Correct staging by <strong>PET</strong>-<strong>CT</strong> Correct staging by <strong>CT</strong><br />

96%<br />

(95% CI: 81; 100)<br />

48%<br />

(95% CI: 29; 68)<br />

Statistical<br />

significance<br />

The odds ratio for correct staging <strong>of</strong> lymphomas based on the Ann Arbor system for <strong>PET</strong>-<br />

<strong>CT</strong> vs. <strong>CT</strong>.<br />

0,002


Table 35.<br />

Odds ratio for correct lymphoma staging: <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong><br />

<strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong><br />

OR NNT<br />

28,00<br />

(95% CI: 3,35; 1227,94)<br />

3<br />

(95% CI: 2; 4)<br />

The odds ratio was calculated at 28.00 (95% CI: 3.35; 1227.94), which means that the<br />

probability <strong>of</strong> correct lymphoma staging according to the Ann Arbor system is 28 times<br />

higher for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong> imaging. The difference is statistically significant.<br />

In order to obtain one additional case <strong>of</strong> correct lymphoma staging it is necessary to<br />

use <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> <strong>CT</strong> with 3 patients with esophageal cancer, NNT = 3 (95% CI: 2; 4).<br />

8.4.3. Safety<br />

The authors identified no undesirable effects <strong>of</strong> the use <strong>of</strong> the diagnostic methods<br />

described.<br />

8.5. Results<br />

As a result <strong>of</strong> searching medical databases two primary studies were found: Freudenberg 2003<br />

(N = 27) and Schaefer 2004 (N = 60), which compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

and <strong>CT</strong> in lymphoma staging and restaging. The reference tests were histopathological tests,<br />

clinical follow-up and conventional staging <strong>of</strong> the disease (clinical and laboratory tests, USG, MRI,<br />

scintigraphy).<br />

A diagnostic efficacy review showed that <strong>PET</strong>-<strong>CT</strong> was diagnostically more efficacious than <strong>CT</strong><br />

both for lymphical and extralymphical locations individually, and for the total cohort.<br />

In staging lesions in the lymphical area, the sensitivity and accuracy <strong>of</strong> both <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> were<br />

100% (95% CI: 82; 100).<br />

In restaging, <strong>PET</strong>-<strong>CT</strong> sensitivity stood at 85% (95% CI: 55; 98) and specificity at 100% (95% CI: 88;<br />

100); both were higher than the respective parameters for <strong>CT</strong>. For <strong>CT</strong> the values were: 69% for<br />

sensitivity (95% CI: 39; 91) and 86% for specificity (95% CI: 67; 96). The diagnostic accuracy <strong>of</strong> the<br />

methods discussed was 95% (95% CI: 83; 99) for <strong>PET</strong>-<strong>CT</strong> and 80% (95% CI: 65; 91) for <strong>CT</strong>.<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity and specificity in detecting pathological lesions located outside <strong>of</strong> lymph<br />

nodes stood at 75% (95% CI: 19; 99) and 100% (95% CI: 78; 100), respectively. In restaging, both<br />

sensitivity and specificity were 100%. With <strong>CT</strong> imaging, the values were: 25% for sensitivity (95% CI: 1;<br />

81) and 100% for specificity (95% CI: 78; 100) for staging, and 75% (95% CI: 19; 99) and 86% (95% CI:<br />

71; 95) respectively for restaging. <strong>PET</strong>-<strong>CT</strong> scanning demonstrated higher accuracy (95% for staging<br />

and 100% for restaging) compared to <strong>CT</strong> (84% and 85% respectively).<br />

Meta-<strong>analysis</strong> <strong>of</strong> the tests included in this study showed that in disease staging in patients with<br />

139


lymphoma, <strong>PET</strong>-<strong>CT</strong> is characterized by higher sensitivity and specificity compared to <strong>CT</strong>. The<br />

sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 93% (95% CI: 82; 98), vs. 80% for <strong>CT</strong> (95% CI: 66; 89). The specificity <strong>of</strong> the<br />

diagnostic methods discussed is 100% (95% CI: 96; 100) and 84% (95% CI: 75; 91) respectively.<br />

140<br />

<strong>PET</strong>-<strong>CT</strong> is characterizes by a higher accuracy <strong>of</strong> 97% (95% CI: 93; 99) s. 78% for <strong>CT</strong> (95% CI: 57; 93).<br />

Freudenberg 2003 analyses the consistency between the reference test and staging based on<br />

the Ann Arbor system. Disease was staged correctly by <strong>PET</strong>-<strong>CT</strong> in 26 (96%) patients, and by <strong>CT</strong> in 13<br />

(48%) patients. The difference between <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> was statistically significant (p=0.002). The<br />

odds <strong>of</strong> obtaining the correct staging were 28 times for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>: OR = 28 (95% CI: 3.35;<br />

1227.94); NNT = 3 (95% CI: 2; 4).<br />

<strong>PET</strong>-<strong>CT</strong> imaging is characterized by higher diagnostic efficacy than <strong>CT</strong> in the staging <strong>of</strong><br />

lymphomas.


9. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS. <strong>CT</strong> IN<br />

ESOPHAGEAL CANCER STAGING<br />

9.1. Disease staging<br />

9.1.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching through medical databases, one primary study was found<br />

(Cerfolio 2005; tab. 146, app. 18.1), that met the inclusion criteria and compared<br />

directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with that <strong>of</strong> other technologies used in<br />

cancer restaging.<br />

9.1.2. Population characteristics<br />

The aim <strong>of</strong> Cerfolio 2005 was a prospective assessment <strong>of</strong> the accuracy <strong>of</strong> three<br />

testing technologies in the restaging <strong>of</strong> esophageal cancer following neoadjuvant<br />

therapy. The studies evaluated were: <strong>PET</strong>-<strong>CT</strong>, transesophageal endoscopic<br />

ultrasound-guided biopsy (EUS) and <strong>CT</strong>.<br />

Only patients for whom scans had been done using all the three technologies<br />

mentioned above prior to surgery were included in the study. Following preliminary<br />

staging, all the patients took neoadjuvant chemotherapy with concomitant<br />

radiotherapy. After the treatment, total restaging was done for these patients (TNM).<br />

The results were entered into a database by a blinded researcher.<br />

Initially, 67 patients were included in the study. Four out <strong>of</strong> that group refused to<br />

take part in the trial. Following neoadjuvant therapy, disease progression occurred<br />

but still they refused to take biopsy. Another 2 patients could take <strong>PET</strong>-<strong>CT</strong> tests<br />

(authors did not provide reasons). The serious health condition <strong>of</strong> another four<br />

patients prevented the procedure or they refused to take the procedure. Ultimately,<br />

48 patients were included in the study. 41 had undergone Ivor Lewis<br />

esophagogastrectomy (resection <strong>of</strong> stomach and esophagus) with<br />

lymphadenectomy.<br />

The characteristics <strong>of</strong> the patients included is presented in table 36.<br />

141


Table 36.<br />

Patient characteristics in Cerfolio 2005<br />

142<br />

Parameter Cerfolio 2005<br />

Number <strong>of</strong> patients included in study 48<br />

Disease staging based on<br />

TNM system<br />

Before therapy After therappy<br />

0 - 15 (31%)<br />

I 0 5 (10%)<br />

IIa 22 (46%) 10 (22%)<br />

IIb 5 (10%) 4 (8%)<br />

III 15 (33%) 2 (4%)<br />

T4 2 (4%) 1 (2%)<br />

IV (M1a) 3 (6%) 5 (10%)<br />

IV (M1b) 1 (2%) 6 (13%)<br />

Women 7 (15%)<br />

Men 41 (85%)<br />

Median age [years] 68 (48-76)<br />

Histopathological examination results<br />

9.1.3. Description <strong>of</strong> intervention<br />

9.1.3.1 <strong>PET</strong>-<strong>CT</strong> imaging<br />

Adenocarcinoma: 43 patients<br />

(85%)<br />

Squamous cell carcinoma: 5 patients<br />

(15%)<br />

In the study, imaging diagnostics was executed using a <strong>PET</strong>-<strong>CT</strong> scanner with a hybrid<br />

head. The radiopharmaceutical used for <strong>PET</strong> was FFDG (F-18 fluoro-deoxy-glucose)<br />

administered intravenously in 555 MBq doses. <strong>PET</strong>-<strong>CT</strong> image was obtained using a GE<br />

Discovery LS <strong>PET</strong>-<strong>CT</strong> Scanner (GE, Milwaukee, Wis). The researcher interpreting the test had<br />

access to <strong>CT</strong> images but did not know EUS-FNA results.<br />

Table 37.<br />

<strong>PET</strong>-<strong>CT</strong> imaging Cerfolio 2005<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong> scanner<br />

type<br />

Cerfolio 2005 Ge Discovery LS<br />

<strong>PET</strong>-<strong>CT</strong> scanner<br />

manufacturer<br />

Ge Medical<br />

Systems<br />

(Milwaukee)<br />

Radiomarker<br />

type<br />

FDG<br />

intravenously<br />

Radiomarker<br />

activity<br />

(MBq)<br />

555<br />

Test<br />

coverage<br />

Thorax,<br />

abdomen,<br />

pelvis


9.1.3.2 Diagnostic technology compared<br />

1. The diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was compared with <strong>CT</strong> and transesophageal<br />

endoscopic ultrasound-guided biopsy.<br />

2. Transesophageal endoscopic ultrasound-guided biopsy was done under anesthetic<br />

and assessed by an experienced ultrasonography specialist, who knew no <strong>CT</strong> or <strong>PET</strong>-<strong>CT</strong><br />

results. The scanning involved the visualization <strong>of</strong> internal organs <strong>of</strong> the abdominal cavity with<br />

the aim <strong>of</strong> identifying the tumor location and the involvement <strong>of</strong> adjacent lymph nodes. The<br />

test was initially done using a GF-UM130 unit, and when lesions were located, an Olympus<br />

UC-30P or VTC 140 endoscope was used for fine-needle biopsy. If lymph nodes were<br />

involved, no biopsy was done for fear <strong>of</strong> aspiration <strong>of</strong> tumor material. Where sampling <strong>of</strong><br />

regional lymph nodes was impossible, an experienced ultrasonography specialist assessed<br />

lymph nodes involvement as positive or negative.<br />

<strong>CT</strong> had to be done with the use <strong>of</strong> a third generation unit as minimum requirement.<br />

Imaging covered the thorax, abdominal cavity and pelvis. Images were assessed by one <strong>of</strong><br />

four radiologists experienced in assessing thorax organs.<br />

9.1.3.3 Reference test<br />

Histopathological examination was used to confirm the presence <strong>of</strong> esophageal cancer.<br />

41 patients in Cerfolio 2005 underwent stomach and esophagus resection using the Ivor<br />

Lewis method (esophagogastrectomy) with lymphadenectomy, followed by<br />

histopathological examination.<br />

9.1.4. Findings<br />

9.1.4.1 Diagnostic efficacy <strong>of</strong> test<br />

Cerfolio 2005 evaluated the diagnostic efficacy <strong>of</strong> tests based on pre-defined end points.<br />

Test efficacy in:<br />

Disease staging:<br />

o T feature (tumor size),<br />

o N feature (involvement <strong>of</strong> lymph nodes),<br />

o M feature (presence <strong>of</strong> distant metastases),<br />

Assessing total response to therapy.<br />

9.1.4.1.1 T-staging<br />

The accuracy <strong>of</strong> each diagnostic test was assessed based on its ability to differentiate<br />

between stages T1-T3, and T0 and T4 (<strong>PET</strong>-<strong>CT</strong> does not differentiate between stages T1, T2<br />

143


144<br />

and T3).<br />

Table 38 presents staging results for the diagnostic technologies as compared to the<br />

reference method.<br />

Table 38.<br />

T-staging results for each <strong>of</strong> the diagnostic methods as compared to the reference test (Cerfolio 2005)<br />

T0<br />

T1–T3<br />

T4<br />

TP FP FN TN<br />

<strong>PET</strong>-<strong>CT</strong> 13 4 3 21<br />

EUS 3 1 13 24<br />

<strong>CT</strong> 4 4 12 21<br />

<strong>PET</strong>-<strong>CT</strong> 18 4 4 15<br />

EUS 20 16 2 3<br />

<strong>CT</strong> 18 14 4 5<br />

<strong>PET</strong>-<strong>CT</strong> 2 0 1 38<br />

EUS 0 1 3 37<br />

<strong>CT</strong> 1 0 2 38<br />

Table 39 presents the diagnostic efficacy parameters by disease stage.<br />

Table 39.<br />

<strong>PET</strong>-<strong>CT</strong>, EUS-FNA and <strong>CT</strong> diagnostic efficacy in tumor restaging<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

LR+<br />

(95% CI)<br />

<strong>PET</strong>-<strong>CT</strong><br />

EUS<br />

<strong>CT</strong><br />

<strong>PET</strong>-<strong>CT</strong><br />

EUS<br />

<strong>CT</strong><br />

<strong>PET</strong>-<strong>CT</strong><br />

EUS<br />

<strong>CT</strong><br />

<strong>PET</strong>-<strong>CT</strong><br />

T0 T1–T3 T4<br />

81%<br />

(54; 96)<br />

19%<br />

(4; 46)<br />

25%<br />

(7; 52)<br />

84%<br />

(64; 96)<br />

96%<br />

(80; 100)<br />

84%<br />

(64; 96)<br />

83%<br />

(68; 93)<br />

66%<br />

(49; 80)<br />

61%<br />

(45; 76)<br />

5,08<br />

(2,01; 12,85)<br />

EUS 4,69<br />

(0,53; 41,24)<br />

<strong>CT</strong><br />

1,56<br />

(0,45; 5,38)<br />

82%<br />

(60; 95)<br />

91%<br />

(71; 99)<br />

82%<br />

(60; 95)<br />

79%<br />

(54; 94)<br />

16%<br />

(3; 40)<br />

26%<br />

(9; 51)<br />

80%<br />

(65; 91)<br />

56%<br />

(40; 72)<br />

56%<br />

(40; 72)<br />

3,89<br />

(1,59; 9,49)<br />

1,08<br />

(0,85; 1,37)<br />

1,11<br />

(0,80; 1,55)<br />

67%<br />

(9; 99)<br />

0%<br />

(0; 71)<br />

33%<br />

(1; 91)<br />

100%<br />

(91; 100)<br />

97%<br />

(86; 100)<br />

100%<br />

(91; 100)<br />

98%<br />

(87; 100)<br />

90%<br />

(77; 97)<br />

95%<br />

(83; 99)<br />

48,75<br />

(2,80; 848,44)<br />

3,25<br />

(0,16; 67,31)<br />

29,25<br />

(1,41; 605,81)


LR-<br />

(95% CI)<br />

DOR<br />

(95% CI)<br />

<strong>PET</strong>-<strong>CT</strong><br />

EUS<br />

<strong>CT</strong><br />

<strong>PET</strong>-<strong>CT</strong><br />

EUS<br />

<strong>CT</strong><br />

0,22<br />

(0,08; 0,63)<br />

0,85<br />

(0,66; 1,09)<br />

0,89<br />

(0,64; 1,24)<br />

22,75<br />

(4,37; 118,34)<br />

5,54<br />

(0,52; 58,76)<br />

1,75<br />

(0,37; 8,30)<br />

0,23<br />

(0,09; 0,58)<br />

0,58<br />

(0,11; 3,09)<br />

0,69<br />

(0,22; 2,21)<br />

16,88<br />

(3,60; 79,19)<br />

1,88<br />

(0,28; 12,61)<br />

1,61<br />

(0,36; 7,12)<br />

0,38<br />

(0,11; 13)<br />

0,91<br />

(0,63; 1,33)<br />

0,63<br />

(0,30; 1,35)<br />

128,33<br />

(4,09; 4029,60)<br />

3,57<br />

(0,12; 105,20)<br />

46,20<br />

(1,47; 1450,60)<br />

With respect to test efficacy in correct identification <strong>of</strong> stage T0, <strong>PET</strong>-<strong>CT</strong> accuracy was<br />

83% (95% CI: 68; 93), <strong>CT</strong> accuracy was 61% (95% CI: 45; 76) and EUS accuracy was 66% (95%<br />

CI: 49; 80).<br />

For stages T1–T3, accuracy was 80% (95% CI: 65; 91) for <strong>PET</strong>-<strong>CT</strong>, 56% (95% CI: 40; 72) for <strong>CT</strong><br />

and 56% (95% CI: 40; 72) for EUS.<br />

For stage T4, the accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was the highest and stood at 98% (95% CI: 87; 100),<br />

while for the other analyzed tests, the accuracy was 90% (95% CI: 77; 97) for EUS, and 95%<br />

(95% CI: 83; 99) for <strong>CT</strong>.<br />

In each case, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was higher than that <strong>of</strong> the other<br />

technologies.<br />

Table 40 lists the proportions <strong>of</strong> patients with the T-feature staged correctly, understaged<br />

and overestaged for the methods compared: <strong>PET</strong>-<strong>CT</strong>, EUS and <strong>CT</strong>.<br />

Table 40.<br />

Proportion <strong>of</strong> cases correctly staged, understaged and overstaged by the diagnostic methods analysed.<br />

<strong>PET</strong>-<strong>CT</strong> EUS <strong>CT</strong><br />

Staged correctly 0,80 0,56 0,56<br />

Understaged 0,12 0,10 0,15<br />

Overstaged 0,07 0,34 0,29<br />

<strong>PET</strong>-<strong>CT</strong> correctly identified the T-status in 80% patients (95% CI: 65; 91), as compared to<br />

56% patients for <strong>CT</strong> and EUS (95% CI: 40; 72).<br />

The odds ratio for correct T-staging and NNT were also calculated for the diagnostic<br />

methods. Table 41 provides the results obtained.<br />

145


Table 41.<br />

Odds ratio for correct T-staging NNT for diagnostic methods analyzed: <strong>PET</strong>-<strong>CT</strong> vs. EUS and <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong><br />

146<br />

<strong>PET</strong>-<strong>CT</strong> vs. EUS<br />

<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong><br />

OR NNT<br />

3,23<br />

(1,09; 10,00)<br />

3,23<br />

(1,09; 10,00)<br />

5<br />

(3; 24)<br />

5<br />

(3; 24)<br />

The odds ratio calculated for <strong>PET</strong>-<strong>CT</strong> vs., EUS and for <strong>PET</strong>-<strong>CT</strong> vs., <strong>CT</strong> is 3.23 (95% CI:<br />

1.09;10.00). It means that the probability <strong>of</strong> correct T-staging with the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 3.23<br />

times higher compared to <strong>CT</strong> and EUS.<br />

In order to obtain one additional case <strong>of</strong> correct T-staging it is necessary to use <strong>PET</strong>-<strong>CT</strong><br />

instead <strong>of</strong> <strong>CT</strong> or EUS with 5 esophageal cancer patients, NNT = 5 (95% CI: 3; 24).<br />

9.1.4.1.2 N-staging<br />

The diagnostic methods compared were used to assess the lymph node involvement.<br />

This feature was not assessed for 7 patients for want <strong>of</strong> full histopathological results.<br />

Table 42.<br />

Diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>, EUS-FNA and <strong>CT</strong> in assessing lymph node involvement<br />

Parameter <strong>PET</strong>-<strong>CT</strong> <strong>CT</strong> EUS-FNA Statistical significance*<br />

TP 5 1 1 -<br />

FP 0 2 2 -<br />

TN 33 31 31 -<br />

FN 3 7 7 -<br />

SE<br />

SP<br />

LR+<br />

LR-<br />

ACC<br />

63%<br />

(24; 91)<br />

100%<br />

(89; 100)<br />

41,55<br />

(2,53; 683,56)<br />

0,39<br />

(0,17; 0,90)<br />

93%<br />

(80; 98)<br />

13%<br />

(0,3; 53)<br />

94%<br />

(80; 99)<br />

2,06<br />

(0,39; 176,84)<br />

0,63<br />

(0,71; 1,23)<br />

78%<br />

(62; 80)<br />

* Statistical significance for the comparison <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> vs. EUS-FNA<br />

13%<br />

(0,3; 53)<br />

94%<br />

(80; 99)<br />

2,06<br />

(0,39; 176,84)<br />

0,63<br />

(0,71; 1,23)<br />

78%<br />

(62; 80)<br />

p = 0,12<br />

p = 0,98<br />

-<br />

-<br />

p = 0,04<br />

Based on the data above, <strong>PET</strong>-<strong>CT</strong> sensitivity is 63% (95% CI: 24; 91), and <strong>CT</strong> and EUS<br />

sensitivity is 13% (95% CI: 0.3; 53), which means that the probability <strong>of</strong> positive results in patients<br />

diagnosed with metastasis to lymph nodes is higher for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong> and EUS.


<strong>PET</strong>-<strong>CT</strong> specificity is 100% (95% CI: 89; 100), while <strong>CT</strong> and EUS specificity is 94% (95% CI: 80;<br />

99), which means that the probability <strong>of</strong> negative results in patients with no metastasis to<br />

lymph nodes identified by the reference test is 100%, 94% and 94% respectively.<br />

The positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 41.55, so the probability <strong>of</strong> positive results in patients<br />

diagnosed with metastasis to lymph nodes is 41.55 times higher than the probability <strong>of</strong> positive<br />

results in patients without metastasis. The positive likelihood ratio for <strong>CT</strong> is 2.06 (95% CI: 0.39;<br />

176.84), which means that the probability <strong>of</strong> positive results in patients diagnosed with<br />

metastasis to lymph nodes is 2.06 <strong>of</strong> the probability <strong>of</strong> positive results in patients with no<br />

metastases diagnosed histopathologically. The positive likelihood ratio for EUS-FNA is 2.06 (95%<br />

CI: 0.39; 176.84), which means that the probability <strong>of</strong> positive results in patients diagnosed with<br />

metastasis to lymph nodes is 2.06 <strong>of</strong> the probability <strong>of</strong> positive results in patients with no<br />

metastasis diagnosed histopathologically.<br />

The negative likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 0.39 (95% CI: 0.17; 0.90), which means that the<br />

probability <strong>of</strong> negative results in patients diagnosed with metastasis to lymph nodes is 0.39<br />

<strong>of</strong> the probability <strong>of</strong> negative results in patients with no metastasis detected. The negative<br />

likelihood ratio calculated for <strong>CT</strong> is 0.63 (0.71; 1.23) so the probability <strong>of</strong> negative results in<br />

patients diagnosed with metastasis to lymph nodes is 0.63 <strong>of</strong> the probability <strong>of</strong> negative<br />

results in patients with no metastasis detected. The negative likelihood ratio calculated for<br />

EUS-FNA is 0.63 (0.71; 1.23) so the probability <strong>of</strong> negative results in patients diagnosed with<br />

metastasis to lymph nodes is 0.63 <strong>of</strong> the probability <strong>of</strong> negative results in patients with no<br />

metastasis detected.<br />

Diagnostic accuracy in N-staging is 93% (95% CI: 80; 98) for <strong>PET</strong>-<strong>CT</strong>, and 78% (95% CI: 62;<br />

80) for both <strong>CT</strong> and EUS-FNA. The differences between <strong>PET</strong>-<strong>CT</strong> and EUS-FNA and <strong>CT</strong><br />

reached statistical significance (p=0.04, test for proportions).<br />

<strong>PET</strong>-<strong>CT</strong> demonstrated the highest sensitivity, specificity, as well as negative and positive<br />

likelihood ratio in N-staging.<br />

9.1.4.1.3 M-staging<br />

The authors <strong>of</strong> Cerfolio 2005 evaluated the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong> vs. EUS<br />

in assessing distant metastasis at stages M1a and M1b.<br />

The efficacy <strong>of</strong> each <strong>of</strong> the technologies in diagnosing metastasis is presented in tables<br />

43 and 44.<br />

147


Table 43.<br />

Diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>, EUS-FNA and <strong>CT</strong> in M-staging: stage M1a<br />

148<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong> EUS<br />

Statistical<br />

significance*<br />

TP 2 0 2 -<br />

FP 1 0 0 -<br />

TN 41 42 42 -<br />

FN 4 6 4 -<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

LR+<br />

(95% CI)<br />

LR-<br />

(95% CI)<br />

DOR<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

33%<br />

(4; 78)<br />

98%<br />

(87; 100)<br />

0%<br />

(0; 46)<br />

100%<br />

(92; 100)<br />

- -<br />

- -<br />

- -<br />

90%<br />

(77; 97)<br />

* Statistical significance for <strong>PET</strong>-<strong>CT</strong> vs. EUS-FNA comparison<br />

88%<br />

(75; 95)<br />

33%<br />

(4; 78)<br />

100%<br />

(92; 100)<br />

0,56<br />

(0,03; 10,02)<br />

1,05<br />

(0,78; 1,42)<br />

0,53<br />

(0,02; 12,83)<br />

92%<br />

(80; 98)<br />

p = 1,00<br />

p = 1,00<br />

-<br />

-<br />

-<br />

p = 0,76<br />

The highest accuracy in assessing the metastatic stage M1a is demonstrated by EUS: 92%<br />

(95% CI: 80; 98); <strong>PET</strong>-<strong>CT</strong> accuracy is 90% (95% CI: 77; 97), and <strong>CT</strong> accuracy is 88% (95% CI: 75;<br />

95). The differences between groups are not statistically significant. The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

and EUS is 33% (95% CI: 4; 78), and for <strong>CT</strong> imaging, sensitivity is 0%, as no true positive results<br />

were reported.<br />

<strong>PET</strong>-<strong>CT</strong> specificity is 98% (95% CI: 87; 100) while for <strong>CT</strong> and EUS imaging, specificity is 100%.<br />

The metastatic stage M1b was assessed by two diagnostic technologies: <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong>.<br />

Table 44.<br />

Diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in M-staging: stage M1b<br />

<strong>PET</strong>-<strong>CT</strong><br />

TP 4 3<br />

FP 4 3<br />

TN - -<br />

FN 2 3<br />

Se<br />

(95% CI)<br />

67%<br />

(22; 96)<br />

<strong>CT</strong><br />

50%<br />

(12; 88)<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity is higher compared to <strong>CT</strong>, stands at 67% (95% CI: 22; 96); for <strong>CT</strong><br />

imaging, sensitivity is 50% (95% CI: 12; 88). The difference is statistically significant.


9.1.4.1.4 CR – complete response<br />

Complete response occurred in 15 (31%) patients. Table 45 presents efficacy values for<br />

detecting complete response using the diagnostic methods compared.<br />

149


Table 45.<br />

Diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>, EUS-FNA and <strong>CT</strong> in assessing complete response<br />

150<br />

Parameter <strong>PET</strong>-<strong>CT</strong> <strong>CT</strong> EUS-FNA<br />

Statistical<br />

significance *<br />

TP 13 4 3 -<br />

FP 4 3 2 -<br />

TN 29 30 31 -<br />

FN 2 11 12 -<br />

SE<br />

SP<br />

LR+<br />

LR-<br />

ACC<br />

87%<br />

(60; 98)<br />

88%<br />

(72; 97)<br />

7,15<br />

(2,79; 18,30)<br />

0,15<br />

(0,04; 0,56)<br />

88%<br />

(95% CI: 75; 95)<br />

27%<br />

(8; 55)<br />

91%<br />

(76; 98)<br />

2,93<br />

(0,75; 11,51)<br />

0,80<br />

(0,58; 1,12)<br />

71%<br />

(95% CI: 56; 83)<br />

20%<br />

(4; 48)<br />

94%<br />

(80; 99)<br />

3,30<br />

(0,61; 17,74)<br />

0,85<br />

(0,65; 1,11)<br />

71%<br />

(95% CI: 56; 83)<br />

p = 0,01<br />

p = 0,60<br />

p = 0,045<br />

*Statistical significance <strong>of</strong> the difference in diagnostic efficacy between <strong>PET</strong>-<strong>CT</strong> and EUS in assessing complete<br />

response.<br />

In the assessment <strong>of</strong> complete response to treatment, <strong>PET</strong>-<strong>CT</strong> demonstrated the highest<br />

sensitivity: 87% (95% CI: 60; 98), compared to 27% (95% CI: 8; 55) for <strong>CT</strong>, and 20% (95% CI: 4;<br />

48) for EUS. The differences between <strong>PET</strong>-<strong>CT</strong> and the reference (index) tests are statistically<br />

significant (p = 0,01 for the comparison with EUS-FNA; test for proportions).<br />

<strong>PET</strong>-<strong>CT</strong> specificity is 88% (95% CI: 72; 97), <strong>CT</strong> specificity is 91% (95% CI: 76; 98), and EUS-FNA<br />

specificity is 94% (95% CI: 80; 99). The differences are not statistically significant.<br />

The highest accuracy was demonstrated by <strong>PET</strong>-<strong>CT</strong>: 88% (95% CI: 75; 95), as compared<br />

with 71% (95% CI: 56; 83) for <strong>CT</strong> and 71% (95% CI: 56; 83) for EUS-FNA. The differences<br />

between <strong>PET</strong>-<strong>CT</strong> and reference tests are statistically significant (p = 0,045 for the comparison<br />

with EUS-FNA, and p = 0.05 for the comparison with <strong>CT</strong>, test for proportions).<br />

9.1.4.2 Safety<br />

The authors identified no undesirable effects <strong>of</strong> the use <strong>of</strong> the diagnostic technologies<br />

compared.


9.1.5. Results<br />

As a result <strong>of</strong> searching medical databases one primary study was found (Cerfolio 2005)<br />

that evaluated the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong> and transesophageal endoscopic<br />

ultrasound-guided biopsy (EUS) in the restaging <strong>of</strong> esophageal cancer and the assessment <strong>of</strong><br />

response to treatment.<br />

In the diagnostics <strong>of</strong> primary tumor (T status), the highest diagnostic accuracy was<br />

demonstrated by <strong>PET</strong>-<strong>CT</strong>: 83% (95% CI: 68; 93) for stage T0, 80% (95% CI: 65; 91) for stages T1-<br />

T3, and 98% (95% CI: 87; 100) for stage T4. In the other tests under assessment, the accuracy <strong>of</strong><br />

<strong>CT</strong> was 61% (95% CI: 45; 76), 56% (95% CI: 40; 72) and 95% (95% CI: 83; 99) respectively, and<br />

the accuracy <strong>of</strong> EUS was 66% (95% CI: 49; 80), 56% (95% CI: 40; 72) and 90% (95% CI: 77; 97).<br />

<strong>PET</strong>-<strong>CT</strong> scanning allowed for correct (consistent with reference test) T-staging in 80%<br />

patients, while <strong>CT</strong> and EUS - in 56% each.<br />

The odds <strong>of</strong> obtaining a correct T-status by using <strong>PET</strong>-<strong>CT</strong> is 3.23 times higher compared to <strong>CT</strong><br />

alone, and to EUS; OR = 3.23 (95% CI: 1.09; 10.00), both for the comparison <strong>PET</strong>-<strong>CT</strong> vs. EUS and<br />

for the comparison <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>; NNT = 5 (95% CI: 3; 24).<br />

In staging lymph nodes involvement (N status), the highest diagnostic accuracy was<br />

achieved for <strong>PET</strong>-<strong>CT</strong>: 93% (95% CI: 80; 98), compared to 78% (95% CI: 62; 80) for <strong>CT</strong> and 78%<br />

(95% CI: 62; 80) for EUS. The difference in accuracy was statistically significant (p=0.04).<br />

The diagnostic efficacy <strong>of</strong> trials in M-staging was evaluated separately for stages M1a and<br />

M1b.<br />

The highest accuracy in determining stage M1a metastasis was demonstrated by EUS – 92%<br />

(95% CI: 80; 98), while the accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 90% (95% CI: 77; 97), and the accuracy <strong>of</strong><br />

<strong>CT</strong> was 88% (95% CI: 75; 95). The differences between the groups were statistically<br />

insignificant. The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and EUS was 33% (95% CI: 4; 78), and the sensitivity <strong>of</strong> <strong>CT</strong><br />

imaging was 0% (95% CI: 0; 40). The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 98% (95% CI: 87; 100), while that <strong>of</strong><br />

<strong>CT</strong> imaging and EUS is 100%.<br />

In the diagnostics <strong>of</strong> stage M1b metastasis, the sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 67% (95% CI: 22; 96)<br />

and was higher than the sensitivity <strong>of</strong> <strong>CT</strong>, which stood at 50% (95% CI: 12; 88).<br />

In the assessment <strong>of</strong> complete response to treatment, <strong>PET</strong>-<strong>CT</strong> showed the highest sensitivity<br />

<strong>of</strong> 87% (95% CI: 60; 98). The sensitivity <strong>of</strong> <strong>CT</strong> was 27% (95% CI: 8; 55), and the sensitivity <strong>of</strong> EUS<br />

was 20% (95% CI: 4; 48). The differences between <strong>PET</strong>-<strong>CT</strong> and the methods compared are<br />

statistically significant. The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning stood at 88% (95% CI: 72; 97), <strong>of</strong> <strong>CT</strong> at<br />

91% (95% CI: 76; 98), and <strong>of</strong> EUS-FNA at 94% (95% CI: 80; 99). The highest accuracy was<br />

demonstrated by <strong>PET</strong>-<strong>CT</strong>, and stood at 88% % (95% CI: 75; 95) vs. 71% (95% CI: 56; 83) for <strong>CT</strong><br />

and 71% (95% CI: 56; 83) for EUS. The differences between <strong>PET</strong>-<strong>CT</strong> and the methods<br />

compared are statistically significant (p=0.045 for the comparison with EUS-FNA, and p=0.05<br />

for the comparison with <strong>CT</strong>).<br />

151


In summary, in restaging esophageal cancer, and in determining complete response to<br />

treatment, <strong>PET</strong>-<strong>CT</strong> imaging demonstrates higher diagnostic efficacy than <strong>CT</strong> or trans-<br />

esophageal ultrasound-guided biopsy.<br />

152<br />

9.2. Diagnostics <strong>of</strong> primary esophageal cancer<br />

9.2.1. Primal tests search results<br />

As a result <strong>of</strong> searching medical databases, one primary trial was found (Kula 2005; tab.<br />

147, app. 18.1) that met the inclusion criteria and compared directly the diagnostic<br />

efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with other methods in esophageal cancer diagnostics.<br />

9.2.2. Population characteristics<br />

The target population were patients with esophageal cancer confirmed histologically or<br />

cytologically, and patients with suspected recurrence <strong>of</strong> esophageal cancer.<br />

The purpose <strong>of</strong> the study was to evaluate the usefulness <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in staging esophageal<br />

cancer. The trial covered 12 men aged from 36 to 78 (59 on average) who had taken<br />

therapy.<br />

The inclusion requirement was esophageal cancer diagnosed previously based on<br />

endoscopic and histopathological tests <strong>of</strong> tumor samples, as well as disease staging<br />

using <strong>CT</strong> <strong>of</strong> the thorax with the use <strong>of</strong> a contrast medium.<br />

The initial patient characteristics is presented in table 46.<br />

Table 46.<br />

Initial patient characteristics in Kula 2005<br />

Staging based on the TNM system<br />

Parameter Kula 2005<br />

Number <strong>of</strong> patients included 12<br />

I 1<br />

IIa 1<br />

IIb 0<br />

III 2<br />

T4 2


IV(M1a) 3<br />

IV(M1b) 3<br />

Prior therapy: chemotherapy 1<br />

Prior therapy:<br />

chemo-radiotherapy<br />

Resection, chemotherapy 2<br />

Surgery 3<br />

Radiotherapy 2<br />

No therapy: exploratory surgery 2<br />

Women 0<br />

Men 12<br />

Age (years)<br />

9.2.3. Description <strong>of</strong> intervention<br />

9.2.3.1 <strong>PET</strong>-<strong>CT</strong> imaging<br />

2<br />

36–78<br />

(average 59.1)<br />

Diagnostic imaging was performed using a <strong>PET</strong>-<strong>CT</strong> scanner with a hybrid head. <strong>PET</strong>-<strong>CT</strong><br />

scanning was done approx. 60-90 minutes after 370-480 MBq <strong>of</strong> FDG was administered.<br />

The radiopharmaceutical could not be given unless the glucose level in blood was<br />

below 8.4 mmol/l.<br />

Table 47.<br />

<strong>PET</strong>-<strong>CT</strong> test characteristics<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

type<br />

Kula 2005 Biograph<br />

<strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

manufacturer<br />

Siemens<br />

Medical<br />

Solutions<br />

(H<strong>of</strong>fman<br />

Estates, III)<br />

Type <strong>of</strong><br />

radiomarker<br />

FDG-<strong>PET</strong><br />

intravenously<br />

9.2.3.2 Diagnostic technologies compared<br />

<strong>PET</strong>-<strong>CT</strong> was compared with <strong>CT</strong>.<br />

Radiomarker<br />

activity<br />

(MBq)<br />

370–480<br />

Test coverage<br />

From orbital<br />

cavities level to<br />

below natis<br />

No data on the type <strong>of</strong> <strong>CT</strong> units used is given in Kula 2005. No detailed description <strong>of</strong><br />

outcome evaluation is given either.<br />

153


154<br />

9.2.3.3 Reference test<br />

In Kula 2005, esophageal cancer had been determined histopathologically or<br />

cytologically for all the patients.<br />

9.2.4. Safety<br />

The authors found no undesirable effects <strong>of</strong> the use <strong>of</strong> the diagnostic methods assessed.<br />

9.2.5. Findings<br />

Primary esophageal cancer was detected in all the 12 patients based on <strong>PET</strong>-<strong>CT</strong><br />

images. No false positive results were identified in the <strong>PET</strong>-<strong>CT</strong> tests. <strong>CT</strong> detected primary<br />

esophageal cancer in 11 primary esophageal cancer patients only. In one case, <strong>PET</strong>-<strong>CT</strong><br />

identified a metastasis to lungs, which could not be imaged by <strong>CT</strong> scanning.<br />

Thus, <strong>PET</strong>-<strong>CT</strong> sensitivity can be described as 100% (95% CI: 74; 100), and <strong>CT</strong> sensitivity as<br />

92% (95% CI: 62; 100).<br />

The remaining data could not be analyzed for want <strong>of</strong> a clear description <strong>of</strong> results or<br />

data for the method compared (<strong>CT</strong>).<br />

9.2.6. Results<br />

As a result <strong>of</strong> searching medical databases one primary trial was found (Kula 2005), which<br />

compared the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with that <strong>of</strong> <strong>CT</strong> in the staging <strong>of</strong> esophageal<br />

cancer (N=12). The reference test was a histopathological or cytological test.<br />

Based on studies accessible, <strong>PET</strong>-<strong>CT</strong> is characterized by higher sensitivity (100%) in<br />

comparison to <strong>CT</strong> (92%) in detecting esophageal cancer.


10. COMPARATIVE ANALYSIS OF THE EFFICACY OF <strong>PET</strong>-<strong>CT</strong> VS.<br />

CONVENTIONAL IMAGING METHODS (<strong>CT</strong>, MRI, USG) IN<br />

CLINICAL STAGING, AND DETE<strong>CT</strong>ING RECURRENCES OF,<br />

MALIGNANT FEMALE GENITAL TUMORS<br />

10.1. Results <strong>of</strong> primary study search<br />

As a result <strong>of</strong> searching medical databases, a single prospective clinical trial was<br />

found (Grisaru 2004, tab. 148, zał. 18.1) that compared the diagnostic efficacy <strong>of</strong><br />

<strong>PET</strong> coupled with x-ray <strong>CT</strong> with conventional imaging methods (computed x-ray<br />

tomography, magnetic resonance imaging and ultrasonography) in staging, and<br />

detecting recurrences <strong>of</strong>, malignant tumors <strong>of</strong> the female genitals.<br />

10.2. Population characteristics<br />

The population in the trial comprised 53 patients diagnosed with malignant<br />

tumors <strong>of</strong> the female genitals (cervical cancer, endometrial cancer, ovarian<br />

cancer, vulvar cancer, vaginal cancer, uterine tube cancer, trophoblast malignant<br />

disease). Indications for <strong>PET</strong>-<strong>CT</strong> diagnostics were based on clinical staging in 18<br />

patients, and suspected recurrence after previous treatment in 35 patients.<br />

Table 48 presents the initial characteristics <strong>of</strong> the population diagnosed.<br />

Table 48.<br />

Characteristics <strong>of</strong> patients diagnosed with malignant female genital tumor in Grisaru 2004.<br />

PARAMETER<br />

Number <strong>of</strong> patients<br />

Median age (SD)<br />

POPULATION<br />

53<br />

56 (15)<br />

The median age <strong>of</strong> subjects was 56 (SD = 15). 21 were diagnosed with cervical cancer<br />

(40%), 8 were diagnosed with endometrial cancer (15%), 19 were diagnosed with<br />

ovarian cancer (36%), while miscellaneous female genital tumors were detected in the<br />

others.<br />

<strong>PET</strong>-<strong>CT</strong> scanning was planned to stage the disease in 34% <strong>of</strong> patients; the other 66% <strong>of</strong><br />

patients underwent the test to determine possible recurrences <strong>of</strong> the disease.<br />

155


156<br />

10.3. Description <strong>of</strong> intervention<br />

10.3.1. <strong>PET</strong>-<strong>CT</strong> imaging<br />

<strong>PET</strong>-<strong>CT</strong> scans were done using the Discovery LS <strong>PET</strong>-<strong>CT</strong> system (GE Medical Systems,<br />

Milwaukee, WI, USA). Fluorine-18 deoxy-glucose was used as a marker. Patients were not<br />

allowed to eat for 4 hours before the test. Also, they were advised to hold their breath<br />

during <strong>CT</strong> scan acquisition. <strong>PET</strong> was performed directly following <strong>CT</strong>, with no change to<br />

the patient's body position. Scans covering the area from cranium to mid thigh were<br />

recorded in 5 to 8 bed positions; the acquisition time <strong>of</strong> a single scan was 5 minutes.<br />

Table 49.<br />

Information on <strong>PET</strong>-<strong>CT</strong> scanner type and scanning method<br />

<strong>PET</strong>-<strong>CT</strong> scanner<br />

type<br />

<strong>PET</strong>-<strong>CT</strong> Scanner<br />

Discovery LS<br />

Radiomarker type<br />

F18-deoxyglucose<br />

Radiomarker<br />

activity<br />

[MBq]<br />

370–666<br />

Contrast medium<br />

type <strong>CT</strong><br />

not used<br />

Coverage<br />

whole body<br />

The scans were interpreted by an experienced physician blinded to the results <strong>of</strong> other<br />

tests.<br />

10.3.2. Diagnostic technologies compared<br />

Before included in the trial, each patient had had her genitals scanned using standard<br />

diagnostic methods (<strong>CT</strong>, MRI and ultrasonography). The authors provide no information on<br />

the time intervals between conventional scanning and <strong>PET</strong>-<strong>CT</strong> scanning.<br />

10.3.3. Reference test<br />

In order to verify the results <strong>of</strong> diagnostic imaging, samples taken during surgeries or<br />

obtained from guided biopsy were tested histopathologically. All negative results <strong>of</strong><br />

histopathological tests were confirmed by further clinical observation and radiological<br />

tests. The authors provide no information on the follow-up period for the population<br />

screened.


10.4. Findings<br />

10.4.1. Disease staging<br />

The EBM parameters <strong>of</strong> diagnostic efficacy (sensitivity, specificity, accuracy, positive<br />

likelihood ratio, negative likelihood ratio and diagnostic odds ratio) were calculated for<br />

two groups separately: patients diagnosed for the purposes <strong>of</strong> disease staging, and<br />

patients with suspected disease recurrence. In both cases, the statistical significance <strong>of</strong><br />

sensitivity and specificity <strong>of</strong> the methods compared were evaluated.<br />

Table 50 presents the number <strong>of</strong> patients with true positive, false positive, false<br />

negative and true negative results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and conventional imaging tests for the<br />

subjects diagnosed for the purposes <strong>of</strong> disease staging. For two patients, conventional<br />

imaging results qualified as indecisive (+/-) in the publication were classified as true<br />

positive.<br />

Table 50.<br />

Number <strong>of</strong> patients with TP, FP, FN and TN results in the group with the disease staged: <strong>PET</strong>-<strong>CT</strong> vs. conventional<br />

methods (<strong>CT</strong>, MRI and USG)<br />

Study<br />

Grisaru 2004<br />

TP<br />

9<br />

FP<br />

0<br />

<strong>PET</strong>-<strong>CT</strong><br />

FN<br />

0<br />

TN<br />

TP<br />

Conventional method<br />

(<strong>CT</strong>, MRI and USG)<br />

Table 51 presents the parameters <strong>of</strong> the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and the<br />

conventional imaging methods (<strong>CT</strong>, MRI and USG) for the patient group diagnosed for the<br />

purposes <strong>of</strong> disease staging.<br />

Table 51.<br />

EBM parameters <strong>of</strong> the diagnostic efficacy in disease staging: <strong>PET</strong>-<strong>CT</strong> vs. conventional methods (<strong>CT</strong>, MRI and USG)<br />

Parameter<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

LR+<br />

(95% CI)<br />

Diagnostic method<br />

Conventional<br />

<strong>PET</strong>-<strong>CT</strong><br />

imaging<br />

(<strong>CT</strong>, MRI and USG)<br />

100%<br />

(66; 100)<br />

100%<br />

(66; 100)<br />

100%<br />

(81; 100)<br />

19,0<br />

(1,3; 284,2)<br />

9<br />

5<br />

56%<br />

(21; 86)<br />

78%<br />

(40; 97)<br />

67%<br />

(41; 87)<br />

2,5<br />

(0,6; 9,7)<br />

FP<br />

2<br />

FN<br />

4<br />

TN<br />

7<br />

Statistical significance<br />

<strong>of</strong> differences<br />

between groups<br />

compared *<br />

p = 0,48<br />

p = 0,04<br />

-<br />

p = 0,13<br />

157


158<br />

LR-<br />

(95% CI)<br />

DOR<br />

(95% CI)<br />

0,05<br />

(0,004; 0,8)<br />

361,0<br />

(6,5; 20143,8)<br />

* p-value for chi-square test (McNemar’s test) Yates corrected<br />

0,6<br />

(0,2; 1,3)<br />

4,4<br />

(0,6; 33,9)<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity in malignant tumor staging is 100% (95% CI: 66; 100), and is higher<br />

than the sensitivity <strong>of</strong> the conventional imaging methods (<strong>CT</strong>, MRI and USG), which is 56%<br />

(95% CI: 21; 86). That means that the probability <strong>of</strong> positive results in patients with<br />

malignant genital tumor metastasis is 100% for <strong>PET</strong>-<strong>CT</strong>, and 56% for the conventional<br />

imaging methods. The result is not statistically significant (p = 0.13).<br />

<strong>PET</strong>-<strong>CT</strong> specificity in disease staging for the group with miscellaneous cancers <strong>of</strong> the<br />

female genitals is 100% (95% CI: 66; 100), and is higher compared to the specificity <strong>of</strong> the<br />

conventional imaging methods, which is 78% (95% CI: 40; 97). That means that for patients<br />

with no metastasis, the probability <strong>of</strong> correct clinical staging is 100% for <strong>PET</strong>-<strong>CT</strong>, and is higher<br />

than for the conventional imaging methods (78%). The difference is not statistically<br />

significant (p = 0.48).<br />

<strong>PET</strong>-<strong>CT</strong> accuracy is 100% (95% CI: 81; 100), and is higher compared with the accuracy<br />

<strong>of</strong> the conventional imaging methods, which is 67% (95% CI: 41; 87). That means that the<br />

probability <strong>of</strong> correct clinical staging is 100% for <strong>PET</strong>-<strong>CT</strong>, and is higher compared with the<br />

conventional imaging methods (67%). The difference is statistically significant (p = 0.04).<br />

The positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 19.0 (95% CI: 1.3; 284.2), and is higher compared<br />

with the ratio for the conventional imaging methods, which is 2.5% (95% CI: 0.6; 9.7). For <strong>PET</strong>-<br />

<strong>CT</strong>, the probability <strong>of</strong> positive results is 19 times higher for patients with malignant genital<br />

tumor metastasis than for patients without such metastasis.<br />

For the conventional methods, the probability <strong>of</strong> positive results is 2.5 times higher for<br />

patients with malignant genital tumor metastasis than for patients without such metastasis.<br />

The negative likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 0.05 (95% CI: 0.004; 0.8), and is higher than the<br />

ratio for the conventional imaging methods, which is 0.6% (95% CI: 0,2; 1,3). For <strong>PET</strong>-<strong>CT</strong>, the<br />

probability <strong>of</strong> negative results in patients with malignant genital tumor metastasis represents<br />

5% <strong>of</strong> the probability <strong>of</strong> negative results in patients without such metastasis.<br />

For the conventional methods, the probability <strong>of</strong> negative results in patients with<br />

malignant genital tumor metastasis represented 60% <strong>of</strong> the probability <strong>of</strong> negative results<br />

in patient with no such metastasis.<br />

The diagnostic odds ratio for <strong>PET</strong>-<strong>CT</strong> in the staging <strong>of</strong> disease in patients with<br />

miscellaneous cancers <strong>of</strong> the female genitals is 361.0 (95% CI: 6.5; 20143.8), and is higher<br />

than the diagnostic odds ratio for the conventional imaging methods, which is 4.4 (95% CI:<br />

0,4; 33,9).<br />

-<br />

-


10.4.2. Assessment <strong>of</strong> disease recurrence<br />

Table 52 provides the count <strong>of</strong> patients with true positive, false positive, false negative or<br />

true negative results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> tests and conventional imaging for the group diagnosed for<br />

the purposes <strong>of</strong> disease recurrence assessment. The outcome <strong>of</strong> a conventional imaging<br />

test for one patient was not taken into account due to an anaphylactic reaction to the<br />

contrast medium given intravenously; a <strong>PET</strong>-<strong>CT</strong> scan for this patient gave a positive result,<br />

and the reference test confirmed disease recurrence. For two patients, the results <strong>of</strong><br />

conventional tests qualified as indecisive (+/-) for the publication are considered true<br />

positive.<br />

Table 52.<br />

Patients with TP, FP, FN and TN results in the group with disease recurrence assessed, <strong>PET</strong>-<strong>CT</strong> vs. conventional methods<br />

(<strong>CT</strong>, MRI and USG)<br />

Study<br />

Grisaru 2004<br />

TP<br />

25<br />

FP<br />

1<br />

<strong>PET</strong>-<strong>CT</strong><br />

FN<br />

1<br />

TN<br />

8<br />

TP<br />

9<br />

Conventional method<br />

(<strong>CT</strong>, MRI and USG)<br />

Table 53 presents EBM parameters <strong>of</strong> the diagnostic methods compared for the group<br />

<strong>of</strong> patients with suspected disease recurrence.<br />

Table 53.<br />

EBM parameters <strong>of</strong> the diagnostic efficacy in assessing disease recurrence: <strong>PET</strong>-<strong>CT</strong> vs. conventional methods (<strong>CT</strong>, MRI<br />

and USG)<br />

Parameter<br />

(95% CI)<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

LR+<br />

(95% CI)<br />

LR-<br />

(95% CI)<br />

Diagnostic method<br />

Conventional imaging<br />

<strong>PET</strong>-<strong>CT</strong><br />

(<strong>CT</strong>, MRI, USG)<br />

96%<br />

(80; 100)<br />

89%<br />

(52; 100)<br />

94%<br />

(81; 99)<br />

8,7<br />

(1,4; 55,0)<br />

0,04<br />

(0,006; 0,3)<br />

DOR<br />

200,0<br />

(95% CI)<br />

(11,2; 3576,7)<br />

* p-value for chi-square test (McNemar’s test) Yates corrected<br />

36%<br />

(18; 57)<br />

56%<br />

(21; 86)<br />

41%<br />

(25; 59)<br />

0,8<br />

(0,3; 2,0)<br />

1,2<br />

(0,6; 2,2)<br />

0,7<br />

(0,1; 3,3)<br />

FP<br />

4<br />

FN<br />

16<br />

TN<br />

Statistical significance<br />

<strong>of</strong> difference between<br />

groups compared *<br />

p < 0,01<br />

p = 0,37<br />

p < 0,01<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity in detecting disease recurrence is 96% (95% CI: 80; 100), and is higher<br />

than the sensitivity <strong>of</strong> the conventional imaging methods (<strong>CT</strong>, MRI and USG), which is 36%<br />

(95% CI: 18; 57). For <strong>PET</strong>-<strong>CT</strong>, the probability <strong>of</strong> positive results in patients diagnosed with<br />

-<br />

-<br />

-<br />

5<br />

159


160<br />

disease recurrence is 60 p.p. higher than for the conventional methods. The difference is<br />

statistically significant in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> (p < 0,01).<br />

A difference in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was also observed in test specificity. <strong>PET</strong>-<strong>CT</strong> specificity is<br />

89% (95% CI: 52; 100) and is higher than the specificity <strong>of</strong> the conventional imaging<br />

methods, which is 56% (95% CI: 21; 86), which means that the probability <strong>of</strong> negative<br />

results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning is 89% in patients with no disease recurrence detected, which is<br />

33 p.p. higher than for the conventional methods. The difference is not statistically<br />

significant (p = 0.37).<br />

<strong>PET</strong>-<strong>CT</strong> accuracy in detecting disease recurrence is 94% (95% CI: 81; 99), and is higher than<br />

the accuracy <strong>of</strong> the conventional imaging methods (<strong>CT</strong>, MRI and USG), which is 41% (95%<br />

CI: 25; 59). The difference is statistically significant in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> study (p < 0,01).<br />

The positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 8.7 (95% CI: 1.4; 55.0), and is higher than the<br />

same ratio for the conventional imaging methods, which stands at 0.8 (95% CI: 0.3; 2.0). For<br />

<strong>PET</strong>-<strong>CT</strong> scanning, the probability <strong>of</strong> positive results in patients diagnosed with disease<br />

recurrence is almost 9 times higher than the probability <strong>of</strong> positive results in patients with no<br />

disease recurrence detected.<br />

For the conventional methods, the probability <strong>of</strong> positive results in patients diagnosed<br />

with disease recurrence represents 80% <strong>of</strong> the probability <strong>of</strong> positive results in patients with<br />

no disease recurrence detected.<br />

The negative likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 0.04 (95% CI: 0.006; 0.3) and is lower compared to<br />

the ration for the conventional imaging methods, which is 1.2% (95% CI: 0,6; 2,2). For <strong>PET</strong>-<strong>CT</strong>,<br />

the probability <strong>of</strong> negative results in patients diagnosed with disease recurrence is 4% <strong>of</strong> the<br />

probability <strong>of</strong> negative results in patients with no disease recurrence detected.<br />

For the conventional methods, the probability <strong>of</strong> negative results in patients diagnosed<br />

with disease recurrence is 110% <strong>of</strong> such probability in patients with no recurrence detected.<br />

The diagnostic odds ratio for detecting disease recurrence in the group with various<br />

female genital cancer types is 200.0 (95% CI: 11.2; 3576.7) for <strong>PET</strong>-<strong>CT</strong>, and is higher than the<br />

diagnostic odds ratio for the conventional imaging methods, which is 0.7 (95% CI: 0,1; 3,3).<br />

10.4.3. Total assessment<br />

Grisaru 2004 presented the sensitivity, specificity, and the negative and positive<br />

likelihood ratios for <strong>PET</strong>-<strong>CT</strong> study and conventional methods (<strong>CT</strong>, MRI and USG) for both<br />

groups <strong>of</strong> patients.<br />

Table 54 provides patient counts for the results determining the diagnostic efficacy <strong>of</strong><br />

tests in the total population <strong>of</strong> patients for who T-status and disease recurrence were<br />

assessed. One patient who had had an anaphylactic reaction to the <strong>CT</strong> contrast medium<br />

given intravenously is left out from the results for the conventional methods; that patient


tested positive in <strong>PET</strong>-<strong>CT</strong> scanning, and the reference test confirmed disease recurrence.<br />

Table 54.<br />

Number <strong>of</strong> patients with TP, FP, FN and TN results, <strong>PET</strong>-<strong>CT</strong> vs. conventional methods (<strong>CT</strong>, MRI and USG)<br />

Study<br />

Grisaru 2004<br />

TP<br />

34<br />

FP<br />

1<br />

<strong>PET</strong>-<strong>CT</strong><br />

FN<br />

1<br />

TN<br />

17<br />

TP<br />

14<br />

Conventional method<br />

(<strong>CT</strong>, MRI and USG)<br />

Table 55 presents sensitivity and specificity values as quoted by the authors <strong>of</strong> the<br />

study, as well as some additional EBM parameters calculated for the imaging methods<br />

compared.<br />

Table 55.<br />

EBM parameters <strong>of</strong> diagnostic efficacy in disease staging and disease recurrence assessment; <strong>PET</strong>-<strong>CT</strong> vs.<br />

conventional methods (<strong>CT</strong>, MRI and USG)<br />

Parameter<br />

(95% CI)<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

LR+<br />

(95% CI)<br />

LR-<br />

(95% CI)<br />

Diagnostic method<br />

Conventional imaging<br />

<strong>PET</strong>-<strong>CT</strong><br />

(<strong>CT</strong>, MRI, USG)<br />

97%<br />

(85; 100)<br />

94%<br />

(73; 100)<br />

96%<br />

(87; 100)<br />

17,5<br />

(2,6; 117,6)<br />

0,03<br />

(0,004; 0,2)<br />

DOR<br />

578,0<br />

(95% CI)<br />

(34,0; 9817,0)<br />

* p-value for chi-square test (McNemar’s test) Yates corrected<br />

41%<br />

(25; 59)<br />

67%<br />

(41; 87)<br />

50%<br />

(36; 64)<br />

1,2<br />

(0,6; 2,7)<br />

0,9<br />

(0,6; 1,4)<br />

1,4<br />

(0,4; 4,6)<br />

FP<br />

6<br />

FN<br />

20<br />

TN<br />

12<br />

Statistical significance<br />

<strong>of</strong> difference between<br />

groups*<br />

p < 0,01<br />

p = 0,13<br />

p < 0,01<br />

<strong>PET</strong>-<strong>CT</strong> is characterized by higher sensitivity and specificity compared to the<br />

conventional imaging methods (<strong>CT</strong>, MRI and USG) in the staging and recurrence<br />

assessment <strong>of</strong> malignant female genital tumors.<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity is 97% (95% CI: 85; 100), and is higher compared with the sensitivity <strong>of</strong><br />

the conventional imaging methods (<strong>CT</strong>, MRI and USG), which is 41% (95% CI: 25; 59). That<br />

means that the probability <strong>of</strong> positive results in patients diagnosed with malignant tumor<br />

metastasis to genitals or patients with disease recurrence detected is 97% for <strong>PET</strong>-<strong>CT</strong> and<br />

41% for the conventional imaging methods. The difference is statistically significant and is<br />

56 p.p. (p < 0.01).<br />

<strong>PET</strong>-<strong>CT</strong> specificity in disease staging and recurrence detection is 94% (95% CI: 73; 100),<br />

-<br />

-<br />

-<br />

161


and is higher compared with the specificity <strong>of</strong> the conventional imaging methods, which<br />

stands at 67% (95% CI: 41; 87). The probability <strong>of</strong> negative results in patients with no<br />

metastasis <strong>of</strong> malignant female genital tumors or with no disease recurrence detected is<br />

94% for <strong>PET</strong>-<strong>CT</strong>, which is 27 p.p. higher than for the conventional imaging methods (67%).<br />

The difference is not statistically significant (p = 0.13).<br />

162<br />

<strong>PET</strong>-<strong>CT</strong> accuracy is 96% (95% CI: 87; 100), and is higher compared with the accuracy <strong>of</strong><br />

the conventional imaging methods (<strong>CT</strong>, MRI and USG), which is 50% (95% CI: 36; 64). The<br />

probability <strong>of</strong> correct assessment is 96% for <strong>PET</strong>-<strong>CT</strong>, which is 46 p.p. higher than for the<br />

conventional imaging methods. The difference between the groups is not statistically<br />

significant (p < 0.01).<br />

Based on the data available in the study, additional parameters <strong>of</strong> diagnostic efficacy<br />

were calculated: negative likelihood ratio, positive likelihood ratio and diagnostic odds<br />

ratio.<br />

The positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 17.5 (95% CI: 2.6; 117.6), and is higher than the ratio<br />

for the conventional imaging methods, which is 1.2% (95% CI: 0.6; 2.7). For <strong>PET</strong>-<strong>CT</strong>, the<br />

probability <strong>of</strong> positive results in patients diagnosed with metastasis <strong>of</strong> malignant genital<br />

tumors or patients with disease recurrence detected is almost 17 times higher compared to<br />

the probability <strong>of</strong> positive results in patients with no metastasis <strong>of</strong> malignant genital tumors or<br />

disease recurrence.<br />

For the conventional methods, the probability <strong>of</strong> positive results in patients diagnosed<br />

with metastasis <strong>of</strong> malignant genital tumors or with disease recurrence detected is 120% <strong>of</strong><br />

the probability <strong>of</strong> positive results in patients with no metastasis <strong>of</strong> malignant genital tumor or<br />

disease recurrence.<br />

The negative likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 0.03 (95% CI: 0.004; 0.2), and is lower than the ratio<br />

for the conventional imaging methods, which is 0.9 (95% CI: 0.6; 1.4).<br />

For <strong>PET</strong>-<strong>CT</strong>, the probability <strong>of</strong> negative results in patients diagnosed with metastasis <strong>of</strong><br />

malignant genital tumors or with disease recurrence detected represented 3% <strong>of</strong> the<br />

probability <strong>of</strong> negative results in patients with no metastasis <strong>of</strong> malignant genital tumor or<br />

disease recurrence.<br />

For the conventional methods, the probability <strong>of</strong> negative results in patients diagnosed<br />

with metastasis <strong>of</strong> malignant genital tumors or with disease recurrence detected is 90% <strong>of</strong><br />

the probability <strong>of</strong> negative results in patients without metastasis <strong>of</strong> malignant genital tumors<br />

or disease recurrence.<br />

For <strong>PET</strong>-<strong>CT</strong>, the diagnostic odds ratio for clinical disease staging and recurrence<br />

detection in the group with miscellaneous genital cancers is 578.0 (95% CI: 34.0; 9817.0),<br />

and is higher compared with the diagnostic odds ratio for the conventional imaging<br />

methods, which stands at 1.4 (95% CI: 0,4; 4,6).


10.5. Results<br />

As a result <strong>of</strong> searching medical databases one primary clinical trial was found (Grisaru<br />

2004) that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> vs. conventional imaging<br />

methods (<strong>CT</strong>, MRI and ultrasonography) in the diagnostics <strong>of</strong> female genital cancer (N=53).<br />

Positive results <strong>of</strong> scanning were verified by histopathological tests <strong>of</strong> material removed<br />

during a surgical procedure or obtained from guided biopsy. Negative results were verified<br />

based on long-term clinical observation and repeated diagnostic imaging.<br />

The assessment <strong>of</strong> the diagnostic efficacy <strong>of</strong> imaging methods discussed included both<br />

staging and detecting recurrences <strong>of</strong> disease.<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostics in clinical staging is higher than the sensitivity <strong>of</strong> the<br />

conventional diagnostic imaging and stands at 100% (95% CI: 66; 100) vs. 56% (95% CI: 21; 86).<br />

The difference is statistically insignificant (p=0.13). The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 100% (95% CI: 66;<br />

100), and that <strong>of</strong> <strong>CT</strong>, MRI and ultrasound scanning is 78% (95% CI: 40; 97). The difference is<br />

statistically insignificant (p=0.48). The diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in the primary staging <strong>of</strong><br />

disease is 100% (95% CI: 81; 100) and is higher than the accuracy <strong>of</strong> the conventional imaging<br />

methods, which is 67% (95% CI: 41; 87). The difference is statistically significant (p=0.04).<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting disease recurrence is 96% (95% CI: 80; 100) and is higher<br />

than the sensitivity <strong>of</strong> the conventional imaging methods, which is 36% (95% CI: 18; 57). The<br />

difference is statistically significant, in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> (p


11. COMPARATIVE ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> AND <strong>CT</strong> EFFICACY IN<br />

THE DIAGNOSTICS OF OVARIAN CANCER<br />

164<br />

11.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching through medical databases, two primary studies were found<br />

(Hauth 2005; tab. 149, app. 18.1 and Makhija 2001; tab. 150, app. 18.1), that satisfied the<br />

inclusion criteria and compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong> with <strong>CT</strong> in the<br />

assessment <strong>of</strong> ovarian cancer recurrences.<br />

11.2. Population characteristics<br />

Hauth 2005 is a prospective clinical trial that included 19 patients. They had taken surgeries<br />

to have ovarian cancer with suspected recurrence removed. The median <strong>of</strong> time from<br />

diagnosis to <strong>PET</strong>-<strong>CT</strong> scanning was one year (range: 0.5–4 years).<br />

Makhija 2001 is a retrospective trial. The population comprised patients with ovarian or<br />

fallopian tube adenocarcinoma recurrence confirmed in histopathological tests taken<br />

following a surgery. Six patients had ovarian cancer, and two had fallopian tube cancer.<br />

The initial characteristics <strong>of</strong> the subject populations is given in table 56.


Table 56.<br />

Characteristics <strong>of</strong> population included in the <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> efficacy in the assessment <strong>of</strong> ovarian cancer<br />

recurrences<br />

* average weighted according to the size <strong>of</strong> population<br />

11.3. Description <strong>of</strong> intervention<br />

11.3.1. <strong>PET</strong>-<strong>CT</strong> imaging<br />

Hauth 2005<br />

<strong>PET</strong>-<strong>CT</strong> scanning was carried out using a Biograph TM unit (Siemens Medical Solutions). Scans<br />

were taken 60 minutes after 350 MBq <strong>of</strong> FDG was administered. Attenuation was revised by<br />

<strong>CT</strong>. The imaging covered whole body. <strong>PET</strong> scans were taken in 6 table positions, for 4 minutes<br />

each. <strong>PET</strong> images were interpreted by two nuclear medicine specialists, and <strong>CT</strong> images by<br />

tworadiologists. <strong>PET</strong>-<strong>CT</strong> images were interpreted by consensus by all the specialists<br />

mentioned.<br />

Makhija 2001<br />

Parameter Hauth 2005 Makhija 2001 Total<br />

Size <strong>of</strong> population 19 8 27<br />

Average age (range) [years] 67 (49–80) 55 (50–73) 63,5*<br />

Primary FIGO stage<br />

[persons]<br />

I IA - 1 1<br />

II<br />

III<br />

IIA 1<br />

IIC<br />

<strong>PET</strong>-<strong>CT</strong> scanning was carried out using a prototype hybrid scanner consisting <strong>of</strong> a ECAT<br />

ART <strong>PET</strong> unit (System <strong>CT</strong>I <strong>PET</strong> ) and a Somatom AR.SP <strong>CT</strong> unit (Siemens Medical Systems). Scans<br />

<strong>of</strong> pelvis and abdomen were taken one hour after 6 to 8 mCi <strong>of</strong> FDG was administered<br />

intravenously. Oral contrast was used in two patients, and dynamic intravenous injections <strong>of</strong><br />

contrast were given to four. Attenuation was revised by the <strong>CT</strong> component. A group <strong>of</strong><br />

radiologists and nuclear medicine specialists interpreted the <strong>PET</strong>-<strong>CT</strong> scans. Lesions <strong>of</strong> SUV<br />

3<br />

IIIA 1<br />

IIIC<br />

13<br />

IV IVB 3 1 4<br />

1<br />

3<br />

5<br />

17<br />

165


equal or higher than 2.5 were regarded malignant. The researchers interpreting <strong>PET</strong>-<strong>CT</strong><br />

images had access to clinical data.<br />

166<br />

11.3.2. Diagnostic technology compared<br />

Hauth 2005<br />

<strong>CT</strong> scanning was carried out using a Somatom Emotion unit (Siemens Medical Solutions).<br />

Patients were contrasted both intravenously and orally. The image was interpreted by two<br />

radiologists. There is no information on blinding the researchers.<br />

Makhija 2001<br />

<strong>CT</strong> scanning. There are no data on the way the trial was carried out, the equipment used<br />

or the way results were evaluated.<br />

11.3.3. Reference test<br />

In Hauth 2005, post-operational histopathological examinations were done as reference<br />

test in five out <strong>of</strong> eleven patients with disease recurrence. The other patients underwent 6-<br />

months’ observation that covered all clinical data available (general examination, set <strong>of</strong><br />

laboratory tests including CA125 antigen, as well as <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> scanning).<br />

The reference test in Makhija 2001 was post-operational histopathological examination.<br />

11.4. Findings<br />

11.4.1. Diagnostic efficacy<br />

The results <strong>of</strong> whole-body contrast scanning using the <strong>PET</strong>-<strong>CT</strong> technology in Hauth 2005<br />

were as follows:<br />

• <strong>PET</strong>-<strong>CT</strong> returned positive results in all the confirmed cases <strong>of</strong> cancer: eleven patients;<br />

for all negative <strong>PET</strong>-<strong>CT</strong> results the reference test showed no recurrence;<br />

• Contrast <strong>CT</strong> detected cancer in eight out <strong>of</strong> eleven positive patients; negative results<br />

<strong>of</strong> <strong>CT</strong> were true for eight and false for three patients.<br />

Table 57.<br />

Number <strong>of</strong> patients with TP, FP, FN and TN results in Hauth 2005<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

TP FP FN TN TP FP FN TN<br />

Hauth 2005 11 0 0 8 8 0 3 8


Table 58 presents diagnostic efficacy parameters as calculated for <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in<br />

detecting recurrences <strong>of</strong> ovarian cancer.<br />

Table 58.<br />

<strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> diagnostic efficacy in assessing ovarian cancer recurrences.<br />

Results /<br />

parameters<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

Statistical significance <strong>of</strong><br />

difference between groups<br />

*<br />

Se (95% CI) 100% (72; 100) 73% (39; 94) p = 0.25<br />

Sp (95% CI) 100% (63; 100) 100.0% (63; 100) NS<br />

Acc (95% CI) 100% (82; 100) 84% (60; 97) p = 0.25<br />

LR+ (95% CI) 1725 (1.16; 255.73) 12,75 (0.84; 193.18) -<br />

LR- (95% CI) 0.04 (0.003; 0.67) 0.31 (0.13; 0.76) -<br />

DOR (95% CI)<br />

391.00 (7.03;<br />

21756.1)<br />

41.29 (1.84; 927.50) -<br />

* p-value for chi-square test(McNemar’s test) Yates corrected<br />

<strong>PET</strong>-<strong>CT</strong> Sensitivity was 100% (95% CI: 72; 100), and <strong>CT</strong> sensitivity was 73% (95% CI: 39; 94).<br />

That means that <strong>PET</strong>-<strong>CT</strong> detected correctly cancer recurrence in all cases, whereas <strong>CT</strong><br />

detected changes in only 73% <strong>of</strong> patients with recurrence.<br />

The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> was 100% (95% CI: 63; 100), which means that for all the<br />

patients with no cancer recurrence the result was negative.<br />

<strong>PET</strong>-<strong>CT</strong> accuracy was 100% (95% CI: 82; 100), which means that all the patients were<br />

diagnosed correctly. The accuracy <strong>of</strong> <strong>CT</strong> was 84% (95% CI: 60; 97), which means that<br />

diagnosis was correct for 84% patients.<br />

Group differences for the above-mentioned parameters had no statistical significance.<br />

The positive likelihood ratio was calculated for <strong>PET</strong>-<strong>CT</strong> at 17.25 (95% CI: 1,16; 255,73), so the<br />

probability <strong>of</strong> positive results in patients with recurrence is 17.25 times higher than for patients<br />

with no recurrence. The positive likelihood ratio was calculated for <strong>CT</strong> at 12.75 (95% CI: 0.84;<br />

193.18), which means that the probability <strong>of</strong> positive results in patients with recurrence is 12.75<br />

times higher than the likelihood for patients with no recurrence.<br />

The negative likelihood ratio was 0.04 for <strong>PET</strong>-<strong>CT</strong> (95% CI: 0.003; 0.67), which means that the<br />

probability <strong>of</strong> negative results in patients with ovarian cancer recurrence represents 0.04 <strong>of</strong><br />

the likelihood for patients with no recurrence. The negative likelihood ratio was 0.31 for <strong>CT</strong><br />

(95% CI: 0.13; 0.76), so the probability <strong>of</strong> positive results in patients with cancer recurrence<br />

represents 0.31 <strong>of</strong> the likelihood for patients with no recurrence.<br />

167


168<br />

The diagnostic odds ratio as calculated for <strong>PET</strong>-<strong>CT</strong> is 391.00 (95% CI: 7,03; 21756,1). That<br />

means that the probability <strong>of</strong> positive results is 391 times higher in patients with cancer<br />

recurrence than the probability in the patient group with no recurrence detected using the<br />

reference test. The diagnostic odds ratio for <strong>CT</strong> is 41.29 (95% CI: 1.84; 927.50), which means<br />

that the probability <strong>of</strong> positive results in patients diagnosed with cancer recurrence by the<br />

reference test is 41.29 times higher than the probability in patients with no recurrence.<br />

<strong>CT</strong> scanning detected ovarian cancer in 12 locations, whereas <strong>PET</strong>-<strong>CT</strong> in 18. In 4 out <strong>of</strong> 6<br />

cases <strong>of</strong> local recurrence or involvement <strong>of</strong> lymph nodes in the pelvis detected by <strong>PET</strong>-<strong>CT</strong>, <strong>CT</strong><br />

returned negative results. Moreover, <strong>CT</strong> failed to detect metastasis located in the thoracic<br />

wall or diaphragm, which were detected by <strong>PET</strong>-<strong>CT</strong>.<br />

Table 59.<br />

Location <strong>of</strong> lesions detected by <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> scanning, Hauth 2005<br />

Location <strong>of</strong> tumor <strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

Local recurrence 3 1<br />

Metastasis to pelvic lymph<br />

nodes<br />

Metastasis to periaortic,<br />

mediastinal and neck<br />

lymph nodes<br />

3 1<br />

6 6<br />

Remote metastasis 4 2<br />

Infiltration <strong>of</strong> the peritoneum 2 2<br />

Total 18 12<br />

In Makhija 2001, <strong>PET</strong>-<strong>CT</strong> detected cancer in five patients, while <strong>CT</strong> detected ovarian<br />

cancer in one patient and in another it accidentally detected a liver cyst, unrelated to<br />

cancer.<br />

The reference test found carcinoma recurrences in all the patients examined. <strong>PET</strong>-<strong>CT</strong> failed<br />

to detect three out <strong>of</strong> eight cases <strong>of</strong> cancer, while <strong>CT</strong> failed to detect seven out <strong>of</strong> eight<br />

recurrences.<br />

Table 60.<br />

Number <strong>of</strong> patients with TP, FP, FN and TN results in Makhija 2001<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

TP FP FN TN TP FP FN TN


Makhija 2001 5 0 3 0 1 0 7 0<br />

The sensitivity <strong>of</strong> the diagnostic tests was calculated on the basis <strong>of</strong> the number <strong>of</strong><br />

correctly and incorrectly diagnosed patients.<br />

Table 61.<br />

Sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in Makhija 2001<br />

Parameter <strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

Statistical significance <strong>of</strong><br />

difference between<br />

groups *<br />

Se (95% CI) 63% (24; 91) 13% (0; 53) p = 0,22<br />

* p-value for chi-square test (McNemar’s test) Yates corrected<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity was 63% (95% CI: 24; 91), and <strong>CT</strong> sensitivity was 13% (95% CI: 0; 53), which<br />

means that <strong>PET</strong>-<strong>CT</strong> identified 63% <strong>of</strong> the patients with carcinoma, and <strong>CT</strong> merely 13%.<br />

According to McNemar’s test, the difference in sensitivity between <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong>, was not<br />

statistically significant (p = 0,22) due to the small number size <strong>of</strong> subject population.<br />

Graph 22 presents the results <strong>of</strong> sensitivity meta-<strong>analysis</strong> (the only meta-analyzable<br />

parameter in view <strong>of</strong> the data deficiency <strong>of</strong> Makhija 2001).<br />

Graph 22.<br />

Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> sensitivity in detecting ovarian cancer recurrence.<br />

Hauth 2005<br />

Makhija 2001 0.63 (0.24, 0.91)<br />

Result <strong>of</strong> meta-nalysis 0.84 (0.60, 0.97)<br />

0.0 0.2 0.4 0.6 0.8 1.0<br />

Sensitivity (95% range <strong>of</strong> confidence)<br />

The result <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> sensitivity meta-<strong>analysis</strong> was 84% (95% CI: 60; 97), which means that<br />

84% <strong>of</strong> patients with cancer recurrence were diagnosed correctly using <strong>PET</strong>-<strong>CT</strong>.<br />

1.00 (0.72, 1.00)<br />

169


Graph 23.<br />

Meta-<strong>analysis</strong> <strong>of</strong> <strong>CT</strong> sensitivity in detecting ovarian cancer recurrence<br />

170<br />

Hauth 2005 0.73 (0.39, 0.94)<br />

Makhija 2001 0.13 (0.00, 0.53)<br />

Result <strong>of</strong> met<strong>analysis</strong><br />

The result <strong>of</strong> <strong>CT</strong> sensitivity meta-<strong>analysis</strong> was 47% (95% CI: 24; 71), which means that 47% <strong>of</strong><br />

patients with cancer recurrence were diagnosed correctly using <strong>CT</strong>.<br />

Table 62 lists meta-<strong>analysis</strong> results for <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> sensitivity.<br />

Table 62.<br />

Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> sensitivity in detecting ovarian cancer recurrence<br />

Results/ parameters <strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

Se (95% CI) 84% (60; 97) 47% (24; 71)<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in the diagnostics <strong>of</strong> ovarian cancer recurrences is 84% (95% CI: 60;<br />

97), and the sensitivity <strong>of</strong> <strong>CT</strong> is 47% (95% CI: 24; 71).<br />

Clearly, the sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in diagnosing ovarian cancer recurrence is significantly<br />

higher than the sensitivity <strong>of</strong> <strong>CT</strong>. However, the high heterogeneity <strong>of</strong> test results and the small<br />

size <strong>of</strong> subject populations make the final results less reliable.<br />

11.4.2. Safety<br />

None <strong>of</strong> the trials included in this study provides information on the safety <strong>of</strong> the diagnostic<br />

procedures used.<br />

0.0 0.2 0.4 0.6 0.8 1.0<br />

Sensitivity (95% range <strong>of</strong> confidence)<br />

0.47 (0.24, 0.71)


11.5. Results<br />

As a result <strong>of</strong> searching medical databases two primary clinical trials were found (Hauth<br />

2005 and Makhija 2001) that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with <strong>CT</strong> in<br />

the assessment <strong>of</strong> ovarian cancer recurrences (N=27). The reference test was<br />

histopathological examination or, if histopathological examination was inaccessible, clinical<br />

follow-up.<br />

<strong>PET</strong>-<strong>CT</strong> is characterized by higher sensitivity than contrast <strong>CT</strong>. The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 84%<br />

(95% CI: 60; 97) and that <strong>of</strong> <strong>CT</strong> is 47% (95% CI: 24; 71).<br />

The specificity <strong>of</strong> these methods, evaluated only in Hauth 2005, was 100% for both <strong>PET</strong>-<strong>CT</strong><br />

and <strong>CT</strong> (95% CI: 63; 100). That implies that a negative result was returned by the imaging<br />

methods under <strong>analysis</strong> for each patient with no cancer recurrence.<br />

The data provided testify to the superiority <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostics over contrast <strong>CT</strong> imaging.<br />

The small size <strong>of</strong> population in the primary studies limits the reliability <strong>of</strong> the <strong>analysis</strong>.<br />

171


12. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS.<br />

TRADITIONAL IMAGING METHODS IN THYROID CANCER<br />

DIAGNOSTICS<br />

172<br />

12.1. Comparative <strong>analysis</strong> <strong>of</strong> the clinical efficacy <strong>of</strong> pet-ct vs.<br />

traditional imaging methods (ct/mri and i131whole-body<br />

scintigraphy) in the diagnostics <strong>of</strong> thyroid cancer<br />

recurrences<br />

12.1.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching medical databases, two original trials (Zimmer 2003; tab. 153, app.<br />

18.1, Nahas 2005; tab. 151, zał. 18.1) were found that compare the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<br />

<strong>CT</strong> with that <strong>of</strong> other imaging methods (<strong>CT</strong>/MRI and whole-body scintigraphy using I 131) in the<br />

diagnostics <strong>of</strong> thyroid cancer recurrence. Zimmer 2003 was a prospective trial, whereas<br />

Nahas 2005 involved a retrospective <strong>analysis</strong> <strong>of</strong> the impact <strong>of</strong> additional <strong>PET</strong>-<strong>CT</strong> findings on<br />

how earlier treatment plans were revised for patients under observation for thyroid cancer<br />

recurrence.<br />

12.1.2. Population characteristics<br />

The subjects <strong>of</strong> both studies were adult patients with thyroid cancer confirmed<br />

histopathologically. All patients were under observation following prior standard treatment<br />

(thyreoidectomy and ablation <strong>of</strong> thyroid gland using iodine 131). The majority <strong>of</strong> patients<br />

(88%) in both Zimmer 2003 and Nahas 2005 had been diagnosed with papillary carcinoma; in<br />

Zimmer 2003, additionally patients with follicular and medullary thyroid cancer took part.<br />

Cancer recurrence was suspected in all the participants <strong>of</strong> Zimmer 2003 based on the above-<br />

limit level <strong>of</strong> thyroglobulin or activity <strong>of</strong> calcitonin in blood, in spite <strong>of</strong> negative results <strong>of</strong><br />

whole-body I 131 scintigraphy. The top limit was: for thyroglobulin in blood - 1.0 ng/mL in<br />

patients treated with suppressive doses <strong>of</strong> thyroid gland hormone; or 10 ng/mL when no<br />

suppressive doses <strong>of</strong> thyroid gland hormone were applied, and for calcitonin activity in<br />

plasma – more than 4 pg/ml.<br />

In Nahas 2005,the authors give no limit values for recurrence markers. The suspicion <strong>of</strong><br />

recurrence was determined based on overall clinical picture and extra tests.<br />

Table 63 shows the initial characteristics <strong>of</strong> the subjects <strong>of</strong> each trial.


Table 63.<br />

Initial characteristics <strong>of</strong> subjects <strong>of</strong> each study<br />

Parameters Zimmer 2003 Nahas 2005<br />

Size <strong>of</strong> population 8 33<br />

Median age (range)<br />

53.6<br />

(SD 20,7)<br />

43.8<br />

(12–72)<br />

Male proportion 12.5% 39.4%<br />

Patients treated by surgery due<br />

to thyroid cancer recurrence<br />

Average number <strong>of</strong> operations<br />

per patient before <strong>PET</strong>-<strong>CT</strong><br />

examination<br />

Patients testing negative in<br />

thyroid scintigraphy<br />

Patients treated with suppressive<br />

doses <strong>of</strong> thyroid gland hormones<br />

Average concentration <strong>of</strong><br />

thyroglobulin in plasma, ng/ml<br />

(from-to)<br />

no data 66.7%<br />

no data 1.5<br />

no data 75.7%<br />

no data 67%<br />

95.2*<br />

(2.5–747)<br />

70.0**<br />

(1.2–629)<br />

* value calculated for 7 patients with various thyroid gland cancer types; a single patient with medullary thyroid<br />

cancer was excluded<br />

** value calculated for 26 patients; in 5 patients thyroglobulin was not markable, in 2 thyroglobulin antibodies were<br />

present<br />

The size <strong>of</strong> population in Zimmer 2003 was 8, and in Nahas 2005 – 33. The median age in<br />

Zimmer 2003 was slightly higher than in Nahas 2005: 53.6 (SD 20.7) vs. 43.8 (between 12 and<br />

72). The male proportion in Zimmer 2003 was 12.5%, and in Nahas 2005 it was 39.4%. In Nahas<br />

2005, 67 % <strong>of</strong> patients were receiving suppressive doses <strong>of</strong> thyroid hormone when they took<br />

<strong>PET</strong>-<strong>CT</strong> tests, whereas Zimmer 2003 informs that all the patients examined had attained<br />

euthyreosis.<br />

12.1.3. Description <strong>of</strong> intervention<br />

12.1.3.1 <strong>PET</strong>-<strong>CT</strong> imaging<br />

In Zimmer 2003, <strong>PET</strong>-<strong>CT</strong> imaging was done with a prototype <strong>PET</strong>-<strong>CT</strong> scanner consisting <strong>of</strong> an<br />

AR.SP spiral <strong>CT</strong> scanner (Siemens Medical Systems, Erlanger, Germany) and an ECAT ART <strong>PET</strong><br />

scanner (<strong>CT</strong>I, Knoxville, TN). A Discovery LS unit made by the General Electric Company was<br />

used in Nahas 2005.<br />

In both studies, deoxy-glucose marked with radioactive F18 fluorine was used as a marker.<br />

In Zimmer 2003, the activity <strong>of</strong> the radiomarker was 260 MBq, and the examination covered<br />

173


the neck and the chest. No data on the activity <strong>of</strong> the marker or the scope <strong>of</strong> the tests are<br />

provided in Nahas 2005.<br />

174<br />

Tests were carried out one hour after FDG was given orally. No iodized contrast medium<br />

was administered intravenously in either <strong>of</strong> the trials. <strong>CT</strong> scanning was followed by <strong>PET</strong>. Two-<br />

dimensional (2D) visualization was used in <strong>PET</strong> examinations.<br />

Table 64 provides detailed information on the type <strong>of</strong> scanner and method used.<br />

Table 64.<br />

Description <strong>of</strong> scanner and method used for <strong>PET</strong>-<strong>CT</strong> examination.<br />

Study<br />

Zimmer 2003<br />

Type <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

Prototype<br />

scanner<br />

Nahas 2005 Discovery LS<br />

Manufactrer <strong>of</strong><br />

<strong>PET</strong>-<strong>CT</strong> scanner<br />

-<br />

General<br />

Electric<br />

Type <strong>of</strong><br />

radiomarker<br />

FDG<br />

intravenously<br />

FDG<br />

intravenously<br />

Coverage<br />

Neck and<br />

chest<br />

Activity <strong>of</strong><br />

radiomarker<br />

[MBq]<br />

260<br />

No data No data<br />

In both cases <strong>PET</strong>-<strong>CT</strong> tests were evaluated by specialists blinded to the patients' clinical<br />

condition and other test results. A team consisting <strong>of</strong> nuclear medicine and radiology<br />

specialists determined the results by consensus. The marker uptake by thyroid gland<br />

parenchyma was assessed visually and quantitatively.<br />

In Zimmer 2003, the patients who tested negative in <strong>PET</strong>-<strong>CT</strong> examination were under<br />

observation for disease recurrence; serial follow-up <strong>PET</strong>-<strong>CT</strong> scans and who-body scintigraphy<br />

were done.<br />

The authors <strong>of</strong> the studies do not report on the duration <strong>of</strong> observation.<br />

12.1.3.2 Diagnostic technologies compared<br />

12.1.3.2.1 <strong>CT</strong>/MRI<br />

Zimmer 2003 provides no data on the procedures <strong>of</strong> computer tomography (<strong>CT</strong>) or<br />

magnetic resonance imaging (MRI), or on the type <strong>of</strong> scanner used or time interval to the<br />

<strong>PET</strong>-<strong>CT</strong> study.<br />

12.1.3.2.2 I131 whole-body scintigraphy (I131 WBS)<br />

Neither <strong>of</strong> the studies provides details <strong>of</strong> the method <strong>of</strong> I 131 whole-body scintigraphy (I 131<br />

WBS) or the time interval between this tests and <strong>PET</strong>-<strong>CT</strong>.


12.1.3.3 Reference Test<br />

In both trials, positive results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> studies were verified based on histopathological tests<br />

<strong>of</strong> material from surgeries. 3 patients (37.5%) in Zimmer 2003, and 20 (61%) patients in Nahas<br />

2005 had had a surgery.<br />

The remaining patients were under long-term clinical observation to confirm negative<br />

results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>; they had I 131 whole-body scintigraphy and <strong>PET</strong>-<strong>CT</strong> examinations. Neither <strong>of</strong><br />

the trials gives details <strong>of</strong> the duration <strong>of</strong> observation for the cohort examined.<br />

12.1.4. Findings<br />

In Zimmer 2003, the authors evaluated the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting<br />

disease recurrences in a group <strong>of</strong> eight patients with thyroid cancer confirmed<br />

histopathologically. For four patients, <strong>PET</strong>-<strong>CT</strong> examinations were preceded by <strong>CT</strong> imaging that<br />

revealed a s<strong>of</strong>t tissues asymmetry, which suggested disease recurrence.<br />

In three patients with papillary thyroid cancer and one patient with medullary thyroid<br />

cancer, <strong>PET</strong>-<strong>CT</strong> detected eleven focuses <strong>of</strong> heightened marker uptake. Seven focuses <strong>of</strong><br />

heightened marker uptake were identified in three patients, for whom postoperative material<br />

was verified histopathologically. One patient had not been operated on but qualified for<br />

chemotherapy based on <strong>PET</strong>-<strong>CT</strong> scanning.<br />

Features indicative <strong>of</strong> cancer recurrence were found in the postoperative material <strong>of</strong> six<br />

out <strong>of</strong> eight lesions. The authors give no data on the presence or absence <strong>of</strong> recurrence in<br />

the remaining two lesions.<br />

No pathological lesions were found by <strong>PET</strong>-<strong>CT</strong> in four other patients. The results were<br />

confirmed through long-term clinical observation.<br />

Table 65 specifies the number <strong>of</strong> patients with true positive, false positive, false negative<br />

and true negative results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong>/MRI in a group <strong>of</strong> patients with suspected thyroid<br />

cancer recurrence in Zimmer 2003. The table leaves out one patient who could not be<br />

<strong>CT</strong>/MRI-scanned.<br />

Table 65.<br />

Number <strong>of</strong> patients with TP, FP, FN and TN results in Zimmer 2003, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>/MRI<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong>/MRI<br />

TP FP FN TN TP FP FN TN<br />

Zimmer 2003 4 0 0 3 2 2 2 1<br />

175


176<br />

The EBM parameters that characterize the diagnostic efficacy in detecting thyroid cancer<br />

recurrence are presented in Table 66.<br />

Table 66.<br />

EBM parameters <strong>of</strong> diagnostic efficacy in detecting thyroid cancer recurrence, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>/MRI<br />

Parameter<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

LR+<br />

(95% CI)<br />

LR-<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

DOR<br />

(95% CI)<br />

Diagnostic method<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong>/MRI<br />

100%<br />

(40; 100)<br />

100%<br />

(29; 100)<br />

7,2<br />

(0,5; 97,8)<br />

0,1<br />

(0,008; 1,6)<br />

100%<br />

(59; 100)<br />

63,0<br />

(1,0; 4042,1)<br />

* p-value for chi-square test (McNemar’s test) Yates corrected<br />

50%<br />

(7; 93)<br />

33%<br />

(1; 91)<br />

0,7<br />

(0,2; 2,7)<br />

1,5<br />

(0,2; 9,8)<br />

43%<br />

(10; 82)<br />

0,5<br />

(0,02; 11,1)<br />

Statistical<br />

significance <strong>of</strong><br />

difference between<br />

groups compared *<br />

p = 0,48<br />

p = 0,48<br />

-<br />

-<br />

p = 0,13<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity in detecting the thyroid cancer recurrence is 100% (95% CI: 40; 100) and is<br />

higher than <strong>CT</strong>/MRI sensitivity, which is 50% (95% CI: 7; 93). That means that the probability <strong>of</strong><br />

positive results for patients with thyroid cancer recurrence is twice as high for <strong>PET</strong>-<strong>CT</strong> than for<br />

<strong>CT</strong>/MRI, and stands at 100%. The difference is not statistically significant (p = 0.48).<br />

<strong>PET</strong>-<strong>CT</strong> specificity in detecting thyroid cancer recurrence is 100% (95% CI: 29; 100) and is<br />

higher than the specificity <strong>of</strong> whole-body scintigraphy, which is 33% (95% CI: 1; 91). That<br />

means that the probability <strong>of</strong> negative results for patients without thyroid cancer recurrence<br />

is 100% for <strong>PET</strong>-<strong>CT</strong>, and is three times higher than the probability <strong>of</strong> negative results for <strong>CT</strong>/MRI.<br />

The difference is not statistically significant (p = 0.48).<br />

The positive likelihood ratio <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 7.2 (95% CI: 0.5; 97.8) and is higher than that <strong>of</strong><br />

<strong>CT</strong>/MRI- 0.7 (95% CI: 0.2; 2.7). The probability <strong>of</strong> positive results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> for patients with<br />

thyroid cancer recurrence is more than 7 times higher than the probability <strong>of</strong> positive results<br />

for patient with no recurrence. In the case <strong>of</strong> <strong>CT</strong>/MRI, the likelihood <strong>of</strong> positive results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

for patients with thyroid cancer recurrence represents 70% <strong>of</strong> the likelihood for patients with<br />

no thyroid cancer recurrence.<br />

The negative likelihood ratio <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 0.1 (95% CI: 0.008; 1.6) and is lower than that <strong>of</strong><br />

<strong>CT</strong>/MRI, which is 1.5 (95% CI: 0.2; 9.8). In the case <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>, the likelihood <strong>of</strong> negative result for<br />

patients with thyroid cancer recurrence represents 10% <strong>of</strong> the likelihood <strong>of</strong> negative results for<br />

-


patients with no recurrence. In the case <strong>of</strong> <strong>CT</strong>/MRI, the likelihood <strong>of</strong> negative results for<br />

patients with thyroid cancer recurrence is 150% <strong>of</strong> the likelihood for patients with no cancer<br />

recurrence.<br />

The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting thyroid cancer recurrence is 100% (95% CI: 59; 100)<br />

and is higher than <strong>CT</strong>/MRI accuracy, which is 43% (95% CI: 10; 82). The proportion <strong>of</strong> people<br />

(both with and without the disease) diagnosed correctly by <strong>PET</strong>-<strong>CT</strong> is 100% and more than<br />

twice as high as for <strong>CT</strong>/MRI; however, the difference observed as evaluated using<br />

McNemar’s test is not statistically significant (p = 0.13).<br />

The diagnostic odds ratio for <strong>PET</strong>-<strong>CT</strong> is 63.0 (95% CI: 1.0; 4042.1), and is higher than the odds<br />

ratio for <strong>CT</strong>/MRI, which is 0.5 (95% CI: 0.02; 11.1). That means that the probability <strong>of</strong> thyroid<br />

cancer for patients with positive <strong>PET</strong>-<strong>CT</strong> results is 63 times higher than the probability for<br />

patients with negative results. However, the probability <strong>of</strong> positive <strong>CT</strong>/MRI results represents<br />

50% <strong>of</strong> the probability for patients with negative result <strong>of</strong> <strong>CT</strong>/MRI imaging.<br />

<strong>PET</strong>-<strong>CT</strong> efficacy in detecting thyroid cancer recurrence was compared to iodine 131<br />

whole-body scintigraphy in Zimmer 2003 and Nahas 2005. Zimmer 2003 included patients with<br />

negative results <strong>of</strong> I 131 whole-body scintigraphy and raised concentration level <strong>of</strong> cancer<br />

markers (thyroglobulin, calcitonin) in blood serum. In Nahas 2005, serial imaging and<br />

biochemical test were done for all the patients at the follow-up stage <strong>of</strong> standard cancer<br />

treatment. 20 subjects (61%) <strong>of</strong> that trial were classified based on the overall clinical picture<br />

and additional examinations. Postoperative material was verified by an experienced<br />

histopathologist. 36 anatomical locations were identified and assessed by standard<br />

histopathological tests. They were located mainly in the thyroid bed and lateral neck<br />

neighborhood. The authors provide results <strong>of</strong> histopathological verification <strong>of</strong> each lesion for<br />

<strong>PET</strong>-<strong>CT</strong> only, without specifying their scintigraphic characteristics, therefore, for the purposes<br />

<strong>of</strong> this <strong>analysis</strong>, true positive, false positive, false negative and true negative results are<br />

presented for each patient.<br />

The number <strong>of</strong> patients assigned to each group in both studies included is presented in the<br />

Table 67.<br />

Table 67.<br />

Number <strong>of</strong> patients with TP, FP, FN and TN results in both studies, <strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body scintigraphy<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong> I 131 whole-body scintigraphy<br />

TP FP FN TN TP FP FN TN<br />

Zimmer 2003 4 0 0 4 0 0 4 4<br />

177


178<br />

Nahas 2005 16 0 3 1 4 0 15 1<br />

Table 68 presents sensitivity values calculated for each study (<strong>PET</strong>-<strong>CT</strong> and I 131 whole-body<br />

scintigraphy).<br />

Table 68.<br />

Sensitivity calculated for each study, <strong>PET</strong>-<strong>CT</strong> vs I 131 whole-body scintigraphy<br />

Study<br />

Se<br />

(95% CI)<br />

<strong>PET</strong>-<strong>CT</strong> I 131 whole-body scintigraphy<br />

Zimmer 2003 100% 0%<br />

Nahas 2005<br />

84%<br />

(60; 97)<br />

21%<br />

(6; 46)<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting thyroid cancer recurrence is bigger than the sensitivity<br />

<strong>of</strong> the whole-body scintigraphy in both studies. In view <strong>of</strong> the type <strong>of</strong> data available, the<br />

confidence interval for scintigraphy findings in Zimmer 2003 cannot be calculated.<br />

Graph 24 illustrates a meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> sensitivity in detecting thyroid cancer<br />

recurrence in both studies included.<br />

Graph 24.<br />

Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> sensitivity in detecting thyroid cancer recurrence<br />

Nahas 2005 0.84 (0.60; 0.97)<br />

Zimmer 2003 1.00 (0.60, 1.00)<br />

Result <strong>of</strong> meta-<strong>analysis</strong> [fixed] 0.87 (0.66; 0.97)<br />

0,0 0,2 0,4 0,6 0,8 1,0<br />

Sensitivity (95% confidence interval)


The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is calculated at 87% (95% CI: 66; 97). The probability <strong>of</strong> positive<br />

results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 87% for patients with recurrence.<br />

Table 69 presents the data accumulated with respect to the sensitivity <strong>of</strong> the methods<br />

compared in recurrence evaluation.<br />

Table 69.<br />

Comparison <strong>of</strong> sensitivity in evaluating recurrence, <strong>PET</strong>-<strong>CT</strong> vs I 131 whole-body scintigraphy<br />

Study<br />

Se<br />

(95% CI)<br />

Diagnostic method<br />

<strong>PET</strong>-<strong>CT</strong> I 131 whole-body scintigraphy<br />

87%<br />

(66; 97)<br />

0–21%<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 87% (95% CI: 66; 97). The probability <strong>of</strong> positive results with <strong>PET</strong>-<strong>CT</strong><br />

scanning is 87% for patients with disease recurrence. Calculated for each <strong>of</strong> the studies<br />

included separately, the sensitivity <strong>of</strong> I 131 whole-body scintigraphy ranges from 0 to 20%.<br />

Table 70 presents specificity values for the methods compared (<strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body<br />

scintigraphy), as calculated separately for both studies included.<br />

Table 70.<br />

Comparison <strong>of</strong> specificity, <strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body scintigraphy<br />

study<br />

Zimmer 2003<br />

Nahas 2005<br />

Sp<br />

(95% CI)<br />

<strong>PET</strong>-<strong>CT</strong> I 131 whole-body scintigraphy<br />

100%<br />

(40; 100)<br />

100%<br />

(3; 100)<br />

100%<br />

(40; 100)<br />

100%<br />

(3; 100)<br />

The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting thyroid cancer recurrence is in both studies<br />

comparable with the specificity <strong>of</strong> whole-body scintigraphy.<br />

For both imaging technologies, the probability <strong>of</strong> negative results for patients with no<br />

thyroid cancer recurrence is 100%.<br />

Table 71 presents diagnostic accuracy values for the methods compared (<strong>PET</strong>-<strong>CT</strong> vs. I 131<br />

whole-body scintigraphy), as calculated separately for both studies included.<br />

179


Table 71.<br />

comparison <strong>of</strong> diagnostic accuracy, <strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body scintigraphy<br />

180<br />

Study<br />

Zimmer 2003<br />

Nahas 2005<br />

Acc<br />

(95% CI)<br />

<strong>PET</strong>-<strong>CT</strong> I 131 whole-body scintigraphy<br />

100%<br />

(63; 100)<br />

85%<br />

(62; 97)<br />

50%<br />

(16; 84)<br />

25%<br />

(9; 49)<br />

The diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is in both cases higher than the diagnostic accuracy <strong>of</strong><br />

whole-body scintigraphy.<br />

The diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in evaluating cancer recurrence as calculated by<br />

meta-<strong>analysis</strong> <strong>of</strong> two trials is presented in graph 25.<br />

Graph 25.<br />

Meta-<strong>analysis</strong> <strong>of</strong> diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

Zimmer 2003 1.00 (0.63, 1.00)<br />

Nahas 2005 0.85 (0.62, 0.97)<br />

Meta-<strong>analysis</strong> result [mixed]<br />

0,5 0,6 0,7 0,8 0,9 1,0<br />

Diagnostic accuracy (95% confidence range)<br />

Graph 26 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> I 131 whole-body<br />

scintigraphy in detecting thyroid cancer recurrence.<br />

0.89 (0.75, 0.97)


Graph 26.<br />

Meta-<strong>analysis</strong> <strong>of</strong> diagnostic accuracy <strong>of</strong> I 131 whole-body scintigraphy<br />

Zimmer 2003 0.50 (0.16; 0.84)<br />

Nahas 2005 0.25 (0.09; 0.49)<br />

Meta-<strong>analysis</strong> result<br />

[fixed]<br />

0,0 0,3 0,6 0,9<br />

Table 72 presents the diagnostic accuracy <strong>of</strong> the methods compared (<strong>PET</strong>-<strong>CT</strong> vs. I 131<br />

whole-body scintigraphy) calculated by meta-<strong>analysis</strong> <strong>of</strong> data provided in both trials.<br />

Table 72.<br />

Diagnostic accuracy in evaluating disease recurrence, <strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body scintigraphy<br />

Test<br />

Acc<br />

(95% CI)<br />

Diagnostic result (95% confidence interval)<br />

Diagnostic method<br />

<strong>PET</strong>-<strong>CT</strong> I 131 whole-body scintigraphy<br />

89%<br />

(75; 97)<br />

33%<br />

(18; 50)<br />

The meta-analyzed diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting thyroid cancer recurrence<br />

is 89% (95% CI: 75; 97) and is higher than the accuracy <strong>of</strong> scintigraphy, which is 33% (95% CI:<br />

18; 50). The proportion <strong>of</strong> people (both <strong>of</strong> sick and healthy) diagnosed correctly is approx. 56<br />

pp. higher for <strong>PET</strong>-<strong>CT</strong> than for I 131 whole-body scintigraphy.<br />

Table 73 presents other EBM parameters that characterize the diagnostic efficacy <strong>of</strong> the<br />

imaging methods compared (<strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body scintigraphy), as calculated by<br />

meta-<strong>analysis</strong> <strong>of</strong> data from both trials.<br />

0.33 (0.18\; 0.50)<br />

181


Table 73.<br />

EBM parameters <strong>of</strong> diagnostic efficacy <strong>of</strong> imaging methods compared, <strong>PET</strong>-<strong>CT</strong> vs I 131 whole-body scintigraphy<br />

182<br />

Parameters<br />

LR+<br />

(95% CI)<br />

LR-<br />

(95% CI)<br />

DOR<br />

(95% CI)<br />

Diagnostic method<br />

<strong>PET</strong>-<strong>CT</strong> I 131 whole-body scintigraphy<br />

5.2<br />

(0.9; 30.9)<br />

0.2<br />

(0.1; 0.6)<br />

28.6<br />

(2.1; 395.7)<br />

1.4<br />

(0.2; 95)<br />

0.9<br />

(0.6; 1.4)<br />

1.6<br />

(0.14; 17.7)<br />

The positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 5.2 (95% CI: 0.9; 30.9), and is higher than the ratio<br />

for I 131 whole-body scintigraphy, which is 1.4 (95% CI: 0.2; 9.5). The probability <strong>of</strong> positive <strong>PET</strong>-<br />

<strong>CT</strong> test results for a patient with thyroid cancer recurrence is 5 times higher than the likelihood<br />

<strong>of</strong> positive results for a patient with no such recurrence. For scintigraphy, the likelihood <strong>of</strong><br />

positive results is almost 1.5 times higher for patients with thyroid cancer recurrence than for<br />

patients without cancer recurrence.<br />

The negative likelihood ratio calculated for <strong>PET</strong>-<strong>CT</strong> is 0.2 (95% CI: 0.1; 0.6), and is lower than<br />

the ratio for whole-body scintigraphy, which is 0.9 (95% CI: 0.6; 1.4). With <strong>PET</strong>-<strong>CT</strong>, the<br />

probability <strong>of</strong> negative results for patients diagnosed with thyroid cancer recurrence<br />

represents 20% <strong>of</strong> the probability for patients without cancer. With scintigraphy, the<br />

probability <strong>of</strong> negative results for patients diagnosed with thyroid cancer represents 90% <strong>of</strong><br />

the probability for patients without thyroid cancer.<br />

The diagnostic odds ratio for <strong>PET</strong>-<strong>CT</strong> is 28.6 (95% CI: 2.1; 395.7), and is higher than the ratio<br />

for whole-body scintigraphy, which is 1.6 (95% CI: 0.14; 17.7). That means that the probability<br />

<strong>of</strong> cancer in patients testing positive with <strong>PET</strong>-<strong>CT</strong> is more than 28 times higher than the<br />

probability for patients testing negative. The probability <strong>of</strong> thyroid cancer in patients testing<br />

positive with <strong>PET</strong>-<strong>CT</strong> is more than 1.5 times higher than the probability for patients testing<br />

negative in I 131 whole-body scintigraphy tests.<br />

12.1.4.1 Safety<br />

The authors <strong>of</strong> the studies included do not provide data on undesirable effects <strong>of</strong> the use<br />

<strong>of</strong> <strong>PET</strong>-<strong>CT</strong> imaging in the trial population.<br />

12.1.5. Impact on therapy<br />

The authors <strong>of</strong> Nahas 2005 analyzed the impact <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> findings on the previous<br />

therapeutic policy for 33 patients who were under observation following standard treatment


<strong>of</strong> papillary thyroid cancer. Physical examination <strong>of</strong> the head and neck was done<br />

repeatedly, with special emphasis on palpation, examination <strong>of</strong> vocal cords flexibility and<br />

examination <strong>of</strong> cranial nerves in order to detect possible recurrences.<br />

<strong>PET</strong>-<strong>CT</strong> tests caused previous treatment plans to be revised for 13 patients, and confirmed<br />

the adopted treatment plans for 9 patients. In total, for 22 patients (67%) <strong>PET</strong>-<strong>CT</strong> yielded<br />

additional information that influenced their therapy. The authors do not specify what revisions<br />

in treatment resulted from <strong>PET</strong>-<strong>CT</strong> findings.<br />

12.1.6. Results<br />

As a result <strong>of</strong> searching through medical databases two primary clinical studies were found<br />

(Zimmer 2003, Nahas 2005), in which <strong>PET</strong>-<strong>CT</strong> was compared to conventional diagnostics<br />

(<strong>CT</strong>/MRI and iodine 131 whole-body scintigraphy - WBS) in detecting thyroid cancer (N = 41).<br />

The imaging methods compared were verified based on histopathological tests, while for<br />

negative results verification was based on long-term clinical observation and a series <strong>of</strong><br />

diagnostic imaging.<br />

In detecting recurrences <strong>of</strong> thyroid cancer, the sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 100% (95% CI: 40; 100),<br />

i.e. twice the sensitivity <strong>of</strong> <strong>CT</strong>/MRI, 50% (95% CI: 7; 93). The difference is not statistically<br />

significant (p = 0.48). In detecting recurrences <strong>of</strong> thyroid cancer, <strong>PET</strong>-<strong>CT</strong> is characterized by a<br />

higher specificity than <strong>CT</strong> and MRI. The calculated values were 100% (95% CI: 29; 100) vs. 33%<br />

(95% CI: 1; 91) respectively. The difference is not statistically significant (p = 0.48). The<br />

diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is higher than that <strong>of</strong> <strong>CT</strong>/MRI, the values being 100% (95% CI:<br />

59; 100) vs. 43% (95% CI: 10; 82) respectively. The difference is not statistically significant (p =<br />

0.13).<br />

Based on a comparison <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with iodine 131 whole body scintigraphy (I 131 WBS), the<br />

imaging sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is higher than that <strong>of</strong> scintigraphy. The established value is 87%<br />

(95% CI: 66; 97) for <strong>PET</strong>-<strong>CT</strong>, but for I 131 WBS it is 0 and 21%. In detecting thyroid cancer<br />

recurrences, specificity is identical for <strong>PET</strong>-<strong>CT</strong> and whole-body scintigraphy and stands at<br />

100%. Similarly, in detecting recurrences <strong>of</strong> thyroid cancer, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

is 89% (95% CI: 75; 97) and is higher than that <strong>of</strong> I 131 WBS, which stands at 33% (95% CI: 18; 50).<br />

In one <strong>of</strong> the studies included, the authors reported that <strong>PET</strong>-<strong>CT</strong> scanning supported early<br />

revisions <strong>of</strong> treatment policies in 22 patients (67%). These patients were under observation<br />

following standard treatment <strong>of</strong> thyroid cancer. Additionally, in some patients, the adopted<br />

treatment policy was confirmed by the data collected from <strong>PET</strong>-<strong>CT</strong> scanning.<br />

In summary, <strong>PET</strong>-<strong>CT</strong> is characterized by a higher diagnostic efficacy in detecting<br />

recurrences <strong>of</strong> thyroid cancer than convectional imaging methods.<br />

183


184<br />

12.2. Comparative efficacy <strong>analysis</strong> <strong>of</strong> f pet-ct vs. traditional<br />

imaging methods (<strong>CT</strong>, I131 whole-body scintigraphy and<br />

USG) in the staging <strong>of</strong> differentiated thyroid cancer<br />

12.2.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching medical databases, one primary prospective clinical study was<br />

found (Freudenberg 2004; tab. 152, app. 18.1), which compared the diagnostic efficacy <strong>of</strong><br />

<strong>PET</strong>-<strong>CT</strong> with other imaging methods such as X-ray <strong>CT</strong>, I 131 whole-body scintigraphy and<br />

ultrasound examination) in the primary staging <strong>of</strong> differentiated thyroid cancer.<br />

12.2.2. Population characteristics<br />

The subjects in Freudenberg 2004 were twelve patients with differentiated thyroid cancer,<br />

who had been qualified for ablation <strong>of</strong> thyroid gland with iodine 131 . The mean age <strong>of</strong> the<br />

patients was 59 (ranging from 31 to 76); men represented 58.3% <strong>of</strong> the population. Three <strong>of</strong><br />

them had been diagnosed histopathologically with follicular thyroid cancer, nine with<br />

papillary thyroid cancer. Eleven patients were examined at the postoperative stage following<br />

thyroidectomy under maximum TSH stimulation (the average activity <strong>of</strong> hormone in blood<br />

was 52 mU/l, SD 21). One patient, in whom lung metastasis had been detected, was<br />

examined after exogenous stimulation with human TSH. The level <strong>of</strong> urinary secretion <strong>of</strong> iodine<br />

was found to be within the physiological limits in all the patients (128 µg/l, SD 50). In six<br />

patients, thyroglobulin concentration in blood was high; none <strong>of</strong> them was diagnosed with a<br />

high concentration level <strong>of</strong> thyroid antibodies.<br />

Table 74 shows the initial characteristics <strong>of</strong> the subjects.<br />

Table 74.<br />

Initial characteristics <strong>of</strong> patients examined<br />

Parameter Freudenberg 2004<br />

Size <strong>of</strong> population 12<br />

Median age (from - to)<br />

59<br />

(31–76)<br />

Male proportion 58.3%<br />

Patients after thyroidectomy 100%<br />

Patients with high concentration level <strong>of</strong><br />

thyroglobulin<br />

50%


Concentration <strong>of</strong> thyroglobulin in blood serum,<br />

ng/ml (fro - to)<br />

Patients diagnosed under maximum TSH<br />

stimulation<br />

Patients with pT1 / pT2 / pT3 / pT4** features<br />

according to the AJCC classification<br />

279*<br />

(0.4–122)<br />

91.6%<br />

0/33%/17%/50%<br />

* Value calculated for 11 patients<br />

** pT1 – tumor


5 times or more was deemed to indicate a tumor or persistent thyroid tissue. This criterion was<br />

confirmed histopathologically in 6 cases (for 69 lesions).<br />

186<br />

12.2.3.2 Diagnostic technology compared<br />

The authors report that all the diagnostic imaging to stage the disease was executed<br />

within a two-week period. Two experienced radiologists interpreted the findings and set the<br />

results via consensus.<br />

12.2.3.2.1 Computer tomography<br />

Computer tomography scans were executed in a standard way; no iodized contrast<br />

medium was administered intravenously. The authors provide no details <strong>of</strong> the type <strong>of</strong><br />

scanner.<br />

12.2.3.2.2 I131 whole-body scintigraphy<br />

High-dose I 131 whole-body scintigraphy (I131 WBS) was done using a two-head gamma<br />

camera. A Bodyscan unit manufactured by Siemens (Erlangen, Germany) was used in the<br />

tests. Examination was carried out within 5–8 days after iodine 131 was administered;<br />

radiomarker activity was 3000 MBq.<br />

12.2.3.2.3 Ultrasound examination <strong>of</strong> the neck<br />

Ultrasound Sonoline Elegra scanner (Siemens, Erlangen, Germany) was used for ultrasound<br />

examinations. The Görgesa criteria, which cover the Solbiati-Index (relation <strong>of</strong> the biggest to<br />

the smallest node size) were applied to stage node involvement, to determine the internal<br />

structure <strong>of</strong> the node and vascularization.<br />

12.2.3.3 Reference test<br />

Histopathological test was used to validate the results for six lesions (8.6%). Wherever such<br />

data were not available, a team <strong>of</strong> specialists (two <strong>of</strong> each: radiologists and nuclear<br />

medicine doctors) worked out a consensus based on all the clinical data available as well as<br />

results <strong>of</strong> diagnostic imaging (including results <strong>of</strong> I 131 WBS and USG), for each patient<br />

separately, with respect to the presence or absence <strong>of</strong> disease, as well as the number and<br />

location <strong>of</strong> changes.<br />

12.2.4. Findings<br />

Freudenberg 2004 presents efficacy details <strong>of</strong> the imaging methods compared, with<br />

respect to disease staging. The number <strong>of</strong> lesions was determined in each location, and the<br />

diagnostic efficacy was evaluated versus the reference method.<br />

Table 76 gives the number <strong>of</strong> lesions detected with the use <strong>of</strong> each imaging method,<br />

along with the results <strong>of</strong> test diagnostic efficiency evaluation.


Table 76.<br />

Number <strong>of</strong> lesions detected with the imaging methods compared vs. the reference test for each feature <strong>of</strong> the TNM<br />

staging system.<br />

Parameter<br />

Primary<br />

tumor (T)<br />

Metastasis to<br />

lymph nodes<br />

(N)<br />

Distant<br />

metastasis<br />

(M)<br />

I 124 <strong>PET</strong>-<strong>CT</strong> I 131 WBS <strong>CT</strong> USG<br />

TP FP FN TN TP FP FN TN TP FP FN TN TP FP FN TN<br />

17 0 0 - 17 0 0 - 6 1 11 - 8 0 9 - 17<br />

6 0 0 - 5 0 1 - 3 1 3 - 2 0 4 - 6<br />

46 1 0 - 35 2 11 - 30 2 16 - - - - - 46<br />

Total 69 1 0 - 57 2 12 - 39 4 30 - - - - - 69<br />

* Re – reference method<br />

Based on data available, the sensitivity <strong>of</strong> each imaging method in lesion staging was<br />

calculated for individual features and for features total. The data are compiled in Table 77.<br />

Table 77<br />

Diagnostic sensitivity in lesion staging for each feature <strong>of</strong> the TNM system: I 124 <strong>PET</strong>-<strong>CT</strong> vs. traditional methods (<strong>CT</strong>, I 131<br />

WBS, USG)<br />

Imaging<br />

method<br />

I 124 <strong>PET</strong>-<strong>CT</strong><br />

I 131 WBS<br />

<strong>CT</strong><br />

USG<br />

Diagnostic sensitivity in lesions staging in individual<br />

locations (95% CI)<br />

T N M<br />

100%<br />

(80; 100)<br />

100%<br />

(80; 100)<br />

35%<br />

(14; 62)<br />

47%<br />

(23; 72)<br />

100%<br />

(54; 100)<br />

83%<br />

(36; 100)<br />

50%<br />

(12; 88)<br />

33%<br />

(4; 78)<br />

* p-value for the chi-square (McNemar’s) test Yates corrected<br />

100%<br />

(92; 100)<br />

76%<br />

(61; 87)<br />

65%<br />

(50; 79)<br />

-<br />

Diagnostic<br />

sensitivity in<br />

lesion staging<br />

irrespective <strong>of</strong><br />

location<br />

100%<br />

(94; 100)<br />

83%<br />

(72; 91)<br />

57%<br />

(44; 68)<br />

43%<br />

(23; 66)<br />

Re<br />

*<br />

Statistic<br />

significance <strong>of</strong><br />

difference:<br />

<strong>PET</strong>-<strong>CT</strong> vs.<br />

method<br />

compared *<br />

(lesion staging<br />

irrespective <strong>of</strong><br />

location)<br />

-<br />

p < 0,01<br />

p < 0,01<br />

p < 0,01<br />

The sensitivity <strong>of</strong> I 124 <strong>PET</strong>-<strong>CT</strong> in T-staging is 100% (95% CI: 80; 100), and is the same for I 131 WBS.<br />

The probability <strong>of</strong> detecting a primary tumor is 100% for I 124 <strong>PET</strong>-<strong>CT</strong>, and is higher than that for<br />

<strong>CT</strong> and USG; the sensitivity <strong>of</strong> these imaging methods is 35% (95% CI: 14; 62) and 47% (95% CI:<br />

23; 72) respectively.<br />

The sensitivity <strong>of</strong> I 124 <strong>PET</strong>-<strong>CT</strong> in N-staging is 100% (95% CI: 54; 100), and is higher than the<br />

sensitivity <strong>of</strong> traditional methods; the sensitivity <strong>of</strong> I 131 WBS, <strong>CT</strong> and USG is 83%(95% CI: 36; 100),<br />

50% (95% CI: 12; 88) and 33% (95% CI: 4; 78) respectively. That means that the likelihood <strong>of</strong> in<br />

187


detecting lesions in this location is 100% with I 124 <strong>PET</strong>-<strong>CT</strong>, while for I 131 WBS, <strong>CT</strong> and USG the<br />

values are 83%, 50% and 33% respectively.<br />

188<br />

I 124 <strong>PET</strong>-<strong>CT</strong> is characterized by higher sensitivity in detecting distant metastasis than I 131 WBS<br />

or USG; the likelihood <strong>of</strong> detecting distant metastasis using I 124 <strong>PET</strong>-<strong>CT</strong> is 100% (95% CI: 92; 100),<br />

whereas for I 131 WBS and <strong>CT</strong>, sensitivity is 76% (95% CI: 61; 87) and 65% (95% CI: 50; 79)<br />

respectively.<br />

Another finding was that the combined sensitivity <strong>of</strong> I 124 <strong>PET</strong>-<strong>CT</strong> was higher in comparison to<br />

the traditional methods (I 131 WBS, <strong>CT</strong> and USG) in thyroid cancer staging. The likelihood <strong>of</strong><br />

detecting a reference test-validated lesion, irrespective <strong>of</strong> location, is 100%(95% CI: 94; 100)<br />

with I 124 <strong>PET</strong>-<strong>CT</strong>, and is higher than with I 131 WBS, <strong>CT</strong> and USG; the sensitivity values calculated<br />

for these methods are: 83% (95% CI: 72; 91), 57% (95% CI: 44; 68) and 43% (95% CI: 23; 66)<br />

respectively. The differences between <strong>PET</strong>-<strong>CT</strong> and each <strong>of</strong> the methods compared are<br />

statistically significant (p < 0.01; McNemar's test).<br />

12.2.4.1 Safety<br />

The study provides no information on any undesirable effects <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> imaging in the cohort.<br />

12.2.5. Results<br />

As a result <strong>of</strong> searching through medical databases, one primary prospective trial was<br />

found (Freudenberg 2004) that compared the diagnostic efficacy <strong>of</strong> I 124 <strong>PET</strong>-<strong>CT</strong> imaging with<br />

convectional diagnostic methods (<strong>CT</strong>, I 131 whole-body scintiscanning and ultrasound<br />

scanning) in the staging <strong>of</strong> thyroid cancer (N = 12). All tests were verified based on<br />

histopathological diagnostics and through formal consensus based on diagnostic imaging<br />

results.<br />

In tumor staging (T-status), the sensitivity <strong>of</strong> I 124 <strong>PET</strong>-<strong>CT</strong> is 100% (95% CI: 80; 100), and is the<br />

same as the sensitivity <strong>of</strong> I 131 whole-body scintigraphy (I 131 WBS), but higher than the sensitivity<br />

<strong>of</strong> <strong>CT</strong> and ultrasound scanning, for which the values are 35% (95% CI: 14; 62) and 47% (95% CI:<br />

23; 72) respectively.<br />

In node involvement staging (N-status), I 124 <strong>PET</strong>-<strong>CT</strong> proved to be the most sensitive. The<br />

values are: 100% for <strong>PET</strong>-<strong>CT</strong> (95% CI: 54; 100), 83% for I 131 WBS (95% CI: 36; 100), 50% for <strong>CT</strong> (95%<br />

CI: 12; 88) and 33% for ultrasonography (95% CI: 4; 78).<br />

In distant metastasis staging (M-status), I 124 <strong>PET</strong>-<strong>CT</strong> is characterized by a higher sensitivity<br />

than I 131 WBS or ultrasonography. The calculated values are 100% (95% CI: 92; 100), 76% (95%<br />

CI: 61; 87) and 65% (95% CI: 50; 79) respectively.<br />

The combined sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> imaging in clinical staging <strong>of</strong> thyroid cancer, irrespective<br />

<strong>of</strong> location, is 100% (95% CI: 94; 100) and is higher than the sensitivity <strong>of</strong> I 131 WBS, <strong>CT</strong> or


ultrasound scanning. The values established for these methods are 83% (95% CI: 72; 91), 57%<br />

(95% CI: 44; 68) and 43% (95% CI: 23; 66) respectively. The differences between <strong>PET</strong>-<strong>CT</strong> and<br />

each <strong>of</strong> the scanning method compared are statistically significant (p < 0.01).<br />

In summary, I 124 <strong>PET</strong>-<strong>CT</strong> is characterized by a higher diagnostic efficacy than the<br />

conventional imaging methods in T-staging <strong>of</strong> differentiated thyroid cancer.<br />

189


13. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS.<br />

TRADITIONAL IMAGING METHODS IN HEAD AND NECK<br />

CANCER DIAGNOSTICS<br />

190<br />

13.1. Diagnostics <strong>of</strong> malignant head and neck tumors (primary<br />

lesions, recurrence after treatment, cervical lymph node<br />

metastasis from an unknown primary focus)<br />

13.1.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching medical databases a single primary trial was found (Branstetter<br />

2005; tab. 155, app. 18.1) that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>of</strong> <strong>CT</strong><br />

in the diagnostics <strong>of</strong> malignant head and neck tumors (primary lesions, recurrences after<br />

treatment, cervical lymph node metastasis from an unknown primary focus).<br />

13.1.2. Population characteristics<br />

Branstetter 2005 is a prospective clinical study on a sample <strong>of</strong> 65 patients: their neck and<br />

head tumors were scanned using the <strong>PET</strong>-<strong>CT</strong> technology. The patients were referred to<br />

examination:<br />

a. for cancer staging (11 patients),<br />

b. for detecting recurrences during treatment (46 patients),<br />

c. for locating unknown primary foci in patients with lymph node involvement (8 patients).<br />

Table 78 presents the characteristics <strong>of</strong> the subject population.<br />

Table 78.<br />

Characteristics <strong>of</strong> subject population in Branstetter 2005<br />

Female<br />

Male<br />

Purpose [people]<br />

Parameter Branstetter 2005<br />

Size <strong>of</strong> population 65<br />

Median age (from – to) [years] 63 (43–83)<br />

Number [persons] 23<br />

Median age (from – to) [years] 61 (43–83)<br />

Number [persons] 42<br />

Median age (from – to) [years] 64 (46–82)<br />

Staging 11<br />

Detection <strong>of</strong> recurrence 46<br />

Detection <strong>of</strong> primary focus 8


13.1.3. Description <strong>of</strong> intervention<br />

13.1.3.1 <strong>PET</strong>-<strong>CT</strong> imaging<br />

<strong>PET</strong>-<strong>CT</strong> scanning was done using two combined scanners: EXA<strong>CT</strong> HR+ (CPS Innovations)<br />

and Somatom Emotion (Siemens Medical Solutions); Reveal (<strong>CT</strong>I Medical Systems). <strong>PET</strong>-<strong>CT</strong><br />

scanning from the cranial base to hypogastrium was done 1 hour following intravenous<br />

administration <strong>of</strong> 8-15 mCi (296–555 MBq) <strong>of</strong> FDG. An intravenous contrast medium was used<br />

in the <strong>CT</strong> component <strong>of</strong> the examination for all the patients without contraindications.<br />

Attenuation was corrected using the <strong>CT</strong> component.<br />

<strong>PET</strong> and <strong>CT</strong> scans were interpreted by groups <strong>of</strong> radiologists and nuclear medicine<br />

specialists working separately, who were blinded to the findings <strong>of</strong> the other imaging test. The<br />

researchers interpreting combined <strong>PET</strong>-<strong>CT</strong> images had access to complete data.<br />

The localization was found radiologically incorrect if tumor was suspected or could not be<br />

rationally excluded.<br />

13.1.3.2 Diagnostic technology compared<br />

<strong>CT</strong> image from <strong>PET</strong>-<strong>CT</strong> examination carried out using two combined scanners: EXA<strong>CT</strong> HR+<br />

(CPS Innovations) and Somatom Emotion (Siemens Medical Solutions); Reveal (<strong>CT</strong>I Medical<br />

Systems). An intravenous contrast medium was used in the <strong>CT</strong> component <strong>of</strong> the examination<br />

for all the patients without contraindications.<br />

13.1.3.3 Reference test<br />

The reference test included:<br />

• biopsy,<br />

• imaging examinations (<strong>CT</strong>, <strong>PET</strong>-<strong>CT</strong>),<br />

• clinical observation for at least 6 months; the average period <strong>of</strong> observation was 8.9<br />

months (from 6 to 12 months).<br />

For all the patients with tumor, diagnosis was confirmed with histopathological<br />

examination.<br />

Observation was possible in 64 patients. One was eliminated from the study because the<br />

period <strong>of</strong> observation was too short.<br />

13.1.4. Findings<br />

13.1.4.1 Diagnostic efficacy<br />

In Branstetter 2005, 125 lesions were identified in 58 patients (91%). Out <strong>of</strong> these 125 lesions,<br />

20 were confirmed by biopsy, 15 by additional imaging and 90 were confirmed clinically. No<br />

191


additional lesions were detected through observation except those revealed by <strong>PET</strong>-<strong>CT</strong><br />

imaging. Statistically, both in terms <strong>of</strong> accuracy in lesion diagnostic and with respect to the<br />

patients, <strong>PET</strong>-<strong>CT</strong> turned out considerably more efficacious than <strong>CT</strong> (p < 0.5); results on the<br />

basis <strong>of</strong> comparison <strong>of</strong> fields under ROC curves.<br />

Table 79.<br />

Test results for each <strong>of</strong> the <strong>of</strong> methods compared vs. reference test: Branstetter 2005; per number <strong>of</strong> lesions<br />

192<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

TP FP FN TN TP FP FN TN<br />

Branstetter 2005 45 6 1 73 34 20 12 59<br />

Table 80 presents the efficacy <strong>of</strong> the testing technologies compared in detecting lesions<br />

(per number <strong>of</strong> lesions, n = 125).<br />

Table 80.<br />

Diagnostic efficacy parameters <strong>of</strong> the methods compared in Branstetter 2005 per number <strong>of</strong> lesions<br />

Parameter<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

LR+<br />

(95% CI)<br />

LR-<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

DOR<br />

(95% CI)<br />

Branstetter 2005<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

98%<br />

(88; 100)<br />

92%<br />

(84; 97)<br />

12,88<br />

(5,96; 27,83)<br />

0,02<br />

(0,003; 0,16)<br />

94%<br />

(89; 98)<br />

547,50<br />

(63,82; 4697,20)<br />

74%<br />

(59; 86)<br />

75%<br />

(64; 84)<br />

2,92<br />

(1,93; 4,43)<br />

0,35<br />

(0,21; 0,58)<br />

74%<br />

(66; 82)<br />

8,36<br />

(3,64; 19,18)<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 98% (95% CI: 88; 100) and the sensitivity <strong>of</strong> <strong>CT</strong> was 74% (95% CI:<br />

59; 86). That means that <strong>PET</strong>-<strong>CT</strong> detected lesions correctly in 98% cases, whereas <strong>CT</strong> detected<br />

only 74% <strong>of</strong> all the cancer lesions.<br />

The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 92% (95% CI: 84; 97), and the specificity <strong>of</strong> <strong>CT</strong> was 75% (95%<br />

CI: 64; 84). That means that <strong>PET</strong>-<strong>CT</strong> test results were negative in 92% <strong>of</strong> patients without<br />

cancer tumor. <strong>CT</strong> test results were negative only in 75% <strong>of</strong> patients without cancer.


<strong>PET</strong>-<strong>CT</strong> accuracy was 94% (95% CI: 89; 98), which means that 94% <strong>of</strong> lesions were correctly<br />

diagnosed. The accuracy <strong>of</strong> <strong>CT</strong> was 74% (95% CI: 66; 82), which means that diagnosis was<br />

accurate for 74% <strong>of</strong> lesions. The difference is significant statistically (p < 0.05; McNemar's test).<br />

The positive likelihood ratio as calculated for <strong>PET</strong>-<strong>CT</strong> is 12.88 (95% CI: 5.96; 27.83), so the<br />

probability <strong>of</strong> a positive results is 12.88 times higher for a tumor focus than the probability <strong>of</strong><br />

positive results for no lesion. The positive likelihood ratio as calculated for <strong>CT</strong> is 2.92 (95% CI:<br />

1.93; 4.43), which means that the probability <strong>of</strong> positive results is 2.92 times higher for tumor<br />

focus than the probability that exists for no lesion.<br />

The negative Likelihood ratio as calculated for <strong>PET</strong>-<strong>CT</strong> is 0.02 (95% CI: 0.003; 0.16), which<br />

means that the probability <strong>of</strong> negative results for tumor lesions represents 0.02 <strong>of</strong> the<br />

probability that exists for no tumor focus. The negative likelihood ratio as calculated for <strong>CT</strong> is<br />

0.35 (95% CI: 0.21; 0.58), so the probability <strong>of</strong> negative results for tumor lesions represents 0.35<br />

<strong>of</strong> the probability for no lesions.<br />

The diagnostic odds ratio as calculated for <strong>PET</strong>-<strong>CT</strong> was 547.5 (95% CI: 63.82; 4697.20), which<br />

means that the probability <strong>of</strong> positive results is 547.5 times higher for tumor lesions than the<br />

probability existing for the group <strong>of</strong> patients in ho no lesions were detected using the<br />

reference test. The diagnostic odds ratio is 8.36 (95% CI: 3.64; 19.18) for <strong>CT</strong>, which means that<br />

the probability <strong>of</strong> positive results in situations when lesions were detected by the reference<br />

test is 8.36 times higher than the probability for patients with no disease focus.<br />

The study provides no results per patient.<br />

13.1.4.2 Safety<br />

No information on the safety <strong>of</strong> the diagnostic procedures was found in the study.<br />

13.1.5. Results<br />

As a result <strong>of</strong> searching through medical databases one primary study was found<br />

(Branstetter 2005) that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with <strong>CT</strong> in the<br />

diagnostics <strong>of</strong> malignant head and neck tumors (primary lesions, recurrences following<br />

therapy, cervical lymph node metastasis from an unknown primary focus; N = 65). Biopsy <strong>of</strong><br />

suspicious lesions, clinical observation, and additional diagnostic imaging were used as the<br />

reference standard.<br />

The sensitivity and specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> were 98% (95% CI: 88; 100) and 92% (95% CI: 84; 97)<br />

respectively, while for <strong>CT</strong> scans the values were 74% (95% CI: 59; 86) and 75% (95% CI: 64; 84).<br />

The accuracy was rated 94% for <strong>PET</strong>-<strong>CT</strong> (95% CI: 89; 98) and 74% for <strong>CT</strong> (95% CI: 66; 82). The<br />

difference is statistically significant.<br />

193


194<br />

13.2. Detection <strong>of</strong> bone involvement by oral cancer<br />

13.2.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching through medical databases one primary study was found (Goerres<br />

2005; tab. 156, app. 18.1) that compared the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with that <strong>of</strong> <strong>CT</strong><br />

and SPE<strong>CT</strong>/<strong>CT</strong> in detecting bone infiltration by oral cancer.<br />

13.2.2. Population characteristics<br />

Goerres 2005 is a retrospective trial on a population <strong>of</strong> 34 more patients with oral cancer in<br />

who involvement <strong>of</strong> lower and upper jaw was suspected based on the clinical picture.<br />

All the patients took surgical treatment, contrast <strong>CT</strong> and SPE<strong>CT</strong>/<strong>CT</strong> scanning <strong>of</strong> the head<br />

and neck, bone scintigraphy, and <strong>PET</strong>-<strong>CT</strong> scanning was acquired from head to pelvic fundus<br />

or covering lower limbs.<br />

The time interval between the <strong>PET</strong>-<strong>CT</strong> and SPE<strong>CT</strong>/<strong>CT</strong> tests was 0-10 days (2.7 day average;<br />

SD 2.4 day; 2 days median). The time difference was 0-24 days (5 days average; SD 5.2 day; 4<br />

days median).<br />

Table 81 presents the characteristics <strong>of</strong> the population.<br />

Table 81.<br />

Population characteristics in Goerres 2005<br />

Location <strong>of</strong> primary focus [persons]<br />

Parameter Goerres 2005<br />

Size <strong>of</strong> population 34<br />

Median age (from - to) [years] 64,2 (46,0–84,6)<br />

Female [persons] 17<br />

13.2.3. Description <strong>of</strong> intervention<br />

13.2.3.1 <strong>PET</strong>-<strong>CT</strong> imaging<br />

Male [persons] 17<br />

Lower alveolar process 28<br />

Upper alveolar process 4<br />

Upper gum in the<br />

retromolar area<br />

The <strong>PET</strong>-<strong>CT</strong> scanning was executed using a Discovery LS (GE Medical Systems) unit. After<br />

four hours’ starving, the patients were given 370 MBq FDG intravenously. Imaging was done 1<br />

2


hour after FDG had been given. Moreover, a contrast medium was administered orally.<br />

Corrections <strong>of</strong> attenuation were done based on <strong>CT</strong> images. The interpretation <strong>of</strong> images was<br />

done by experienced radiology and nuclear medicine specialists.<br />

13.2.3.2 Diagnostic test compared<br />

<strong>CT</strong> imaging was done using a Somatom VolumeZoom (Siemens) unit. Scans were taken<br />

from cervical base to collarbone with the patient in the supine position. Contrast was given<br />

intravenously before the examination.<br />

Scintigraphy was taken using the SPE<strong>CT</strong>/<strong>CT</strong> technology, with the following contrast media<br />

and equipment:<br />

• 3 hours before the examination a patient was given, intravenously, the contrast medium<br />

Teceos (Schering), which contained 99m technetium <strong>of</strong> the activity <strong>of</strong> 650 MBq;<br />

• preliminary whole-body study was done with a Bodyscan set (Siemens);<br />

• final SPE<strong>CT</strong>/<strong>CT</strong> study was done with the use <strong>of</strong> a Hawkeye Millennium VG8 instrument (GE<br />

Medical Systems).<br />

<strong>CT</strong> image was used to correct attenuation.<br />

13.2.3.3 Reference test<br />

All the patients in Goerres 2005 had their tumors resected surgically and examined<br />

histopathologically. Bone surgeries were performed only if one <strong>of</strong> the imaging methods<br />

suggested bone involvement or if such suspicion appeared during resection.<br />

13.2.4. Findings<br />

13.2.4.1 Diagnostic efficacy<br />

Goerres 2005 evaluated contrast <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> efficacy in detecting bone involvement<br />

by oral cancer. Two <strong>of</strong> the patients tested false positive with <strong>PET</strong>-<strong>CT</strong>. SPE<strong>CT</strong>/<strong>CT</strong> scanning<br />

yielded three false positive results and one false negative. In one patient the result <strong>of</strong> contrast<br />

<strong>CT</strong> was false positive.<br />

Table 82.<br />

Test results for each <strong>of</strong> the methods compared vs. the reference test in Goerres 2005; calculated per patient<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong> SPE<strong>CT</strong>/<strong>CT</strong> <strong>CT</strong><br />

TP FP FN TN TP FP FN TN TP FP FN TN<br />

Goerres 2005 12 2 0 20 11 3 1 19 11 0 1 22<br />

195


196<br />

Table 83 presents the diagnostic efficacy parameters as calculated for <strong>PET</strong>-<strong>CT</strong>, SPE<strong>CT</strong>/<strong>CT</strong><br />

and <strong>CT</strong>.<br />

Table 83.<br />

Diagnostic efficacy parameters for the technologies compared in Goerres 2005, calculated per patient<br />

Parameter<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

LR+<br />

(95% CI)<br />

LR-<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

DOR<br />

(95% CI)<br />

Goerres 2005<br />

<strong>PET</strong>-<strong>CT</strong> SPE<strong>CT</strong>/<strong>CT</strong> <strong>CT</strong><br />

100%<br />

(74; 100)<br />

91%<br />

(71; 99)<br />

8,85<br />

(2,73; 28,65)<br />

0,04<br />

(0,003; 0,66)<br />

94%<br />

(80; 99)<br />

205,00<br />

(9,08; 4627,50)<br />

92%<br />

(62; 100)<br />

86%<br />

(65; 97)<br />

6,72<br />

(2,32; 19,51)<br />

0,10<br />

(0,02; 0,64)<br />

88%<br />

(73; 97)<br />

69,67<br />

(6,44; 754,16)<br />

92%<br />

(62; 100)<br />

100%<br />

(85; 100)<br />

40,69<br />

(2,61; 635,61)<br />

0,12<br />

(0,03; 0,53)<br />

97%<br />

(85; 100)<br />

345,00<br />

(13,00; 9155,10)<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity was 100% (95% CI: 74; 100), SPE<strong>CT</strong>/<strong>CT</strong> sensitivity was 92% (95% CI: 62; 100),<br />

and <strong>CT</strong> sensitivity was 92% (95% CI: 62; 100). That means that <strong>PET</strong>-<strong>CT</strong> detected correctly bone<br />

involvement in all cases, whereas <strong>CT</strong> and SPE<strong>CT</strong>/<strong>CT</strong> detected lesions in 92% patients<br />

diagnosed with bone involvement. The difference is not statistically significant.<br />

The specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 91% (95% CI: 71; 99), the specificity <strong>of</strong> SPE<strong>CT</strong>/<strong>CT</strong> was 86% (95%<br />

CI: 65; 97), and the specificity <strong>of</strong> <strong>CT</strong> was 100% (95% CI: 85; 100). It means that for all the<br />

patients with no bone involvement the <strong>CT</strong> result was negative, the results were negative for<br />

<strong>PET</strong>-<strong>CT</strong> and SPE<strong>CT</strong>/<strong>CT</strong> in 91% and 86% patients with no lesions in bones respectively. The<br />

difference is not statistically significant.<br />

The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 94% (95% CI: 80; 99), the accuracy <strong>of</strong> SPE<strong>CT</strong>/<strong>CT</strong> was 88% (95%<br />

CI: 73; 97), and the accuracy <strong>of</strong> <strong>CT</strong> was 97% (95% CI: 85; 100). That means that the<br />

proportions <strong>of</strong> patients diagnosed correctly using <strong>PET</strong>-<strong>CT</strong>, SPE<strong>CT</strong>/<strong>CT</strong> and <strong>CT</strong> technologies were<br />

94%, 88% and 97% respectively. The difference is not statistically significant.<br />

The positive likelihood ratio as calculated for <strong>PET</strong>-<strong>CT</strong> is 8,85 (95% CI: 2,73; 28,65), therefore<br />

the probability <strong>of</strong> positive results is 8.85 times higher for patients diagnosed with bone<br />

involvement than the probability for patients without involvement. The positive likelihood ratio<br />

for SPE<strong>CT</strong>/<strong>CT</strong> is 6.72 (95% CI: 2,32; 19,51). That means that the probability <strong>of</strong> positive results is


6.72 times higher for patients with bone invasion than the probability <strong>of</strong> positive results for<br />

patients without bone involvement. The positive likelihood ratio as calculated for <strong>CT</strong> is 40.69<br />

(95% CI: 2,61; 635,61). That means that the probability <strong>of</strong> positive results is 40,69 times higher<br />

for patients diagnosed with bone invasion than the probability that exists among patients with<br />

no bone invasion.<br />

The negative likelihood ratio as calculated for <strong>PET</strong>-<strong>CT</strong> is 0,04 (95% CI: 0,003; 0,66). That<br />

means that the probability <strong>of</strong> negative results for patients diagnosed with bone involvement<br />

represents 0.04 <strong>of</strong> the probability that exists for patients with no invasion <strong>of</strong> bone by oral<br />

cancer. The negative likelihood ratio as calculated for SPE<strong>CT</strong>/<strong>CT</strong> is 0.10 (95% CI: 0,02; 0,64),<br />

therefore the probability <strong>of</strong> negative result for patients diagnosed with bone involvement<br />

represents 0.10 <strong>of</strong> the probability for people with no infiltration. The negative likelihood ratio as<br />

calculated for <strong>CT</strong> is 0.12 (95% CI: 0,03; 0,53), therefore the probability <strong>of</strong> negative results for<br />

patients diagnosed with bone involvement represents 0.12 <strong>of</strong> the probability for patients with<br />

no infiltration.<br />

The diagnostic odds ratio as calculated for <strong>PET</strong>-<strong>CT</strong> was 205.00 (95% CI: 9,08; 4627,5). That<br />

means that the probability <strong>of</strong> positive results is 205 times higher for patients with bones<br />

involvement compared to the probability that exists for patients testing negative for bone<br />

involvement in reference tests. The diagnostic odds ratio <strong>of</strong> SPE<strong>CT</strong>/<strong>CT</strong> is 69.67 (95% CI: 6,44;<br />

754,16), which means that the probability <strong>of</strong> positive results is 69.67 times higher for patients<br />

diagnosed with bone involvement by the reference test than for patients with no infiltration.<br />

The diagnostic odds ratio <strong>of</strong> <strong>CT</strong> is 345.00 (95% CI: 13,00; 9155,10), which means that the<br />

probability <strong>of</strong> positive result 345 times higher for patients testing positive for bone involvement<br />

in reference tests than for patients with no infiltration.<br />

13.2.4.2 Safety<br />

No information on the safety <strong>of</strong> the diagnostic procedures used was found in the trial<br />

included in this <strong>analysis</strong>.<br />

13.2.5. Results<br />

As a result <strong>of</strong> searching through medical databases one primary trial was found<br />

(Goerres 2005) that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with SPE<strong>CT</strong>/<strong>CT</strong> and<br />

<strong>CT</strong> in identifying bone involvement by oral cavity cancer (N = 34). Histopathological<br />

examination following a surgical procedure was used as the reference standard.<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was the highest and stood at 100% (95% CI: 74; 100), the sensitivity <strong>of</strong><br />

SPE<strong>CT</strong>/<strong>CT</strong> was 92% (95% CI: 62; 100). The sensitivity <strong>of</strong> <strong>CT</strong> was 92% (95% CI: 62; 100). The<br />

specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 91% (95% CI: 71; 99), compared to 86% for SPE<strong>CT</strong>/<strong>CT</strong> (95% CI: 65; 97)<br />

and 100% for <strong>CT</strong> (95% CI: 85; 100).<br />

197


198<br />

The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning was 94% (95% CI: 80; 99), compared to 88% for SPE<strong>CT</strong>/<strong>CT</strong><br />

(95% CI: 73; 97) and 97% for <strong>CT</strong> (95% CI: 85; 100), which represents 94%, 88% and 97%, <strong>of</strong><br />

properly diagnosed patients for <strong>PET</strong>-<strong>CT</strong>, SPE<strong>CT</strong>/<strong>CT</strong> and <strong>CT</strong> respectively. The differences in<br />

accuracy, sensitivity and specificity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>, <strong>CT</strong> and SPE<strong>CT</strong>/<strong>CT</strong> tests in the diagnostics <strong>of</strong><br />

local involvement <strong>of</strong> bones by oral cavity cancer are not statistically significant<br />

13.3. Impact <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostics on treatment revisions<br />

13.3.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching medical databases, two primary clinical studies were found (Koshy<br />

2005; tab. 157, app. 18.1, Wild 2005; tab. 154, app 18.1), which compared directly the<br />

diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with traditional methods <strong>of</strong> clinical assessment and imaging in<br />

the diagnostics <strong>of</strong> head and neck cancer.<br />

13.3.2. Population characteristics<br />

The retrospective study Wild 2005 included 21 patients with cancers <strong>of</strong> the nasal cavity,<br />

paranasal sinuses, orbital cavity, pterygopalatine fossa and infratemporal fossa. Patients with<br />

their pterygopalatine fossa or infratemporal fossa tumors restaged were included in the study<br />

only if the tumor was located in the paranasal sinsuses or the nasal cavity. All neoplasm cases<br />

were confirmed histologically prior to imaging. <strong>PET</strong>-<strong>CT</strong> scanning was done as additional to <strong>CT</strong><br />

or MRI. Included were only those patients, for whom both imaging results and observation<br />

data enabling clinical assessment were available. <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> or MRI examinations could<br />

not be separated by treatment procedures. 26 examinations were executed for the purposes<br />

<strong>of</strong> staging or restaging: 9 were done for disease staging, and 15 patients took 17 restaging<br />

scans. The time interval between a traditional imaging study and <strong>PET</strong>-<strong>CT</strong> was below 2 weeks.<br />

<strong>PET</strong>-<strong>CT</strong> tests were done twice for five patients.<br />

Koshy 2005 included 36 patients with previously untreated squamous cell carcinoma <strong>of</strong> the<br />

head and neck, who had taken radiotherapy. <strong>PET</strong>-<strong>CT</strong> scanning was executed in all the<br />

patients as part <strong>of</strong> their planned radiotherapy on the same day as, or a week from, <strong>CT</strong><br />

scanning. All the patients with one exception had been recently diagnosed with head and<br />

neck cancer. Routine procedures were followed for the patients, comprising medical check-<br />

up, <strong>CT</strong> (with contrast) <strong>of</strong> the head and neck, and chest X-ray. Patients with metastasis from<br />

unknown primary foci underwent endoscopic biopsies or tonsillectomy. Patients with tumors<br />

<strong>of</strong> nasopharynx and paranasal sinuses had MRI examinations <strong>of</strong> the head and neck. Patients<br />

with lungs metastasis suspected based on an X-ray study had <strong>CT</strong> chest tests.


Table 84 presents the characteristics <strong>of</strong> the population.<br />

Table 84.<br />

Population characteristics<br />

Parameter Koshy 2005 Wild 2005<br />

Size <strong>of</strong> population 36 21<br />

Median age +/-SD (from - to) [years] 56 (32–80)<br />

Location <strong>of</strong> primary focus<br />

[persons]<br />

59,1+/-16,4<br />

(16–87)<br />

Female [persons] 8 6<br />

Male [persons] 28 15<br />

Maxillary sinus - 9<br />

Sphenoidal sinus - 3<br />

Orbital cavity or orbital<br />

cavity & ethomid sinus<br />

- 2<br />

Paranasal sinuses 3 -<br />

Nasopharynx 5 -<br />

Nasal cavity - 3<br />

Oropharynx 17 -<br />

Larynx 4 -<br />

Oral cavity 2 -<br />

Lower part <strong>of</strong> pharynx 2 -<br />

Unknown primary focus 3 -<br />

pterygopalatine fossa and<br />

infratemporal fossa<br />

(restaging)<br />

13.3.3. Intervention description<br />

13.3.3.1 <strong>PET</strong>-<strong>CT</strong> imaging<br />

- 4<br />

In Wild 2005, <strong>PET</strong>-<strong>CT</strong> imaging was done using a Discovery LS scanner (GE Medical Systems).<br />

After 4 hours’ starving, the patients were given 370 MBq <strong>of</strong> FDG intravenously. Imaging was<br />

done 1 hour after the contrast medium was administered. Images were taken from head to<br />

pelvic fundus. <strong>CT</strong> images were used to correct attenuation.<br />

199


200<br />

<strong>PET</strong>-<strong>CT</strong> scanning in Koshy 2005 was executed using a Discovery LS unit (GE Medical<br />

Systems) after 4 hours’ starving, 45 to 60 minutes after 370–440 MBq <strong>of</strong> FDG was given<br />

intravenously to patients immobilized by means <strong>of</strong> a mask. Corrections <strong>of</strong> attenuation were<br />

done using <strong>CT</strong> images. <strong>PET</strong>-<strong>CT</strong> images were interpreted with the use <strong>of</strong> available clinical<br />

information, both by radiology and nuclear medicine specialists. Imaging covered the whole<br />

body.<br />

13.3.3.2 Diagnostic technology compared<br />

In Wild 2005, <strong>CT</strong> scanning was executed using a Somatom VolumeZoom scanner (Siemens)<br />

with intravenous contrast, from cervical base to collar bones.<br />

All MRI examinations were executed using a 1.5 T GE Signa instrument after contrast was<br />

given intravenously.<br />

In Koshy 2005, <strong>CT</strong> scanning was done using a GE light speed scanner (General Electric)<br />

after contrast was given intravenously to patients immobilized by means <strong>of</strong> a mask. The<br />

examination covered the head and neck; only patients with lungs metastasis had chest X-ray<br />

and <strong>CT</strong> examinations.<br />

Also, patients took medical check-ups, and, in case <strong>of</strong> nasopharynx or paranasal sinus<br />

tumors, they had MRI examinations <strong>of</strong> the head and neck (no data on equipment).<br />

13.3.3.3 Reference test<br />

In Wild 2005, all patients with lesions had histological test.<br />

In Koshy 2005, when distant metastasis or synchronous tumors were suspected, the<br />

reference test comprised histological examination and a combination <strong>of</strong> additional<br />

diagnostic imaging and clinical observation. No diagnostic imaging or clinical observation<br />

data are available.<br />

13.3.4. Results<br />

13.3.4.1 Impact on therapy<br />

Table 85 provides a detailed list <strong>of</strong> revisions to treatment <strong>of</strong> head and neck cancer<br />

triggered by <strong>PET</strong>-<strong>CT</strong> scanning.


Table 85.<br />

Impact <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> on revisions to head and neck cancer therapy<br />

Changes in treatment<br />

after <strong>PET</strong>-<strong>CT</strong><br />

Chemotherapy in lieu <strong>of</strong><br />

radiotherapy and<br />

surgery<br />

Additional therapeutic<br />

surgery<br />

Palliative radiotherapy<br />

with additional<br />

chemotherapy in lieu <strong>of</strong><br />

therapeutic<br />

radiotherapy<br />

Koshy 2005 Wild 2005<br />

- 1<br />

- 1<br />

- 1<br />

Additional surgery 1 1<br />

Chemotherapy in lieu <strong>of</strong><br />

surgery<br />

Additional<br />

chemotherapy<br />

Discontinuation <strong>of</strong><br />

chemotherapy<br />

Changes <strong>of</strong><br />

chemotherapy schemes<br />

Adjustment <strong>of</strong> radiation<br />

area<br />

Adjustment <strong>of</strong> radiation<br />

doses<br />

- 2<br />

- 1<br />

1 -<br />

2 -<br />

5 2<br />

4 -<br />

In Wild 2005, the area <strong>of</strong> radiation was adjusted for two patients based on <strong>PET</strong>-<strong>CT</strong> findings.<br />

Two patients took chemotherapy instead <strong>of</strong> surgery; one patient took chemotherapy instead<br />

<strong>of</strong> radiotherapy and surgery planned before; lesions detected by <strong>PET</strong>-<strong>CT</strong> were removed<br />

surgically in two patients; one person took chemotherapy instead <strong>of</strong> no treatment; one<br />

person was given palliative chemoradiotherapy instead <strong>of</strong> therapeutic radiotherapy.<br />

In Koshy 2005, some modifications were made to treatment procedures: the radiation area<br />

was adjusted for five patients; the radiation dose was adjusted for four patients; one patient<br />

did not take chemotherapy although it had been planned before; one patient underwent<br />

surgery and radiochemotherapy. Among the patients for who treatment plans were revised,<br />

the disease was upstaged on the TNM scale for five patients, for one the disease was<br />

downstaged and for three it did not change.<br />

Table 86 lists revisions in therapy for patients with head and neck cancer.<br />

201


Table 86.<br />

Revisions in treatment <strong>of</strong> head an neck cancer based on <strong>PET</strong>-<strong>CT</strong> findings<br />

202<br />

Study (N) Changes <strong>of</strong> treatment Confirmation <strong>of</strong> treatment<br />

Wild 2005 (21) 9 12<br />

Koshy 2005 (36) 9 27<br />

Changes in the strategy <strong>of</strong> treatment were made based on <strong>PET</strong>-<strong>CT</strong> findings for 43% <strong>of</strong><br />

patients in Wild 2005 and for 25% in Koshy 2005.<br />

Graph 27 presents the results <strong>of</strong> a meta-<strong>analysis</strong> <strong>of</strong> Wild 2005 and Koshy 2005 details<br />

relating to therapy revisions.<br />

Graph 27.<br />

Meta-<strong>analysis</strong> (fixed effects model) <strong>of</strong> the proportion <strong>of</strong> patients for head and neck cancer therapy was revised<br />

following <strong>PET</strong>-<strong>CT</strong> examination<br />

Wild 2005 0,43 (0,22, 0,66)<br />

Koshy 2005 0,25 (0,12, 0,42)<br />

Result <strong>of</strong> meta<strong>analysis</strong><br />

The use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> examination in addition to standard diagnostic methods helped revise<br />

treatment for 32% patients with head and neck cancer (95% CI: 21; 44). Thanks to treatment<br />

revisions, some extra health effects were achieved and unnecessarily burdensome<br />

procedures were avoided.<br />

13.3.4.2 Safety<br />

0,0 0,2 0,4 0,6 0,8<br />

Proportion <strong>of</strong> patients with treatment revised (95% confidence interval)<br />

Neither <strong>of</strong> the trials reports on the safety <strong>of</strong> the diagnostic methods used.<br />

0,32 (0,21, 0,44)


13.3.5. Results<br />

As a result <strong>of</strong> searching through medical databases two primary studies were found (Wild<br />

2005, Koshy 2005) which compared directly the impact <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning on therapeutic<br />

decisions with standard head and neck cancer staging methods (<strong>CT</strong>, MRI, medical<br />

examination; N = 57).<br />

Histopathological examination was used as the reference standard in Wild 2005, while in<br />

Koshy 2005, when distant metastasis or synchronous tumors were suspected, histopathological<br />

tests or a combination <strong>of</strong> additional diagnostic imaging and clinical observation were<br />

executed (no data are available on the reference standard in the remaining patients).<br />

In Wild 2005, <strong>PET</strong>-<strong>CT</strong> findings triggered revisions <strong>of</strong> treatment suggested based on<br />

conventional imaging in 43% patients, while in Koshy 2005 the value was 25%, which<br />

produced the total <strong>of</strong> 32% cases (95% CI: 21; 44).<br />

203


14. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS.<br />

CONVENTIONAL IMAGING METHODS IN PANCREATIC<br />

CANCER DIAGNOSTICS<br />

204<br />

14.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching through medical databases, one primary prospective clinical trial<br />

was found (Heinrich 2005; tab. 158, app. 18.1). It compares the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

with conventional imaging (<strong>CT</strong> with contrast medium, ERCP, MRI, EUS, diagnostic<br />

laparoscopy) in pancreatic cancer diagnostics.<br />

14.2. Population characteristics<br />

59 patients with suspicions <strong>of</strong> pancreatic cancer were included in the trial. The diagnostic<br />

efficacy <strong>of</strong> the imaging methods compared was evaluated with respect to both detecting<br />

the primary focus <strong>of</strong> pancreatic cancer and T-staging.<br />

Initial characteristics <strong>of</strong> the population is presented in table 87.<br />

Table 87.<br />

Initial characteristics <strong>of</strong> patients in Heinrich 2005.<br />

PARAMETER POPULATION<br />

Size <strong>of</strong> population 59<br />

Median age in years (from-to)<br />

61<br />

(40–80)<br />

Male proportion (%) 50.8%<br />

Patients with primary lesion identified in the head<br />

<strong>of</strong> the pancreas<br />

Patients with primary lesion identified in the tail <strong>of</strong><br />

the pancreas<br />

Patients with test results verified through<br />

histopathological tests<br />

86.4%<br />

13.6%<br />

88.1%<br />

Patients diagnosed with malignant lesions 81.3%<br />

Patients diagnosed with benign lesions 10.2%


The median age <strong>of</strong> the cohort was 61 (from 40 to 80). 30 men (50.8%) and 29 women<br />

(49.2%) participated in the trial. Cancer involved the head <strong>of</strong> the pancreas in 51 patients<br />

(86.4%) and the tail <strong>of</strong> the pancreas in 8 (13.6%) patients.<br />

14.3. Intervention description<br />

14.3.1. <strong>PET</strong>-<strong>CT</strong> imaging<br />

The <strong>PET</strong>-<strong>CT</strong> examination was conducted by means <strong>of</strong> a <strong>PET</strong>-<strong>CT</strong> Discovery LS scanner<br />

(General Electric Medical Systems, Waukesha, WI), which consisted <strong>of</strong> a Light Speed Plus<br />

spiral <strong>CT</strong> scanner and an Advanced NXi <strong>PET</strong> scanner. The patients were instructed not to eat<br />

4 to 6 hours before the examination. The patients had been given contrast medium orally<br />

before scanning; no iodized contrast medium was administered intravenously. 18F-<br />

fluorodeoxyglucose with the activity <strong>of</strong> 350 to 450 MBq was used as a marker. First, a <strong>CT</strong> study<br />

was carried out by taking scans at an exhalation phase, from the tip <strong>of</strong> the head down to the<br />

pelvic fundus. <strong>PET</strong> scans were taken immediately after <strong>CT</strong> scans according to the same<br />

pattern as for <strong>CT</strong>. The acquisition time for a single scan was 4 minutes.<br />

Table 88 presents a characteristics <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong> scanner used in the examination, and the<br />

procedures applied.<br />

Table 88.<br />

Characteristics <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanner and the procedures used in testing.<br />

Type <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

Type <strong>of</strong> <strong>PET</strong><br />

radiomarker<br />

Discovery LS F 18 -deoxyglucose<br />

Type <strong>of</strong> <strong>CT</strong><br />

contrast medium<br />

oral contrast<br />

medium; no<br />

intravenous<br />

contrast medium<br />

Scope <strong>of</strong><br />

examination<br />

from tip <strong>of</strong> the<br />

head to pelvic<br />

fundus<br />

Radiomarker<br />

activity [MBq]<br />

350–450 MBq<br />

<strong>CT</strong> findings were analyzed simultaneously by two or more experienced nuclear medicine<br />

doctors and radiologists. The authors <strong>of</strong> the trials provide no data on how those were blinded<br />

to the findings <strong>of</strong> conventional imaging methods.<br />

Interpretation <strong>of</strong> findings involved locating areas with increased marker uptake as well as<br />

determining the outline <strong>of</strong> those areas based on fusion <strong>PET</strong>-<strong>CT</strong> images. Also, <strong>CT</strong> images were<br />

analyzed separately in order to identify additional lesions with no marker uptake.<br />

205


206<br />

14.3.2. Diagnostic technology compared<br />

<strong>PET</strong>-<strong>CT</strong> findings were compared with the findings <strong>of</strong> conventional methods.<br />

The following tests were done: <strong>CT</strong> with contrast medium (1-3 mm section); chest<br />

radiogram; endoscopic ultrasound scanning with fine-needle aspiration biopsy (BAC) <strong>of</strong><br />

primary tumor and metastatic focuses; endoscopic retrograde cholangiopancreatography<br />

(ERCP), serial testing <strong>of</strong> CA 19-9 in blood serum; and diagnostic laparoscopy.<br />

Staging was carried out in stages; at the beginning, each patient had a <strong>CT</strong> test with<br />

contrast medium as well as <strong>PET</strong>-<strong>CT</strong> scanning. If based on the findings the patient was<br />

qualified for surgery, further tests were carried out for pancreatic cancer staging (endoscopic<br />

ultrasonography, CA 19-9 in blood and diagnostic laparoscopy).<br />

The median time between contrast <strong>CT</strong> and <strong>PET</strong>-<strong>CT</strong> was 10 days.<br />

14.3.3. Reference test<br />

Ultrasound findings were verified through observations during surgery and<br />

histopathological test <strong>of</strong> material removed and bioptates.<br />

Negative test result in patients who had not undergone surgery was validated based on<br />

long-term clinical follow-up. The authors <strong>of</strong> the research reported that the median duration <strong>of</strong><br />

observation was 15 months (scope: 6-18 months) for 7 patients with pancreas lesions not<br />

validated histopathologically. During that period, endoscopic ultrasonography with fine<br />

needle aspiration biopsy, as well as the <strong>CT</strong> or MRI examinations were repeated to confirm the<br />

benign character <strong>of</strong> lesions.<br />

14.4. Findings<br />

14.4.1. Tumor detection and <strong>analysis</strong><br />

The findings <strong>of</strong> diagnostic imaging were verified histopathologically for 52 out <strong>of</strong> 59<br />

patients who had had <strong>PET</strong>-<strong>CT</strong> tests.<br />

The number <strong>of</strong> patients who tested true positive, false positive, false negative and true<br />

negative in <strong>PET</strong>-<strong>CT</strong> and contrast <strong>CT</strong> examinations carried out in a population <strong>of</strong> patients with<br />

suspected pancreatic cancer is presented in table 89.


Table 89.<br />

Number <strong>of</strong> patients with TP, FP, FN and TN results in Heinrich 2005: <strong>PET</strong>-<strong>CT</strong> vs. contrast <strong>CT</strong>.<br />

Examination<br />

<strong>PET</strong>-<strong>CT</strong> Contrast <strong>CT</strong><br />

TP FP FN TN TP FP FN TN<br />

Heinrich 2005 41 4 5 9 bd bd bd bd<br />

Based on the data above, the diagnostic efficacy parameters <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> were calculated<br />

and, along with the parameters reported by the authors for contrast <strong>CT</strong>, are presented in<br />

table 90.<br />

Table 90.<br />

Diagnostic efficacy parameters <strong>of</strong> the imaging methods compared: <strong>PET</strong>-<strong>CT</strong> vs. contrast <strong>CT</strong><br />

Parameter<br />

(95% CI)<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

Diagnostic method<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong> with contrast medium<br />

89%<br />

(76; 96)<br />

69%<br />

(39; 91)<br />

85%<br />

(73; 93)<br />

Statistical<br />

significance <strong>of</strong><br />

difference between<br />

groups compared<br />

93% p = 0,69<br />

21% p = 0,07<br />

bd not found<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity in detecting primary tumor is 89% (95% CI: 76; 96) and is lower than the<br />

sensitivity <strong>of</strong> <strong>CT</strong> with contrast medium, which is 93%. The difference is not statistically significant<br />

(p = 0.69). That means that the probability <strong>of</strong> positive <strong>PET</strong>-<strong>CT</strong> results for a patient diagnosed<br />

with pancreatic cancer is 4 pp lower compared to <strong>CT</strong> with contrast medium.<br />

<strong>PET</strong>-<strong>CT</strong> specificity in detecting primary tumor is 69% (95% CI: 39; 91) and is higher than the<br />

specificity <strong>of</strong> <strong>CT</strong> with contrast medium, which stands at 21%. That means that the probability<br />

<strong>of</strong> negative results in a patient without pancreatic cancer is 69% for the <strong>PET</strong>-<strong>CT</strong>, and is three<br />

times higher compared to <strong>CT</strong> with contrast medium (21%). The difference is not statistically<br />

significant (p = 0.07).<br />

<strong>PET</strong>-<strong>CT</strong> accuracy in detecting primary tumor is 85% (95% CI: 73; 93). The difference<br />

between groups is not statistically significant either.<br />

14.4.2. Staging<br />

The authors <strong>of</strong> Heinrich 2005 evaluated the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> imaging with<br />

respect to pancreatic cancer staging.<br />

207


208<br />

Out <strong>of</strong> 25 patients diagnosed with pancreas adenocarcinoma, who had had a surgery, 24<br />

had metastasis to lymph nodes. The number <strong>of</strong> true positive results for <strong>PET</strong>-<strong>CT</strong> was 3. The<br />

calculated sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting metastasis to lymph nodes is 21.4% (95% CI: 4,7;<br />

50,8).<br />

Out <strong>of</strong> 46 patients with pancreatic cancer confirmed histopathologically, 16 had distant<br />

metastasis confirmed histopathologically.<br />

<strong>PET</strong>-<strong>CT</strong> identified distant metastasis in 13 patients; for 5 <strong>of</strong> them <strong>PET</strong>-<strong>CT</strong> was the only<br />

method that detected distant metastasis located in the liver and retroperitoneal lymph<br />

nodes, abdominal wall, cervical lymph nodes and lungs. In 12 patients, distant metastases<br />

demonstrated an increased marker uptake; in one patient FDG-negative lesions were found<br />

in lungs; and in one patient both FDG-positive and FDG-negative lesions were found in lungs.<br />

The <strong>PET</strong>-<strong>CT</strong> technology failed to detect lesions in three patients. No false positive results were<br />

reported.<br />

Conventional methods detected lesions in nine patients; in two, metastasis to liver was<br />

diagnosed incorrectly (one patient with chronic pancreas inflammation and the other with<br />

pancreas adenoma). In 2 patients, lesions were detected only by diagnostic laparoscopy,<br />

while in 5 only by <strong>PET</strong>-<strong>CT</strong>.<br />

Table 91 presents the numbers <strong>of</strong> patients with a true positive, false positive, false negative<br />

and true negative results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning and contrast <strong>CT</strong> scanning in the population<br />

diagnosed with distant metastasis <strong>of</strong> pancreatic cancer.<br />

Table 91.<br />

Number <strong>of</strong> patients with TP, FP, FN and TN results in M-staging, <strong>PET</strong>-<strong>CT</strong> vs. conventional methods<br />

Study<br />

<strong>PET</strong>-<strong>CT</strong> Conventional methods<br />

TP FP FN TN TP FP FN TN<br />

Heinrich 2005 13 0 3 43 9 2 7 41<br />

Table 92 presents EBM parameters calculated for the diagnostic methods compared, in a<br />

group <strong>of</strong> patients diagnosed with distant metastases <strong>of</strong> pancreatic cancer.


Table 92.<br />

EBM parameters <strong>of</strong> diagnostic efficacy for the diagnostic methods compared; <strong>PET</strong>-<strong>CT</strong> vs. conventional methods<br />

Parameter<br />

Se<br />

(95% CI)<br />

Sp<br />

(95% CI)<br />

LR+<br />

(95% CI)<br />

LR-<br />

(95% CI)<br />

Acc<br />

(95% CI)<br />

DOR<br />

(95% CI)<br />

Diagnostic method<br />

<strong>PET</strong>-<strong>CT</strong> Conventional methods<br />

81%<br />

(54; 96)<br />

100%<br />

(92; 100)<br />

69.9<br />

(4.4; 1111.5)<br />

0.2<br />

(0.1; 0.5)<br />

95%<br />

(86; 99)<br />

335.6<br />

(16.3; 6913.9)<br />

56%<br />

(30; 80)<br />

95%<br />

(84; 99)<br />

12,1<br />

(2.9; 50.1)<br />

0,46<br />

(0.3; 0.8)<br />

85%<br />

(73; 93)<br />

26.4<br />

(4.7; 148.5)<br />

Statistical<br />

significance <strong>of</strong><br />

differences<br />

between groups<br />

compared<br />

p = 0.22<br />

p = 0.48<br />

-<br />

-<br />

p = 0.08<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity in detecting distant metastases is 81% (95% CI: 54; 96) and is higher than<br />

the sensitivity <strong>of</strong> the conventional imaging methods, which is 56% (95% CI: 30; 80). The<br />

probability <strong>of</strong> positive results in patients diagnosed with distant metastases is 25 pp higher for<br />

<strong>PET</strong>-<strong>CT</strong> than for the conventional methods. The difference in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is not statistically<br />

significant (p = 0.22).<br />

Specificity also favors <strong>PET</strong>-<strong>CT</strong>, for which it stands at 100% (95% CI: 92; 100) and is higher<br />

than the specificity <strong>of</strong> the conventional imaging methods, which is 95% (95% CI: 84; 99). That<br />

means that the probability <strong>of</strong> negative results in a patient without distant metastases is 100%,<br />

for <strong>PET</strong>-<strong>CT</strong>, which is 5 pp higher than the probability <strong>of</strong> negative results for the conventional<br />

methods. The differences are not statistically significant (p = 0.48).<br />

The positive likelihood ratio <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 69.9 (95% CI: 4.4; 1111.5), and is higher compared to<br />

the conventional methods, for which the value is 12.1 (95% CI: 2.9; 50.1). The probability <strong>of</strong><br />

positive <strong>PET</strong>-<strong>CT</strong> results in a patient with distant metastasis is almost 70 times higher than the<br />

probability <strong>of</strong> positive result in a patient without distant metastases.<br />

For the conventional methods, the probability <strong>of</strong> positive result is more than 12 times higher<br />

in patients with distant metastasis than in patients without metastasis.<br />

The negative likelihood ratio <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 0.2 (95% CI: 0.1; 0.5) and is lower compared to the<br />

ratio <strong>of</strong> the conventional methods, which is 0.46 (95% CI: 0.3; 0.8). For <strong>PET</strong>-<strong>CT</strong>, the probability<br />

<strong>of</strong> negative results in a patient with distant metastasis represents 20% <strong>of</strong> the probability <strong>of</strong><br />

negative results in a patient without distant metastasis.<br />

-<br />

209


210<br />

For the conventional methods, the probability <strong>of</strong> negative result in a patient with distant<br />

metastasis represents 46% <strong>of</strong> the probability in a patient without distant metastasis.<br />

The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting distant metastasis is 95% (95% CI: 86; 99) and is higher<br />

than the accuracy <strong>of</strong> the conventional methods (<strong>CT</strong>, MRI and USG), which is 85% (95% CI: 73;<br />

93). However, the difference did not reach the level <strong>of</strong> statistical significance (p = 0.08).<br />

The diagnostic odds ratio in detecting distant metastasis <strong>of</strong> pancreatic cancer is 335.6<br />

(95% CI: 16.3; 6913.9) with <strong>PET</strong>-<strong>CT</strong>, and is higher than the diagnostic odds ratio with the<br />

conventional imaging methods, for which the value is 26.4 (95% CI: 4.7; 148.5).<br />

The authors <strong>of</strong> the study report that if <strong>PET</strong>-<strong>CT</strong> scanning is used after the conventional<br />

methods to detect distant metastasis if pancreatic cancer, diagnostic sensitivity goes up to<br />

88%. The difference is not statistically significant (p = 0.06).<br />

14.4.3. Revisions <strong>of</strong> oncological procedures<br />

The authors <strong>of</strong> the study reported that two patients who participated in the trial had a<br />

synchronous rectosigmoid junction cancer diagnosed by <strong>PET</strong>-<strong>CT</strong>. Radical palliative<br />

rectosigmoid resection was performed in a patient diagnosed negatively for distant<br />

metastases <strong>of</strong> pancreatic cancer. Palliative resection <strong>of</strong> a synchronous rectal tumour was<br />

performed in a patient with pancreatic cancer metastasis in the abdominal wall. None <strong>of</strong> the<br />

patients above developed clinical symptoms <strong>of</strong> rectal cancer; for none <strong>of</strong> them lesions<br />

associated with tumor were identified by physical examination.<br />

Considering these patients, as well as five other patients for who <strong>PET</strong>-<strong>CT</strong> was the only<br />

technology to have detected distant metastasis, oncological procedures were revised for six<br />

out <strong>of</strong> 37 patients in whom pancreas lesions were recognized as resectable.<br />

Table 93 presents the impact <strong>of</strong> both methods compared on the oncological procedures<br />

for patients with pancreatic cancer (staging).<br />

Table 93.<br />

Probability <strong>of</strong> revision <strong>of</strong> oncological procedure in patients diagnosed using the methods compared: <strong>PET</strong>-<strong>CT</strong> vs.<br />

conventional methods<br />

Parameter<br />

Probability <strong>of</strong> revision<br />

(95% CI)<br />

Diagnostic method<br />

<strong>PET</strong>-<strong>CT</strong> Conventional methods<br />

33%<br />

(20; 48)<br />

20%<br />

(9; 34)<br />

Statistical significance<br />

<strong>of</strong> difference between<br />

groups compared<br />

p = 0.03


The odds <strong>of</strong> revision <strong>of</strong> oncological procedure for pancreatic cancer patients when <strong>PET</strong>-<strong>CT</strong><br />

scans are used for staging is 33% (95% CI: 20; 48) and is 13 pp higher compared to the<br />

conventional methods. The difference in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is statistically significant (p = 0.03).<br />

Moreover, <strong>PET</strong>-<strong>CT</strong> is more efficacious in determining the type <strong>of</strong> lesion as it provides a<br />

detailed anatomical imaging <strong>of</strong> its outline.<br />

14.4.4. Safety<br />

The authors <strong>of</strong> the study do not report on possible side effects in patients diagnosed by<br />

<strong>PET</strong>-<strong>CT</strong> and conventional methods.<br />

14.5. Results<br />

As a result <strong>of</strong> searching through medical databases one primary clinical trial was found<br />

(Heinrich 2005) that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with conventional<br />

imaging methods (contrast <strong>CT</strong>, ERCP, MRI, EUS, diagnostic laparoscopy) in diagnosing primary<br />

lesions and in staging pancreatic cancer (N = 59). The comparison <strong>of</strong> imaging methods was<br />

verified by histopathological tests <strong>of</strong> material removed during a surgical procedure or<br />

bioptates, or by long-term clinical observation.<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning in detecting primary lesions <strong>of</strong> pancreatic cancer is 89%<br />

(95% CI: 76; 96), which is lower than the sensitivity <strong>of</strong> contrast <strong>CT</strong>, which is 93%. <strong>PET</strong>-<strong>CT</strong> imaging<br />

has a higher sensitivity rating than contrast <strong>CT</strong> in detecting pancreatic cancer, the estimated<br />

values being 69% (95% CI: 39; 91) vs. 21%, respectively. The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 85% (95% CI:<br />

73; 93). No statistically significant differences were observed between the groups for any <strong>of</strong><br />

the parameters <strong>of</strong> efficacy discussed.<br />

In M-staging, <strong>PET</strong>-<strong>CT</strong> is characterized by a higher sensitivity than conventional imaging<br />

methods, the estimated values being 81% (95% CI: 54; 96) vs. 56% (95% CI: 30; 80) respectively.<br />

The difference in favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is not statistically significant (p = 0.22). Imaging specificity<br />

favors <strong>PET</strong>-<strong>CT</strong>, for which it stands at 100% (95% CI: 92; 100) vs. 95% (95% CI: 84; 99) for the<br />

conventional methods compared. The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in M-staging is 95% (95% CI: 86; 99)<br />

and is higher than the accuracy <strong>of</strong> the conventional methods (<strong>CT</strong>, NMR and USG), 85% (95%<br />

CI: 73; 93). The difference, although close to the threshold, did not reach statistical<br />

significance (p = 0.08).<br />

A revision <strong>of</strong> oncological procedures as a result <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning was reported for 16%<br />

patients (95% CI: 6; 32) with resectable pancreatic cancer. Procedure revision is more likely if<br />

<strong>PET</strong>-<strong>CT</strong> scanning is used in clinical staging than if the conventional methods are used, the<br />

211


estimated values being 33% (95% CI: 20; 48) vs. 20% (9; 34) respectively. The difference in<br />

favor <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is statistically significant (p = 0.03).<br />

212


15. COMPARATIVE EFFICACY ANALYSIS OF <strong>PET</strong>-<strong>CT</strong> VS. <strong>CT</strong> IN<br />

THE STAGING AND EVALUATING RESPONSE TO<br />

TREATMENT OF GASTROINTESTINAL STROMAL TUMOR<br />

15.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching medical databases, one primary clinical study was found (Antoch<br />

2004; tab. 159, app. 18.1) that evaluated the diagnostic efficacy <strong>of</strong> <strong>PET</strong> coupled with<br />

roentgen <strong>CT</strong> in detecting gastrointestinal stromal tumour (GIST).<br />

15.2. Population characteristics<br />

20 patients with gastrointestinal stromal tumor confirmed clinically were included in the<br />

study provided GIST diagnosis was confirmed for them through histopathological examination<br />

<strong>of</strong> primary tumor or metastasis.<br />

The median age <strong>of</strong> the patients was 60 (from 39 to 77). Men represented 55% <strong>of</strong> the<br />

population. The proportion <strong>of</strong> patients with a primary focus located in the stomach was 60%,<br />

in small intestine – 35%, and in mesentery – 5%.<br />

Results <strong>of</strong> initial neoplasm staging, and subsequently results <strong>of</strong> <strong>analysis</strong> <strong>of</strong> response to<br />

imatinib treatment over 6 months were an indication for diagnosis.<br />

Scans were taken prior to therapy and again in the first, third and sixth month <strong>of</strong> it.<br />

The imaging procedures were not available for all the patients at the same time. The total<br />

number <strong>of</strong> tests was 20 initially, 20 in the first month, 10 in the third month, 9 in the sixth month,<br />

and 20 in the follow-up period, which, along with clinical data, served as a reference test. The<br />

<strong>analysis</strong> was based on 79 tests altogether.<br />

15.3. Description <strong>of</strong> intervention<br />

15.3.1. <strong>PET</strong>-<strong>CT</strong> imaging<br />

Diagnostic <strong>PET</strong>-<strong>CT</strong> imaging was performed by means <strong>of</strong> a Biograph scanner (Siemens<br />

Medical Solutions) that consisted <strong>of</strong> a <strong>CT</strong> scanner (Somatom Emotion; Siemens) and a <strong>PET</strong><br />

scanner (ECAT HR+; Siemens).<br />

213


214<br />

The Biograph produced two separate <strong>CT</strong> and <strong>PET</strong> images that were assessed individually,<br />

and again after fusion, to evaluate therapeutic efficacy. 18F-fluorodeoxyglucose (FDG) was<br />

used as a marker.<br />

<strong>PET</strong> imaging was carried out 60 minutes after the average dose <strong>of</strong> 358 MBq 18F-FDG (±42<br />

MBq) was administered. The patients had been asked not to eat for 4 hours minimum before<br />

the examination. The glucose level in blood was confirmed and blood samples taken before<br />

the marker was administered. <strong>PET</strong> scanning duration was adjusted to the patients’ weight. For<br />

patients with body weight below 65 kg, <strong>emission</strong> time was 3 minutes, for 65–85 kg it was 4<br />

minutes, and for patients with body weight above 85 kg it was 5 minutes. For the purposes <strong>of</strong><br />

GIST diagnosis all the patients had <strong>PET</strong>-<strong>CT</strong> scans <strong>of</strong> the whole body taken. Imaging (which<br />

included staging) was limited to areas where a tumor had been detected, yet in order to<br />

detect new distant metastasis whole-body scans were taken every 3 months.<br />

Table 94.<br />

<strong>PET</strong>-<strong>CT</strong> details<br />

Type <strong>of</strong><br />

<strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

Biograph<br />

Manufacturer <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

Siemens Medical<br />

Solutions (H<strong>of</strong>fman<br />

Estates, USA)<br />

Type <strong>of</strong><br />

radiopharmaceutical<br />

Coverage<br />

Radiomarker<br />

activity [MBq]<br />

FDG whole body 358<br />

<strong>PET</strong> scans were interpreted by two nuclear medicine doctors, and <strong>CT</strong> data by two<br />

radiologists. They were blinded to the findings <strong>of</strong> other imaging tests. <strong>PET</strong>-<strong>CT</strong> fusion results were<br />

interpreted through consensus examined by the same doctors.<br />

15.3.2. Diagnostic technology compared<br />

<strong>PET</strong>-<strong>CT</strong> findings were compared with <strong>CT</strong> findings (i.e. <strong>CT</strong> as a component <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>). <strong>CT</strong><br />

was carried out using a Somatom Emotion <strong>CT</strong> scanner (Siemens) which is a part <strong>of</strong> a Biograph<br />

<strong>PET</strong>-<strong>CT</strong> scanner (Siemens Medical Solutions).<br />

Table 95.<br />

<strong>CT</strong> details<br />

Type <strong>of</strong> <strong>CT</strong> scanner <strong>CT</strong> scanner manufacturer<br />

Contrast type and method <strong>of</strong><br />

administration<br />

<strong>CT</strong> Somatom Emotion Siemens Medical Solutions intravenously and orally


The <strong>CT</strong> scans carried out to assess the efficacy <strong>of</strong> therapy was classified in keeping with<br />

the directives <strong>of</strong> the World Health Organization (WHO) and The Response Evaluation Criteria<br />

in Solid Tumors (RECIST).<br />

15.3.3. Reference test<br />

Clinical observation following the completion <strong>of</strong> a six-month-long treatment and<br />

observation <strong>of</strong> response to treatment were used as the reference method. The average time<br />

<strong>of</strong> observation was 381 days (SD 134). All the clinical data available were collected during<br />

that time, including physical examinations, laboratory examinations, and radiological<br />

imaging procedures (<strong>CT</strong>, MRI, <strong>PET</strong>-<strong>CT</strong>). All the patients had whole-body <strong>PET</strong>-<strong>CT</strong> scanning.<br />

The participants were divided into two groups: one consisted <strong>of</strong> patients who responded<br />

to treatment, the other included patients who did not. The evaluation was done by a team <strong>of</strong><br />

a nuclear medicine doctor, a radiologist and an oncologist. They had access to all clinical<br />

and radiological data, and had not taken part in any previous imaging diagnosis.<br />

The patients were classified as responsive or non-responsive to treatment, in keeping with<br />

the criteria <strong>of</strong> WHO, RECIST as well as EORTC (European Organization for Research and<br />

Treatment <strong>of</strong> Cancer).<br />

15.4. Findings<br />

15.4.1. Staging<br />

In initial staging, 282 neoplastic lesions were located in 20 patients using <strong>PET</strong>-<strong>CT</strong>, but 249<br />

using <strong>CT</strong> with contrast medium. The differences between the diagnostic methods were<br />

statistically significant (p < 0.0001).<br />

Initially, <strong>CT</strong> did not detect any lesions in 2 patients (10%), therefore the evaluation <strong>of</strong><br />

response to treatment was limited to 18 people for this method.<br />

15.4.2. Response to treatment<br />

The authors <strong>of</strong> the study analyzed the similarity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> findings in evaluating<br />

response to treatment vs. the reference method. After 1, 3 and 6 months, <strong>PET</strong>-<strong>CT</strong> findings<br />

corresponded to the results <strong>of</strong> the reference test in terms <strong>of</strong> statistical significance. In none <strong>of</strong><br />

those periods significant agreement was observed for <strong>CT</strong> findings. Details are presented in<br />

Table 96.<br />

215


Table 96.<br />

Scanning findings vs. reference test<br />

216<br />

Imaging method<br />

<strong>CT</strong> (WHO)<br />

<strong>CT</strong> (RECIST)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Scan timing after therapy<br />

(months)<br />

p-value<br />

1 NF<br />

3 NF<br />

6 NF<br />

1 NF<br />

3 NF<br />

6 NF<br />

1 < 0.001<br />

3 0.001<br />

6 0.005<br />

The study also evaluated whether response to treatment was correct in the first, third and<br />

sixth month. Details are presented in Table 97.<br />

Table 97.<br />

Correct response to treatment<br />

Period <strong>PET</strong>-<strong>CT</strong> (N = 20) <strong>CT</strong> (N = 18)<br />

Statistical significance<br />

<strong>of</strong> tests differences<br />

1 st month 95% 44% p = 0.001<br />

3 rd month 100% 60% NF<br />

6 th month 100% 57% NF<br />

<strong>PET</strong>-<strong>CT</strong> helped determine correctly response to treatment in the majority <strong>of</strong> patients<br />

compared with <strong>CT</strong> results in each period <strong>of</strong> observation. The observed differences were<br />

statistically significant in the first month.<br />

15.4.3. Safety<br />

The authors do not report on the side effects <strong>of</strong> the diagnostic techniques used.<br />

15.5. Results<br />

A single clinical trial was found (Antoch 2004) that compared the diagnostic efficacy <strong>of</strong><br />

<strong>PET</strong>-<strong>CT</strong> with that <strong>of</strong> <strong>CT</strong> imaging in GIST staging and in the evaluation <strong>of</strong> treatment response in<br />

patients with gastrointestinal stromal tumor (N = 20). Long-term clinical observation was used<br />

as a reference standard.


In primary disease staging, <strong>PET</strong>-<strong>CT</strong> imaging is more effective as a diagnostics method than<br />

<strong>CT</strong>. The difference between the diagnostic methods compared was statistically significant (p<br />

< 0.0001). In terms <strong>of</strong> response to treatment, significant congruence <strong>of</strong> results between the<br />

method compared and the reference standard was identified only for <strong>PET</strong>-<strong>CT</strong>. When <strong>PET</strong>-<strong>CT</strong><br />

scanning was used, response to treatment was diagnosed correctly in 95%, 100% and 100%<br />

patients in 1, 3 and 6-month follow-up periods respectively. For <strong>CT</strong>, the respective values were<br />

44%, 60% and 57%. The difference between particular diagnostic methods reached statistical<br />

significance in the 1 st month (p = 0.001).<br />

217


16. COMPARATIVE EFFICACY ANALYSIS OF THE <strong>PET</strong>-<strong>CT</strong> AND<br />

<strong>CT</strong> IN ASSESSING RESIDUAL LESIONS IN COLORE<strong>CT</strong>AL<br />

LIVER METASTASIS<br />

218<br />

16.1. Results <strong>of</strong> primary trial search<br />

As a result <strong>of</strong> searching medical databases, one primary prospective clinical study was<br />

found (Veit 2006; tab. 160, app. 18.1) that compares the diagnostic efficacy <strong>of</strong> <strong>PET</strong> coupled<br />

with roentgen <strong>CT</strong> in detecting residual lesions following radi<strong>of</strong>requency ablation <strong>of</strong> colorectal<br />

liver metastases.<br />

16.2. Population characteristics<br />

Included in the trial were 13 patients (11 men, 2 women) aged 55 to 71 (63 on average)<br />

with colorectal cancer and liver metastases confirmed histopathologically, who, due to<br />

contraindications to surgery, were classified for high-current percutaneous radio-frequency<br />

ablation. The patients included in the trial had contraindications to surgical resection <strong>of</strong><br />

metastasis (recurring liver metastasis after hepatectomy, cardiologic contraindications to<br />

surgery, functional liver anomaly, or metastasis located close to a blood vessel, which makes<br />

surgery impossible).<br />

16.3. Description <strong>of</strong> intervention<br />

16.3.1. <strong>PET</strong>-<strong>CT</strong> imaging<br />

<strong>PET</strong>-<strong>CT</strong> was performed using a Biograph <strong>PET</strong>-<strong>CT</strong> system (Siemens Medical Solutions,<br />

H<strong>of</strong>fman Estates, IL, USA) which consisted <strong>of</strong> a <strong>CT</strong> scanner with dual-slice configuration and a<br />

<strong>PET</strong> scanner. <strong>CT</strong> scans were taken at the beginning <strong>of</strong> the examination. Before whole-body<br />

examination, 140 ml <strong>of</strong> ionized contrast medium was administered intravenously; 0.2% water<br />

solution <strong>of</strong> locust bean gum as negative contrast and 2.5% <strong>of</strong> mannitol were given orally. Prior<br />

to guided examination <strong>of</strong> the liver, 100 ml <strong>of</strong> ionised contrast medium was administered.<br />

<strong>PET</strong> scans were done after <strong>CT</strong> examination. F18-deoxyglucose was used as marker; activity<br />

<strong>of</strong> the radiomarker was 350 MBq per dose. The acquisition time for a single scan was adjusted<br />

to the patient’s weight, and was 3 minutes per bed position for a patient with body weight


elow 65 kg, 4 minutes for patients with body weight between 65 and 85, and 5 minutes per<br />

bed position for a patient with body weight above 85 kg.<br />

A description <strong>of</strong> the equipment used in <strong>PET</strong>-<strong>CT</strong> examinations is presented in table 98.<br />

Table 98.<br />

Scanner and <strong>PET</strong>-<strong>CT</strong> procedure<br />

Type <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

<strong>PET</strong>-<strong>CT</strong> Biograph<br />

system<br />

<strong>PET</strong> marker<br />

Radiomarker<br />

activity [MBq]<br />

18F-deoxyglukose 350<br />

Type <strong>of</strong> <strong>CT</strong><br />

contrast medium<br />

intravenous ionised<br />

contrast medium;<br />

contrast medium<br />

producing<br />

negative<br />

amplification <strong>of</strong> the<br />

large intestine area<br />

Comments<br />

whole-body examination<br />

before ablation;<br />

guided examination <strong>of</strong><br />

the liver within two days<br />

<strong>of</strong> ablation<br />

All the patients had whole-body <strong>PET</strong>-<strong>CT</strong> scans (from cranium to upper thigh) one day<br />

before ablation; follow-up guided examination <strong>of</strong> the liver was carried out for all the patients<br />

2 days before surgery. For patients excluded from the trial within two days <strong>of</strong> ablation, follow-<br />

up <strong>PET</strong>-<strong>CT</strong> scans had the same coverage as pre-ablation tests (whole body).<br />

The image obtained as a result <strong>of</strong> <strong>PET</strong> and <strong>CT</strong> scan fusion was analyzed. <strong>PET</strong>-<strong>CT</strong> findings<br />

were interpreted by two experienced nuclear medicine doctors; in case <strong>of</strong> discrepancies,<br />

the result was determined by consensus. The doctors had access to the clinical data <strong>of</strong> the<br />

patients examined, except for <strong>CT</strong> findings.<br />

In order to detect residual lesions, images obtained using each imaging method before<br />

and after ablation was analyzed.<br />

Focuses <strong>of</strong> intense glucose metabolism found by <strong>PET</strong>-<strong>CT</strong> scanning next to necrosis areas<br />

resulting from ablation were considered indicative <strong>of</strong> residual lesions. Also, a qualitative and<br />

quantitative evaluation <strong>of</strong> focuses <strong>of</strong> increased glucose uptake was done.<br />

16.3.2. Diagnostic technology compared<br />

<strong>PET</strong>-<strong>CT</strong> findings were compared to the results from contrast <strong>CT</strong> obtained at the time <strong>of</strong> <strong>PET</strong>-<br />

<strong>CT</strong> scanning. <strong>CT</strong> findings were interpreted by two experienced radiologists; if opinions were<br />

divided, the result was agreed by consensus. The radiologists who performed the ablation did<br />

not participate in <strong>CT</strong> interpretation.<br />

Irregular peripheral contrast amplification as well as multi-nodular edges <strong>of</strong> a lesion were<br />

considered indicative <strong>of</strong> residual lesions. When residual lesions were detected, ablation was<br />

repeated during the same session.<br />

219


220<br />

16.3.3. Reference test<br />

Imaging diagnoses were verified based on the findings <strong>of</strong> liver imaging tests (<strong>CT</strong>, <strong>PET</strong>-<strong>CT</strong>,<br />

MRI) carried out in all patients after the ablation, laboratory tests results and clinical<br />

observation. For two patients results <strong>of</strong> histopathological test <strong>of</strong> material from guided biopsy<br />

were available. In six patients, a higher concentration level <strong>of</strong> colorectal neoplasm markers<br />

was identified.<br />

In case <strong>of</strong> tumor recurrence, another <strong>CT</strong>-guided ablation <strong>of</strong> the recurrence areas was<br />

performed during the follow-up period.<br />

The average follow-up period was 393 days (between 205 and 720).<br />

16.4. Findings<br />

Ablation was performed in 13 patients with colorectal liver metastasis; 11 had a <strong>PET</strong>-<strong>CT</strong><br />

examination immediately after ablation. Two patients (each with one metastasis and one<br />

radio-frequency current ablation) had no follow-up <strong>PET</strong>-<strong>CT</strong> examination directly after ablation<br />

due to poor general condition; for these only <strong>CT</strong> and MRI findings were available.<br />

19 ablation sessions were performed on 11 patients with 16 metastases confirmed<br />

(including repeated ablation). For these patients, 15 <strong>PET</strong>-<strong>CT</strong> examinations were performed<br />

within 2 days after the procedure; the remaining 4 examinations were performed to evaluate<br />

treatment effects during the observation period, its median being 58 days. In total, 32 follow-<br />

up <strong>PET</strong>-<strong>CT</strong> examinations were performed (including 19 examinations mentioned above).<br />

The authors <strong>of</strong> the study provide data on the sensitivity and accuracy <strong>of</strong> both methods in<br />

evaluating residual lesions after radio-frequency ablation <strong>of</strong> colorectal liver metastases.<br />

Table 99 presents the findings.<br />

Table 99.<br />

Sensitivity and accuracy <strong>of</strong> the diagnostic methods compared in detecting residual lesions after ablation <strong>of</strong><br />

colorectal liver metastasis, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong><br />

Parameter<br />

Diagnostic method<br />

<strong>PET</strong>-<strong>CT</strong> <strong>CT</strong><br />

Sensitivity 65% 44%<br />

Accuracy<br />

68%<br />

47%


<strong>PET</strong>-<strong>CT</strong> sensitivity in detecting residual lesions after radio-frequency ablation <strong>of</strong> colorectal<br />

liver metastasis is 65%, and is higher than <strong>CT</strong> sensitivity, which is 44%. That means that the<br />

probability <strong>of</strong> positive result for a patient with residual lesions confirmed in the liver after<br />

ablation is 65% for <strong>PET</strong>-<strong>CT</strong> and 44% for <strong>CT</strong> with contrast medium. The authors do not report on<br />

the statistical significance <strong>of</strong> the difference.<br />

The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 68%, and is higher than the accuracy <strong>of</strong> conventional <strong>CT</strong><br />

imaging (47%).<br />

In four patients with five metastases confirmed, residual lesions were detected by <strong>PET</strong>-<strong>CT</strong> in<br />

the course <strong>of</strong> further observation, while <strong>CT</strong> findings were negative. The ablation procedure<br />

was repeated. The use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> with these patients allowed revising treatment early on, to<br />

the patients’ benefit (longer disease-free period <strong>of</strong> life).<br />

16.5. Results<br />

As a result <strong>of</strong> searching through medical databases, a single primary prospective clinical<br />

trial was found (Veit 2006) that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong><br />

imaging in detecting residual lesions <strong>of</strong> colorectal liver metastasis following a therapy that<br />

used radio-frequency current ablation (N = 13).<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting residual disease is 65%, and is higher than that <strong>of</strong> <strong>CT</strong>,<br />

which stands at 44%. The authors provide no data on the statistical significance <strong>of</strong> the results<br />

above.<br />

The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in detecting residual lesions is 68%, and is higher than the accuracy<br />

<strong>of</strong> <strong>CT</strong>, which stands at 47%.<br />

Analysis <strong>of</strong> imaging methods compared showed that in detecting residual lesions <strong>of</strong><br />

colorectal liver metastasis following radi<strong>of</strong>requency ablation, a higher efficacy is<br />

demonstrated by <strong>PET</strong>-<strong>CT</strong> diagnostics than by <strong>CT</strong> imaging.<br />

221


17. COMPARATIVE ANALYSIS OF THE EFFICACY OF <strong>PET</strong>-<strong>CT</strong> VS<br />

OTHER IMAGING TECHNIQUES (MRI OR <strong>CT</strong>) IN THE<br />

STAGING OF NEOPLASM IN VARIED LOCATIONS<br />

222<br />

17.1. Neoplasm in varied locations – disease staging (<strong>PET</strong>-<strong>CT</strong> vs.<br />

MRI)<br />

17.1.1. Results <strong>of</strong> trial search<br />

As a result <strong>of</strong> searching medical databases, two primary prospective trials were<br />

found: Antoch 2003 (tab. 140, app. 18.1), and Schmidt 2005 (tab. 141, app. 18.1),,<br />

which discussed the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in staging neoplasm in varied locations.<br />

For both <strong>of</strong> these trials, hybrid <strong>PET</strong>-<strong>CT</strong> was compared with magnetic resonance<br />

(MRI), and both methods were verified based on a reference test, which was<br />

histopathological examination and/or clinical observation.<br />

17.1.2. Population characteristics<br />

Patients with neoplasm in varied locations participated in all the trials reviewed. Pre-<br />

surgery staging was performed for 84% <strong>of</strong> the patients included in Antoch 2003, and for<br />

7% in Schmidt 2005. Staging in case <strong>of</strong> suspected recurrence was performed for 16% <strong>of</strong><br />

patients in Antoch 2003 and for 15% in Schmidt 2005. In Schmidt 2005 covered additional<br />

patients with whom the diagnostic methods compared were used for T-staging (10%)<br />

and post-treatment restaging (68%).<br />

In neither <strong>of</strong> the trials, the patients inclusion or exclusion criteria were specified.<br />

Table 100 provides initial characteristics <strong>of</strong> patients included in the trials.<br />

Table 100.<br />

Initial characteristics <strong>of</strong> patients included in trials<br />

Parameter<br />

Size <strong>of</strong> population<br />

Mean age [years]<br />

Male proportion<br />

lung and bronchial cancers<br />

cancer <strong>of</strong> unknown<br />

primary origin<br />

head and neck cancer<br />

Antoch 2003<br />

98<br />

58<br />

64%<br />

30%<br />

12%<br />

13%<br />

Schmidt 2005<br />

38<br />

56**<br />

44%**<br />

0%<br />

10,5%<br />

0%<br />

Total<br />

136<br />

57*<br />

58%<br />

22%<br />

12%<br />

9%


Cancer type<br />

(patient<br />

proportion)<br />

melanoma<br />

genitourinary cancers<br />

esophageal cancer<br />

thyroid cancer<br />

mesothelioma<br />

liver cancer<br />

bone cancer<br />

breast cancer<br />

testicular cancer<br />

lymphoma<br />

sarcoma<br />

13%<br />

* average weighted with patient population number participating in 2 studies<br />

**data for 41 patients<br />

8%<br />

6%<br />

6%<br />

6%<br />

10,5%<br />

115 patients were initially included in Antoch 2003, contraindications to imaging were<br />

identified in 17 <strong>of</strong> them (acute allergic reaction to intravenous contrast medium in 8<br />

patients; in 9 patients contraindications to MRI were identified for reasons such as heart<br />

simulators, metal implants or claustrophobia).<br />

41 patients were included in Schmidt 2005, but only 38 were qualified. The reasons for<br />

excluding two patients were contraindications to MRI, such as heart simulator and<br />

claustrophobia. For one patient, MRI testing was stopped due to strong pain caused by<br />

advanced metastasis to bones.<br />

The total <strong>of</strong> 136 patients were included in this <strong>analysis</strong>, with the average age <strong>of</strong> 56-68<br />

and the male proportion <strong>of</strong> 44–64%.<br />

17.1.3. Description <strong>of</strong> intervention<br />

17.1.3.1 <strong>PET</strong>-<strong>CT</strong><br />

Biograph produced by Siemens Medical Solutions (H<strong>of</strong>fman Estates, USA) was used for<br />

disease staging in Antoch 2003, and Philips Gemini by Philips Medical Systems was used<br />

in Schmidt 2005.<br />

For both trials included, the radiopharmaceutical used was FDG (fluoro-deoxy-glucose),<br />

with the activity <strong>of</strong> 350 MBq in Antoch 2003, and 202-372 MBq in Schmidt 2005. The<br />

radiopharmaceutical was applied 60 minutes before <strong>PET</strong>-<strong>CT</strong> scanning in both trials. In<br />

Schmidt 2005, FDG was preceded by 20 mg <strong>of</strong> furosemide and buskopan administered<br />

intravenously.<br />

3%<br />

2%<br />

0%<br />

0%<br />

0%<br />

0%<br />

0%<br />

37%<br />

5%<br />

0%<br />

0%<br />

0%<br />

29%<br />

3%<br />

3%<br />

3%<br />

12%<br />

6%<br />

15%<br />

6%<br />

4%<br />

2%<br />

1%<br />

8%<br />

1%<br />

1%<br />

1%<br />

223


224<br />

Table 101 gives details <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong> tests.<br />

Table 101.<br />

Description <strong>of</strong> intervention<br />

Trial<br />

Antoch<br />

2003<br />

Schmidt<br />

2005<br />

<strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

type<br />

Biograph<br />

Philips<br />

Gemini<br />

<strong>PET</strong>-<strong>CT</strong> scanner<br />

manufacturer<br />

Siemens Medical<br />

Solutions (H<strong>of</strong>fman<br />

Estates, USA)<br />

Philips Medical<br />

Systems<br />

Radiopharma<br />

ceutical type<br />

and<br />

administration<br />

method<br />

FDG<br />

intravenously<br />

FDG<br />

Study range<br />

whole body<br />

no data<br />

Radiomarker<br />

activity<br />

[MBq]<br />

350<br />

202–372<br />

In Schmidt 2005, patients could not eat for at least 6 hours before, so the sugar level did<br />

not exceed 120 mg/dL. In Antoch 2003, before giving the radiopharmaceutical, the correct<br />

level <strong>of</strong> glucose concentration in blood was checked.<br />

In both studies, in order to perform the <strong>CT</strong> part <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning, the patients received<br />

contrast medium intravenously. In Antoch 2003, large intestine enlargement was achieved by<br />

administering glucose-free barium.<br />

The average <strong>PET</strong>-<strong>CT</strong> testing time was 27 minutes (range: 22-39 minutes) in Antoch 2003,<br />

and 103 minutes in Schmidt 2005. The authors <strong>of</strong> the latter noted that it took 60 minutes to<br />

prepare a patient and 43 minutes to scan.<br />

For both studies, the <strong>PET</strong>-<strong>CT</strong> scans were assessed by a radiologist and a nuclear<br />

medicine expert. The assessors were blinded to the results <strong>of</strong> imaging studies other than <strong>PET</strong>-<br />

<strong>CT</strong>.<br />

17.1.3.2 Compared diagnostic test<br />

For the two studies assessed in this part <strong>of</strong> the <strong>analysis</strong> (Antoch 2003, Schmidt 2005), <strong>PET</strong>-<strong>CT</strong><br />

scanning was compared with magnetic resonance imaging (MRI).<br />

In Antoch 2003, magnetic resonance was performed using Sonata System 1.5-T (Siemens<br />

Medical Solutions, Erlangen, Germany), and in Schmidt 2005, the Magnetom Avanto 1.5-T<br />

scanner was used (Siemens Medical Solutions).<br />

Table 102 provides a detailed description <strong>of</strong> the magnetic resonance tests in each trial.


Table 102.<br />

Description <strong>of</strong> diagnostic tests compared<br />

Study<br />

Antoch 2003<br />

Schmidt 2005<br />

MRI scanner type<br />

Sonata System 1.5-T<br />

Magnetom Avanto<br />

1.5-T<br />

MRI scanner<br />

manufacturer<br />

Siemens Medical<br />

Solutions<br />

Siemens Medical<br />

Solutions<br />

Contrast medium<br />

type and<br />

administration<br />

method<br />

intravenous<br />

contrast medium<br />

intravenous<br />

contrast medium<br />

Study range<br />

whole body<br />

whole body<br />

In Antoch 2003, axial scanning from head to thigh was done, which corresponded to the<br />

area scanned using <strong>PET</strong>-<strong>CT</strong>. Initially, the thorax and abdomen were scanned without a<br />

contrast medium used, and than the paramagnetic contrast medium Megnevist (Schering,<br />

Germany) was given intravenously at 3 ml/sec and the dose <strong>of</strong> 0.2 mmol/kg, and after 12<br />

seconds, seven other scans were performed enhanced by contrast in the following order:<br />

abdomen (arterial phase), thorax, abdomen (portal-venous phase), pelvis, thighs, head and<br />

abdomen (late venous phase).<br />

In Schmidt 2005, the whole body was scanned using parallel acquisition techniques (PAT).<br />

Initially, the head/neck, pelvis, thighs, calves and thorax/abdomen were scanned. After<br />

gadolinum-DTPA was administered at 3 ml/sec with the dose <strong>of</strong> 2 mmol/ks, and 20 ml <strong>of</strong> salt<br />

solution was given, the liver, brain and whole abdomen were scanned axially. Another PAT<br />

agent was used for axial imaging <strong>of</strong> brain, lungs and abdomen, and for sagittal imaging <strong>of</strong><br />

spine and frontal imaging <strong>of</strong> calfs.<br />

The average MRI testing time was 26 minutes (range: 20-34 minutes) in Antoch<br />

2003, and 70 minutes (range: 56-76 minutes) in Schmidt 2005. The authors <strong>of</strong> the latter noted<br />

that the scanning alone took approx. 55 minutes.<br />

17.1.3.3 Reference test<br />

In both studies, histopathological examination and/or imaging follow-up were the<br />

reference test.<br />

In Antoch 2003, 47% patients underwent tumor resection, and the tumor was assessed<br />

pathologically, therefore only for these patients the accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> vs. MRI was<br />

evaluated. Similarly, metastases to lymph nodes was verified based on pathological tests for<br />

44% <strong>of</strong> the patients, and distant metastases for 14%. Follow-up was the reference method<br />

for patients without histopathological verification <strong>of</strong> N and M features. The average<br />

observation time was 273 days (range: 75–515 days). During that period clinical<br />

assessment, laboratory tests and diagnostic imaging (<strong>CT</strong>, MRI and/or <strong>PET</strong>-<strong>CT</strong>) were done,<br />

225


followed by histopathological diagnostics. Data on the reference test results were<br />

collected by a doctor blinded to the results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI scanning.<br />

226<br />

In Schmidt 2005, clinical, histological and radiological tests in the course <strong>of</strong> 6-month-long<br />

observations were used as reference methods for all pathological changes detected,<br />

especially to verify divergent and questionable data. 14 targeted <strong>CT</strong>-guided biopsies, Tests<br />

done as part <strong>of</strong> follow-up included 14 <strong>CT</strong> scans, 14 <strong>PET</strong> scans, 16 <strong>PET</strong>-<strong>CT</strong> scans, 14 bone<br />

scintigraphies, 10 MRI tests, 1 whole body MRI, 18 X-ray pictures and 5 ultrasound test.<br />

Histological test were done in case a primary tumor or disease recurrence were suspected.<br />

17.1.4. Findings<br />

17.1.4.1 Diagnostic accuracy in detecting primary and recurring carcinomas<br />

This end point was evaluated only in Schmidt 2005. The authors noted that both <strong>PET</strong>-<strong>CT</strong><br />

and MRI helped detect 7 cancer cases, including 3 primary and 4 recurring tumors in 38<br />

patients qualified for the clinical trial.<br />

The number <strong>of</strong> patients with true positive (TP), false positive (FP), false negative (FN)<br />

and true negative (TN) result <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI as calculated based on the data<br />

provided by the authors in Schmidt 2005 is presented in table 103.<br />

Table 103.<br />

Number <strong>of</strong> patients with TP, FP, FN and TN results in detecting cancer in patients with varied location <strong>of</strong> the disease<br />

using <strong>PET</strong>-<strong>CT</strong> and MRI.<br />

TP<br />

FP<br />

<strong>PET</strong>-<strong>CT</strong><br />

FN<br />

7*<br />

0*<br />

0*<br />

*Calculated based on data available<br />

TN<br />

31*<br />

Table 104 presents the diagnostic parameters: sensitivity (Se), specificity (Sp),<br />

accuracy (Acc), Positive likelihood ratio (LR+), negative likelihood ratio (LR-) and<br />

diagnostic odds ratio (DOR) as calculated for the methods compared (<strong>PET</strong>-<strong>CT</strong> and MRI).<br />

Table 104.<br />

Accuracy in detecting cancer in patients with the disease in varied location using <strong>PET</strong>-<strong>CT</strong> vs MRI .<br />

Parameter<br />

Se (95% CI)<br />

Sp (95% CI)<br />

LR+ (95% CI)<br />

LR- (95% CI)<br />

TP<br />

6*<br />

<strong>PET</strong>-<strong>CT</strong><br />

FP<br />

0*<br />

100% (59; 100)*<br />

100% (89; 100)*<br />

60,00* (3,81; 944,10)*<br />

0,06* (0,00; 0,93)*<br />

MRI<br />

FN<br />

1*<br />

MRI<br />

86% (42; 100)*<br />

TN<br />

31*<br />

100% (89; 100)*<br />

52,00* (3,26; 829,97)*<br />

0,19* (0,04; 0,81)*


Acc (95% CI)<br />

DOR<br />

*Calculated based on data available<br />

100% (91; 100)*<br />

945,00* (17,31; 51595)*<br />

97% (86; 100)*<br />

273,00* (9,97; 7477,05)*<br />

Based on the data above, <strong>PET</strong>-<strong>CT</strong> sensitivity is 100% (95% CI: 59; 100), and MRI sensitivity is<br />

86% (95% CI: 42; 100), which means that the probability <strong>of</strong> positive results for patients with<br />

primary or recurring cancer is 100% and 86% for <strong>PET</strong>-<strong>CT</strong> and MRI, respectively.<br />

The specificity <strong>of</strong> both methods is 100% (95% CI: 89; 100), so the probability <strong>of</strong> negative<br />

result for healthy patients is 100% for both <strong>PET</strong>-<strong>CT</strong> and MRI.<br />

The diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 100% (95% CI: 91; 100), and for MRI the value is 97%<br />

(95% CI: 86; 100). No statistically significant differences between the groups were observed<br />

for none <strong>of</strong> the parameters above.<br />

The positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> is 60,00 (95% CI: 3,81; 944,10), so the probability <strong>of</strong><br />

positive results for patients with primary and recurring cancers is 60 times higher than for<br />

healthy patients. The positive likelihood ratio (LR+) for magnetic resonance is 52,00 (95% CI:<br />

3,26; 829,97). It means that the probability <strong>of</strong> positive results for cancer patients is 52 times<br />

higher than for patients with no cancer detected using the index test.<br />

The negative likelihood ratio (LR-) for <strong>PET</strong>-<strong>CT</strong>, is 0.06 (95% CI: 0,00; 0,93), so the probability <strong>of</strong><br />

negative results in patients with primary or recurring cancers is 0.06 <strong>of</strong> that for patients without<br />

cancer. The negative likelihood ratio (LR-) for MRI is 0.19 (0.04; 0.81), so the probability <strong>of</strong><br />

negative result for cancer patients is 0.19 <strong>of</strong> that for patients without the disease.<br />

The diagnostic odds ratio (DOR) calculated for <strong>PET</strong>-<strong>CT</strong> is 945 (95% CI: 17,31; 51595), which<br />

means that the probability <strong>of</strong> positive results is 945 times higher for patients with primary or<br />

recurring cancers than for patients not diagnosed with cancer using the reference test. The<br />

diagnostic odds ratio (DOR) for MRI is 273 (95% CI: 9,97; 7477,05), which means that the<br />

probability <strong>of</strong> positive results in patients with primary or recurring cancers is 273 times higher<br />

than for patients without such cancers.<br />

In Schmidt 2005, four patients with primary tumors in unknown locations were diagnosed<br />

using <strong>PET</strong>-<strong>CT</strong> and MRI in order to identify the locations. However, for none <strong>of</strong> the patients the<br />

primary origin was detected using the diagnostic methods compared. No primary tumor<br />

was identified in these patients during 6 months-long observations.<br />

17.1.4.2 T-staging<br />

T-status was assessed in both trials included (Antoch 2003, Schmidt 2005).<br />

In all tests, the T-status was assessed based on the TNM staging system created by the<br />

AJCC (American Joint Committee on Cancer). <strong>PET</strong>-<strong>CT</strong> and MRI results were verified<br />

histopathologically and based on radiologcal follow-up.<br />

227


228<br />

The consistency <strong>of</strong> the T-status established using the diagnostic methods<br />

compared with the reference test results as described by the authors is given in<br />

table 105.<br />

Table 105.<br />

Consistency <strong>of</strong> the T feature with the reference test for patients with neoplasm in varied locations, <strong>PET</strong>-<strong>CT</strong> vs MRI<br />

Study<br />

Antoch<br />

2003<br />

N*<br />

46<br />

Correct<br />

staging<br />

(accuracy)<br />

n (%)<br />

37 (80%)<br />

Overstage<br />

4 (9%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

5 (11%)<br />

Schmidt<br />

2005<br />

38 37 (97%) 0 (0%)<br />

1 (3%)<br />

*Number <strong>of</strong> patients verified against the reference test results<br />

Correct<br />

staging<br />

(accuracy)<br />

n (%)<br />

24 (52%)<br />

37 (97%)<br />

Overstaged<br />

6 (13%)<br />

0 (0%)<br />

MRI<br />

Incorrect staging<br />

n (%)<br />

Based on the data above, for all the tests, the proportion <strong>of</strong> correctly staged<br />

patients was higher for <strong>PET</strong>-<strong>CT</strong> than for magnetic resonance (MRI). <strong>PET</strong>-<strong>CT</strong> accuracy<br />

is 80 and 97%, and MRI accuracy is 52% and 97%. Overstaged and understaged<br />

results are 0 and 9% and 3 and 11% respectively for <strong>PET</strong>-<strong>CT</strong>, and 0 and 13% and 3<br />

and 35% respectively for MRI.<br />

Understaged<br />

16 (35%)<br />

The authors in Antoch 2003 described <strong>PET</strong>-<strong>CT</strong> as demonstrating diagnostic accuracy that is<br />

statistically significantly higher than MRI (p < 0.001). Differences in accuracy between <strong>PET</strong>-<br />

<strong>CT</strong> and MRI were not statistically significant in Schmidt 2005.<br />

In Schmidt 2005, the sensitivity and specificity <strong>of</strong> the methods compared were the same:<br />

86% and 100% respectively.<br />

Graph 28 illustrates a meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostic accuracy in T-staging. As there<br />

was significant heterogeneity between trials (p = 0.0122) meta-<strong>analysis</strong> was performed<br />

using the random effects method.<br />

1 (3%)


Graph 28.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in T-staging in patients with neoplasm in varied locations<br />

Antoch 2003 0,80 (0,66, 0,91)<br />

Schmidt 2005 0,97 (0,86, 1,00)<br />

Meta-<strong>analysis</strong> result [random]<br />

0,6 0,7 0,8 0,9 1,0<br />

Diagnostic accuracy (95% confidence interval)<br />

0,89 (0,68, 1,00)<br />

<strong>PET</strong>-<strong>CT</strong> diagnostic accuracy as calculated by meta-<strong>analysis</strong> <strong>of</strong> the two trials is 89% (95% CI:<br />

68; 100).<br />

Graph 29 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> T-staging using the<br />

magnetic resonance technology. As there was significant heterogeneity between trials (p <<br />

0.0001) the meta-<strong>analysis</strong> was performed using the random effects method.<br />

229


Graph 29.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> MRI in T-staging in patients with neoplasm in varied locations<br />

230<br />

The average value <strong>of</strong> the diagnostic accuracy <strong>of</strong> magnetic resonance (MRI) in T-staging<br />

is 79% (95% CI: 26; 99).<br />

The average values <strong>of</strong> diagnostic efficacy calculated for <strong>PET</strong>-<strong>CT</strong> and MRI are given in<br />

table 106.<br />

Antoch 2003<br />

Result <strong>of</strong> meta-<strong>analysis</strong><br />

[random]<br />

Schmidt 2005 0,97 (0,86, 1,00)<br />

Table 106.<br />

Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI accuracy in T-staging in patients with neoplasm in varied locations.<br />

Parameter<br />

Acc (95% CI)<br />

0,0 0,2 0,4 0,6 0,8 1,0<br />

<strong>PET</strong>-<strong>CT</strong><br />

89% (68; 100)<br />

MRI<br />

79% (26; 99)<br />

Based on the data above, the diagnostic accuracy in T-staging is higher for <strong>PET</strong>-<strong>CT</strong> than<br />

for magnetic resonance (MRI).<br />

Diagnostic accuracy (95% confidence interval)<br />

Graph 30 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the proportion <strong>of</strong> patients for whom the results <strong>of</strong> T-<br />

staging using <strong>PET</strong>-<strong>CT</strong> or MRI scanning were consistent with the reference test.<br />

0,52 (0,37, 0,67)<br />

0,79 (0,26, 0,99)


Graph 30.<br />

Odds ratio for the consistency between the reference test and T- staging results for patients with neoplasm in varied<br />

locations, <strong>PET</strong>-<strong>CT</strong> vs. MRI<br />

Antoch 2003 3,77 (1,37, 10,83)<br />

Schmidt 2005 1,00 (0,01, 80,59)<br />

Meta-<strong>analysis</strong> result [fixed] 3,29 (1,37, 7,90)<br />

The odds ratio as calculated by meta-<strong>analysis</strong> is 3.29 (95% CI: 1.37; 7.90), which means<br />

that the probability <strong>of</strong> T-staging results consistent with reference test results for <strong>PET</strong>-<strong>CT</strong> is 3.29<br />

times higher than for magnetic resonance. The result is statistically significant.<br />

In order to obtain one additional case <strong>of</strong> correct T-staging, it is necessary to use <strong>PET</strong>-<strong>CT</strong><br />

instead <strong>of</strong> MRI with 7 patients, NNT = 7 (95% CI: 4; 20).<br />

17.1.4.3 N-staging<br />

Disease staging with respect to metastasis to lymph nodes was analyzed in two <strong>of</strong> the trials<br />

included.<br />

In both studies (Antoch 2003 and Schmidt 2003), disease staging was done based on the<br />

TNM lung cancer staging system developed by the AJCC (American Joint Committee on<br />

Cancer).<br />

In Antoch 2003, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> vs. MRI was evaluated for 98 patients,<br />

and the results were verified by the reference test. In Schmidt 2005, 120 lymph nodes from 38<br />

patients were examined, out <strong>of</strong> which 60 were affected by metastasis.<br />

Table 107 presents the consistency between the N-staging results obtained using the<br />

diagnostic methods compared and the results <strong>of</strong> the reference test as described by the<br />

authors in both trials.<br />

0,01 0,1 0,2 0,5 1 2 5 10 100<br />

Odds ratio (95% confidence interval)<br />

231


Table 107.<br />

Consistency <strong>of</strong> N-staging results with the reference test in patients with neoplasm in varied locations, <strong>PET</strong>-<strong>CT</strong> vs. MRI<br />

232<br />

Study<br />

Antoch<br />

2003<br />

N*<br />

98<br />

Correct<br />

staging<br />

(accuracy)<br />

n (%)<br />

91 (93%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Overstaged<br />

5 (5%)<br />

Schmidt<br />

2005<br />

38 37 (97%) 1 (3%)<br />

*Number <strong>of</strong> patients verified using reference test<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

2 (2%)<br />

0 (0%)<br />

Correct<br />

staging<br />

(accuracy)<br />

n (%)<br />

77 (79%)<br />

32 (82%)<br />

MRI<br />

Overstaged<br />

13 (13%)<br />

3 (8%)<br />

Incorrect staging<br />

n (%)<br />

Based on the data above, for all the tests evaluated, the proportion <strong>of</strong> correctly<br />

diagnosed patients was higher for <strong>PET</strong>-<strong>CT</strong> than for magnetic resonance (MRI). <strong>PET</strong>-<strong>CT</strong><br />

accuracy is 93% and 97%, and the accuracy <strong>of</strong> MRI is 79% and 82%. The proportion<br />

<strong>of</strong> overstaged and understaged cases is 3%–5% and 0%–2% respectively for <strong>PET</strong>-<strong>CT</strong><br />

and 8%–13% and 8% respectively for MRI.<br />

In Antoch 2003, the differences in accuracy between the diagnostic methods<br />

compared were statistically significant (p = 0,001). In Schmidt 2005, the differences<br />

were not statistically significant.<br />

Graph 31 illustrates a meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> accuracy in N-staging.<br />

Graph 31.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in N-staging in patients with neoplasm in varied locations<br />

Antoch 2003 0,93 (0,86, 0,97)<br />

Schmidt 2005 0,97 (0,86, 1,00)<br />

Result <strong>of</strong> meta-<strong>analysis</strong> [fixed] 0,94 (0,89, 0,97)<br />

0,85 0,90 0,95 1,00<br />

Diagnostic accuracy (95% confidence interval)<br />

Understaged<br />

8 (8%)<br />

3 (8%)


<strong>PET</strong>-<strong>CT</strong> diagnostic accuracy as calculated by meta-<strong>analysis</strong> <strong>of</strong> the two studies is 94% (95%<br />

CI: 89; 97).<br />

The results <strong>of</strong> a meta-<strong>analysis</strong> <strong>of</strong> the accuracy <strong>of</strong> MRI in N-staging are presented in graph<br />

32.<br />

Graph 32.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> MRI in N-staging in patients with neoplasm in varied locations.<br />

Antoch 2003 0,93 (0,86, 0,97)<br />

Schmidt 2005 0,97 (0,86, 1,00)<br />

Meta-<strong>analysis</strong> result [fixed] 0,94 (0,89, 0,97)<br />

The average MRI diagnostic accuracy as calculated by meta-<strong>analysis</strong> is 80% (95% CI: 73; 86).<br />

The average values <strong>of</strong> diagnostic efficacy <strong>of</strong> N-staging calculated for <strong>PET</strong>-<strong>CT</strong> and MRI is<br />

given in table 108.<br />

Table 108.<br />

Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI accuracy in N-staging in patients with neoplasm in varied locations.<br />

Parameter <strong>PET</strong>-<strong>CT</strong> MRI<br />

Acc (95% CI) 94% (89; 97) 80% (73; 86)<br />

Based on the data above, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in the assessment <strong>of</strong> the<br />

involvement <strong>of</strong> lymph nodes is higher that that <strong>of</strong> magnetic resonance (MRI).<br />

Graph 33 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the proportion <strong>of</strong> patients for whom the results<br />

<strong>of</strong> N-staging using <strong>PET</strong>-<strong>CT</strong> vs. MRI scanning were consistent with the reference tests<br />

results.<br />

0,85 0,90 0,95 1,00<br />

Diagnostic accuracy (95% confidence interval)<br />

233


Graph 33.<br />

Odds ratio for the consistency between the reference test and N-staging result in patients with neoplasm in varied<br />

locations, <strong>PET</strong>-<strong>CT</strong> vs. MRI<br />

234<br />

Antoch 2003 3,55 (1,35, 10,36)<br />

Schmidt 2005 6,94 (0,76, 327,53)<br />

Meta-<strong>analysis</strong> results<br />

[fixed]<br />

The odds ratio is 4.00 (95% CI: 1.74; 9.19), so the probability <strong>of</strong> consistency between the<br />

results <strong>of</strong> N-staging and the reference test results is 4 times higher for <strong>PET</strong>-<strong>CT</strong> than for<br />

magnetic resonance. The result is statistically significant.<br />

In order to obtain one additional case <strong>of</strong> correct N-staging it is necessary to use <strong>PET</strong>-<strong>CT</strong><br />

instead <strong>of</strong> MRI with 8 patients with cancer; NNT = 8 (95% CI: 5; 17).<br />

Table 109 presents additional parameters concerning the diagnostic accuracy <strong>of</strong> the<br />

methods compared: sensitivity (Se) and specificity (Sp), as provided by the authors <strong>of</strong> the<br />

trials.<br />

Table 109.<br />

The diagnostic accuracy <strong>of</strong> N-staging in patients with neoplasm in varied locations. <strong>PET</strong>-<strong>CT</strong> vs. MRI<br />

Parametr<br />

0,5 1 2 5 10 100 1000<br />

Odds ration (95% confidence interval)<br />

<strong>PET</strong>-<strong>CT</strong> MRI<br />

Antoch 2003 Schmidt 2005<br />

Statistical<br />

significance<br />

(<strong>PET</strong>-<strong>CT</strong> vs<br />

MRI)<br />

<strong>PET</strong>-<strong>CT</strong> MRI<br />

4,00 (1,74, 9,19)<br />

Statistical<br />

significance<br />

(<strong>PET</strong>-<strong>CT</strong> vs<br />

MRI)<br />

Se 95% 79% bd 100% 79% bd<br />

Sp 92% 78% bd 95% 95% bd


The diagnostic sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 95% and 100% and for MRI it is 79% in both trials. The<br />

specificity reported by the authors <strong>of</strong> both studies is 92% and 95% for <strong>PET</strong>-<strong>CT</strong>, and 78%-95% for<br />

magnetic resonance.<br />

The diagnostic parameters calculated based on the number <strong>of</strong> lymph nodes affected by<br />

metastases was additionally reported in Schmidt 2005. Detailed results are given in table 110.<br />

Table 110.<br />

Diagnostic accuracy evaluation parameters for N-staging using <strong>PET</strong>-<strong>CT</strong> vs MRI, as calculated in Schmidt 2005 in<br />

relation to the number <strong>of</strong> lymph nodes assessed.<br />

Parameter<br />

Se<br />

n/N<br />

Sp<br />

n/N<br />

Number<br />

<strong>of</strong> lymph<br />

nodes assessed<br />

120<br />

<strong>PET</strong>-<strong>CT</strong><br />

98%<br />

59/60<br />

83%<br />

50/60<br />

MRI<br />

80%<br />

48/60<br />

75%<br />

45/60<br />

Statistical<br />

significance<br />

(<strong>PET</strong>-<strong>CT</strong> vs MRI)<br />

no data<br />

Considering the number <strong>of</strong> lymph nodes invaded by metastases, sensitivity and<br />

specificity are higher for <strong>PET</strong>-<strong>CT</strong> than for magnetic resonance, and are 98% and 83%<br />

respectively for <strong>PET</strong>-<strong>CT</strong>, and 80% and 75% respectively for MRI.<br />

Schmidt 2005 established that the median size <strong>of</strong> lymph nodes invaded by<br />

metastasis diagnosed based on <strong>PET</strong>-<strong>CT</strong> is 15 mm (SD = 8), and based on MRI it is 18 mm<br />

(SD = 8).<br />

In Schmidt 2005, <strong>PET</strong>-<strong>CT</strong> helped diagnose a greater number <strong>of</strong> lymph nodes in all<br />

size-based groups, and a statistically significantly greater number <strong>of</strong> small lymph<br />

nodes (below 10 mm). Two large lymph nodes (>20 mm) were additionally located in<br />

the mediastinal and retrotibial regions using this technology.<br />

17.1.4.4 Assessment <strong>of</strong> distant metastases (M feature)<br />

The assessment <strong>of</strong> cancer stage based on the TNM system with respect to the<br />

presence <strong>of</strong> distant metastasis was performed in two studies included in this <strong>analysis</strong>:<br />

Antoch 2003 and Schmidt 2005.<br />

In Antoch 2003, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was compared with MRI for 98 patients,<br />

and the results were verified using the reference method. In Schmidt 2005, 268 pathological<br />

lesions were assessed in 38 patients, including 191 malignant lesions and 77 benign lesions.<br />

The consistency <strong>of</strong> the results <strong>of</strong> M-staging one with the use <strong>of</strong> the technologies<br />

compared with the reference (index) test as reported by the authors is given in<br />

table 111.<br />

NS<br />

235


Table 111.<br />

Consistency <strong>of</strong> distant metastasis staging (M feature) with reference test in patients with neoplasm in varied locations,<br />

<strong>PET</strong>-<strong>CT</strong> vs MRI<br />

236<br />

Study<br />

Antoch<br />

2003<br />

N*<br />

98<br />

Correct<br />

staging<br />

(accuracy)<br />

n (%)<br />

92 (94%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Overstaged<br />

3 (3%)<br />

Schmidt<br />

2005<br />

38 37 (97%) 1 (3%)<br />

*Number <strong>of</strong> patients verified using reference test<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

3 (3%)<br />

0 (0%)<br />

Correct<br />

staging<br />

(accuracy)<br />

n (%)<br />

91 (93%)<br />

38 (100%)<br />

Overstaged<br />

3 (3%)<br />

0 (0%)<br />

MRI<br />

Incorrect staging<br />

n (%)<br />

Based on the data above the accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 94% and 97%, and the accuracy <strong>of</strong><br />

MRI is 93%–100%. Overstaged and understaged results stood at 3% and 0%–3% respectively<br />

for <strong>PET</strong>-<strong>CT</strong>, and 0%–3% and 0%–4% for MRI respectively.<br />

In Antoch 2005, slightly higher diagnostic accuracy was reported for <strong>PET</strong>-<strong>CT</strong> (94%) as<br />

compared to MRI (93%), but the differences were not statistically significant (p > 0.99). The<br />

differences between groups were not statistically significant in Schmidt 2005 either (p > 0,99).<br />

<strong>CT</strong>.<br />

Graph 34 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> M-staging using <strong>PET</strong>-<br />

Graph 34.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in M-staging in patients with neoplasm in varied locations<br />

Antoch 2003 0,94 (0,87, 0,98)<br />

Schmidt 2005 0,97 (0,86, 1,00)<br />

Results <strong>of</strong> meta-<strong>analysis</strong> [fixed]<br />

0,86 0,91 0,96 1,01<br />

Diagnostic accuracy (95%) confidence interval)<br />

0,94 (0,90, 0,98)<br />

Understaged<br />

4 (4%)<br />

0 (0%)


The <strong>PET</strong>-<strong>CT</strong> diagnostic accuracy calculated by meta-<strong>analysis</strong> <strong>of</strong> the two studies is 94% (95%<br />

CI: 90; 98).<br />

Graph 35 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> MRI in M-staging. As<br />

there was significant heterogeneity between trials (p = 0.0359) the meta-<strong>analysis</strong> was<br />

performed using the random effects method.<br />

Graph 35.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> MRI in M-staging in patients with neoplasm in varied locations<br />

Antoch 2003<br />

0,93 (0,86, 0,97)<br />

Schmidt 2005 1,00 (0,91, 1,00)<br />

Meta-<strong>analysis</strong> result [random] 0,96 (0,86, 1,00)<br />

0,85 0,90 0,95 1,00<br />

Diagnostic accuracy (95% confidence interval)<br />

Magnetic resonance diagnostic accuracy as calculated by meta-<strong>analysis</strong> <strong>of</strong> the two<br />

studies is 96% (95% CI: 86; 100).<br />

Table 112 lists the average values <strong>of</strong> diagnostic accuracy as calculated for <strong>PET</strong>-<strong>CT</strong> and<br />

MRI in M-staging.<br />

Table 112.<br />

Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI accuracy in M-staging in patients with neoplasm in varied locations.<br />

Parameter<br />

Acc (95% CI)<br />

<strong>PET</strong>-<strong>CT</strong><br />

94% (90; 98)<br />

MRI<br />

96% (86; 100)<br />

Based on the data above, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in M-staging is lower than<br />

the diagnostic accuracy <strong>of</strong> MRI.<br />

Graph 36 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the proportion <strong>of</strong> patients for whom M-staging<br />

using <strong>PET</strong>-<strong>CT</strong> vs. MRI gave results consistent with the index test.<br />

237


Graph 36.<br />

Odds ratio for the consistency <strong>of</strong> M-staging results with the reference test in patients with neoplasm in varied<br />

locations, <strong>PET</strong>-<strong>CT</strong> vs MRI<br />

238<br />

Antoch 2003 1,18 (0,33, 4,42)<br />

Schmidt 2005 0,32 (0,00, 39,00)<br />

Meta-<strong>analysis</strong> result [fixed]<br />

0,2 0,5 1 2 5 10 100<br />

Odds ratio (95%<br />

confidence interval)<br />

1,00 (0,34, 2,95)<br />

The odds ratio calculated by meta-<strong>analysis</strong> is 1.00 (95% CI: 0,34; 2,95). It means that the<br />

probability <strong>of</strong> the results <strong>of</strong> M-staging being consistent with the reference method is<br />

identical for both <strong>of</strong> the diagnostic technologies compared, and the result is not<br />

statistically significant.<br />

The parameters <strong>of</strong> the diagnostic efficacy <strong>of</strong> the methods compared (accuracy,<br />

specificity and sensitivity) presented in the trials are listed in table 113. In Antoch 2003, the<br />

authors provide efficacy data for both methods in detecting metastasis in individual organs.<br />

Table 113.<br />

Evaluation <strong>of</strong> the efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI in detecting distant metastasis in patients with neoplasm in<br />

varied locations.<br />

Parameter<br />

Se<br />

Metastasis<br />

location<br />

lungs<br />

liver<br />

bones<br />

other organs<br />

total<br />

lungs<br />

liver<br />

bones<br />

<strong>PET</strong>-<strong>CT</strong><br />

n/N (%)<br />

(89%)<br />

(86%)<br />

(62%)<br />

(73%)<br />

(93%)<br />

(94%)<br />

(96%)<br />

(96%)<br />

Antoch 2003<br />

MRI<br />

n/N (%)<br />

(82%)<br />

(93%)<br />

(85%)<br />

(67%)<br />

(90%)<br />

(94%)<br />

(95%)<br />

(92%)<br />

Statistical<br />

significance<br />

<strong>PET</strong>-<strong>CT</strong> vs MRI<br />

no data<br />

no data<br />

no data<br />

no data<br />

NS<br />

no data<br />

no data<br />

no data<br />

<strong>PET</strong>-<strong>CT</strong><br />

n/N (%)<br />

no data<br />

no data<br />

no data<br />

no data<br />

(100%)<br />

no data<br />

no data<br />

no data<br />

Schmidt 2005<br />

MRI<br />

n/N (%)<br />

no data<br />

no data<br />

no data<br />

no data<br />

(100%)<br />

no data<br />

no data<br />

no data<br />

Statistical<br />

significance<br />

<strong>PET</strong>-<strong>CT</strong> vs MRI<br />

no data<br />

no data<br />

no data<br />

no data<br />

NS<br />

no data<br />

no data<br />

no data


Sp<br />

Acc<br />

other organs<br />

total<br />

lungs<br />

liver<br />

bones<br />

other organs<br />

total<br />

(73%)<br />

(95%)<br />

(89%)<br />

(95%)<br />

(92%)<br />

(95%)<br />

92/98<br />

(94%)<br />

(67%)<br />

(95%)<br />

(82%)<br />

(95%)<br />

(91%)<br />

(93%)<br />

91/98<br />

(93%)<br />

no data<br />

NS<br />

no data<br />

no data<br />

no data<br />

no data<br />

p > 0,99<br />

no data<br />

(98%)<br />

no data<br />

no data<br />

no data<br />

no data<br />

97%<br />

no data<br />

(100%)<br />

no data<br />

no data<br />

no data<br />

no data<br />

100%<br />

no data<br />

NS<br />

no data<br />

no data<br />

no data<br />

no data<br />

p > 0,99<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> for individual organs was 93-100%, and that <strong>of</strong> magnetic<br />

resonance was 90-100%. <strong>PET</strong>-<strong>CT</strong> specificity is 95% and 98% and MRI specificity was 95%–<br />

100%.<br />

Based on Antoch 2003, <strong>PET</strong>-<strong>CT</strong> sensitivity is higher compared to MRI in detecting<br />

metastasis to lungs (89% vs. 82%), organs total (93% vs. 90%) and organs other than lungs,<br />

liver and bones (73% vs. 67%); in detecting metastasis to liver and bones, <strong>PET</strong>-<strong>CT</strong> sensitivity<br />

is lower than that <strong>of</strong> MRI, and was 86% vs. 93% for liver, and 62% vs. 85% for bones.<br />

The specificity and accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning were higher or equal to the respective<br />

parameters <strong>of</strong> MRI in detecting metastasis in all the organs scanned.<br />

Schmidt 2005 reported additional diagnostic parameters calculated based on the<br />

number <strong>of</strong> pathological lesions. Detailed results are presented in table 114.<br />

Table 114.<br />

Parameters <strong>of</strong> diagnostic efficacy in M-staging with the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI, calculated in Schmidt 2005 based on<br />

the number <strong>of</strong> pathological changes.<br />

Parameter<br />

Se<br />

n/N<br />

Sp<br />

n/N<br />

Acc<br />

Number<br />

<strong>of</strong> confirmed<br />

pathological<br />

changes<br />

268<br />

<strong>PET</strong>-<strong>CT</strong><br />

82%<br />

157/191<br />

82%<br />

63/77<br />

MRI<br />

96%<br />

184/191<br />

82%<br />

63/77<br />

With respect to the number <strong>of</strong> organs affected by metastases, <strong>PET</strong>-<strong>CT</strong> sensitivity is<br />

lower compared to MRI (82% vs. 96%), and specificity is the same for both (82%).<br />

82%<br />

92%<br />

Statistical<br />

significance<br />

(<strong>PET</strong>-<strong>CT</strong> vs MRI)<br />

no data<br />

<strong>PET</strong>-<strong>CT</strong> was more accurate compared to MRI in detecting metastasis to lungs (37 vs. 36)<br />

and to s<strong>of</strong>t tissues (6 vs. 3), and MRI was more accurate compared to <strong>PET</strong>-<strong>CT</strong> in<br />

detecting metastasis to liver (71 vs. 62) and bones (76 vs. 50).<br />

Metastases with the median size was 14 mm (SD = 13 mm, n = 40) were detected using<br />

<strong>PET</strong>-<strong>CT</strong>, and for MRI the median size <strong>of</strong> metastases was 12 mm (SD = 11, n = 61).<br />

NS<br />

no<br />

dat<br />

a<br />

239


240<br />

17.1.4.5 TNM staging system<br />

In Antoch 2003 and Schmidt 2005, the stage <strong>of</strong> disease was assessed based on the TNM<br />

staging system created by the AJCC (American Joint Committee on Cancer).<br />

Table 115 provides detailed results concerning the consistency between the<br />

diagnostic methods evaluated, and the reference test, as described by the authors <strong>of</strong><br />

each study.<br />

Table 115.<br />

Consistency between the reference test and cancer staging results (TNM feature) in patients with neoplasm in varied<br />

locations, <strong>PET</strong>-<strong>CT</strong> vs. MRI<br />

Study<br />

Antoch<br />

2003<br />

N<br />

98<br />

Correct<br />

staging<br />

(accuracy)<br />

n (%)<br />

75 (77%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Overstaged<br />

11 (11%)<br />

Schmidt<br />

2005<br />

38 36* (96%) no data<br />

*Calculated based on data available<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

12 (12%)<br />

no data<br />

Correct<br />

staging<br />

(accuracy)<br />

n (%)<br />

53 (54%)<br />

35* (91%)<br />

MRI<br />

Overstaged<br />

19 (19%)<br />

no data<br />

Incorrect staging<br />

n (%)<br />

Based on the data above, the proportion <strong>of</strong> correctly staged patients was higher<br />

for <strong>PET</strong>-<strong>CT</strong> than for magnetic resonance (MRI) in both studies. <strong>PET</strong>-<strong>CT</strong> accuracy is <strong>of</strong> 77<br />

and 96%, and the accuracy <strong>of</strong> MRI is 54% and 91%. Antoch 2005 reports that the<br />

differences in diagnostic accuracy <strong>of</strong> the testing technologies compared were<br />

statistically significant. The differences in Schmidt 2005 were not statistically significant.<br />

Understaged<br />

26 (27%)<br />

no data<br />

In Antoch 2003, the proportion <strong>of</strong> overstaged and understaged results was 11% and 12%<br />

respectively for <strong>PET</strong>-<strong>CT</strong>. and 19% and 27% respectively for MRI.<br />

Graph 37 illustrates a meta -<strong>analysis</strong> <strong>of</strong> the accuracy <strong>of</strong> clinical TNM-staging for <strong>PET</strong>-<strong>CT</strong>. As<br />

there was significant heterogeneity between trials (p = 0.0072), the meta-<strong>analysis</strong> was<br />

performed using the random effects method.


Graph 37.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in cancer staging in patients with neoplasm in varied locations<br />

Antoch 2003 0,77 (0,67, 0,85)<br />

Schmidt 2005 0,95 (0,82, 0,99)<br />

Meta-<strong>analysis</strong> result [random] 0,86 (0,65, 0,98)<br />

The diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in TNM-staging as calculated by meta-<strong>analysis</strong> <strong>of</strong> two<br />

studies is 86% (95% CI: 65; 98).<br />

0,6 0,7 0,8 0,9 1,0<br />

Diagnostic accuracy (95% confidence interval)<br />

Graph 38 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> magnetic resonance in<br />

cancer staging. As there was significant heterogeneity between trials, the meta-<strong>analysis</strong><br />

was performed using the random effects method (p < 0,0001).<br />

241


Graph 38.<br />

Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> MRI in cancer staging in patients with neoplasm in varied locations.<br />

Meta-<strong>analysis</strong> result<br />

[random]<br />

242<br />

Antoch 2003 0,54 (0,44, 0,64)<br />

Schmidt 2005 0,92 (0,79, 0,98)<br />

The diagnostic accuracy <strong>of</strong> MRI in cancer staging (TNM feature) as calculated by meta-<br />

<strong>analysis</strong> <strong>of</strong> the two studies is 75% (95% CI: 33; 99).<br />

The average values <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI in cancer staging (TNM)<br />

are presented in table 116.<br />

Table 116.<br />

Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI accuracy in TNM-staging in patients with neoplasm in varied locations.<br />

Parameter<br />

Acc (95% CI)<br />

0,0 0,2 0,4 0,6 0,8 1,0<br />

<strong>PET</strong>-<strong>CT</strong><br />

86% (65; 98)<br />

MRI<br />

75% (33; 99)<br />

Based on the data above, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in cancer staging (TNM) is<br />

higher compared to magnetic resonance (MRI).<br />

Diagnostic accuracy (95% confidence interval)<br />

0,75 (0,33, 0,99)<br />

Graph 39 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the proportion <strong>of</strong> patients for whom the results<br />

<strong>of</strong> cancer staging using <strong>PET</strong>-<strong>CT</strong> or MRI scanning were consistent with the reference test.


Graph 39.<br />

Odds ratio for the consistency <strong>of</strong> TNM-staging results with the reference test in patients with neoplasm in varied<br />

locations, <strong>PET</strong>-<strong>CT</strong> vs MRI<br />

Antoch 2003 2,77 (1,44, 5,38)<br />

Schmidt 2005 1,54 (0,17, 19,41)<br />

Meta-<strong>analysis</strong> result<br />

[fixed]<br />

0,1 0,2 0,5 1 2 5 10 100<br />

The odds ratio as calculated by meta-<strong>analysis</strong> amounts to 2,61 (95% CI: 1.46; 4.67), so the<br />

probability <strong>of</strong> cancer staging results being consistent with the reference test for <strong>PET</strong>-<strong>CT</strong> is 2.61<br />

times higher than for MRI. The result is statistically significant.<br />

In order to obtain one additional case <strong>of</strong> correct cancer staging it is necessary to use <strong>PET</strong>-<strong>CT</strong><br />

instead <strong>of</strong> MRI with 6 patients, NNT = 6 (95% CI: 4; 15).<br />

17.1.4.6 Impact on therapy<br />

Information on how the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> classical imaging methods (MRI) causes<br />

therapy tp be changed is provided only in one <strong>of</strong> the studies analyzed (Antoch 2003).<br />

The results presented by the authors <strong>of</strong> the study are given in table 117.<br />

Table 117.<br />

Influence <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> vs MRI scanning results on therapy changes in patients with neoplasm in varied locations<br />

Therapeutic decision<br />

Other therapeutic decisions; total<br />

Palliative treatment instead <strong>of</strong> operation<br />

Operation instead <strong>of</strong> palliative treatment<br />

Neoadjuvant therapy instead <strong>of</strong> operation<br />

Limited surgery instead <strong>of</strong> extensive surgery<br />

Odds ratio (95% confidence interval)<br />

<strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> MRI<br />

n/N (%)<br />

12/98 (12%)<br />

4/98 (4%)<br />

5/98 (5%)<br />

1/98 (1%)<br />

2/98 (2%)<br />

2,61 (1,46, 4,67)<br />

MRI instead <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

n/N (%)<br />

2/98 (2%)<br />

1/98 (1%)<br />

1/98 (1%)<br />

0/98 (0%)<br />

0/98 (0%)<br />

Staging the disease with the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> MRI resulted in a different<br />

243


ecommendation for further treatment in the case <strong>of</strong> 12 patients (12%), and when magnetic<br />

resonance was used instead <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> therapeutic decision in were changed for 2 patients<br />

(2%).<br />

244<br />

17.1.4.7 Safety<br />

No information on the safety <strong>of</strong> the diagnostic procedures applied is given in either <strong>of</strong> the<br />

studies under <strong>analysis</strong>.<br />

17.1.5. Results<br />

Two primary clinical trials (Antoch 2003 and Schmidt 2005) were found, where the diagnostic<br />

efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in the staging <strong>of</strong> neoplasms in varied locations was directly compared to<br />

magnetic resonance imaging – MRI (N=136). Both trials were qualified based on a reference<br />

methods, which in this part <strong>of</strong> the study were histopathological examination and/or clinical<br />

follow-up.<br />

A single-trial-based rating <strong>of</strong> the efficacy <strong>of</strong> primary and secondary neoplasm detection<br />

showed that the sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is higher in comparison to magnetic resonance imaging:<br />

100% (95% CI: 59; 100) vs. 86% (95% CI: 42; 100). The specificity <strong>of</strong> both methods was 100%. The<br />

differences between groups were not statistically significant.<br />

For T-staging, the diagnostic accuracy established by meta-<strong>analysis</strong> <strong>of</strong> two trials was 89% (95%<br />

CI: 68; 100) for <strong>PET</strong>-<strong>CT</strong> and 79% (95% CI: 26; 99) for MRI. Calculated by meta-<strong>analysis</strong>, the odds<br />

ratio for T-staging results being consistent with the reference test using <strong>PET</strong>-<strong>CT</strong> versus MRI, is 3.29<br />

(95% CI: 1.37; 7.90), and the value is statistically significant; NNT=7 (95% CI: 4; 20).<br />

Analysis showed that the <strong>PET</strong>-<strong>CT</strong> method was more accurate compared to MRI in the<br />

assessment <strong>of</strong> the involvement <strong>of</strong> lymph nodes by cancer metastasis (N status). The accuracy<br />

established by meta-<strong>analysis</strong> is 94% (95% CI: 89; 97) for <strong>PET</strong>-<strong>CT</strong>, as compared to 80% for MRI (95%<br />

CI: 73; 86). The odds <strong>of</strong> the staging results for the involvement <strong>of</strong> lymph nodes by cancer<br />

metastasis being consistent with the reference test were four times higher for the <strong>PET</strong>-<strong>CT</strong> method<br />

than for magnetic resonance. The result is statistically significant; OR = 4.00 (95% CI: 1.74; 9.19);<br />

NNT = 8 (95% CI: 5; 17). The diagnostic sensitivity reported by the authors is 95% and 100% for <strong>PET</strong>-<br />

<strong>CT</strong>, and is 79% for MRI, while specificity is 92%-95% for <strong>PET</strong>-<strong>CT</strong> and 78%-95% for MRI.<br />

Established by meta-<strong>analysis</strong> <strong>of</strong> two trials, the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-CY vs. <strong>CT</strong> in distant<br />

metastasis staging (M status) is 94% (95% CI: 90; 98) for <strong>PET</strong>-<strong>CT</strong> and 96% (95% CI: 86; 100) for<br />

magnetic resonance. The odds <strong>of</strong> obtaining reference method-consistent staging results for<br />

distant metastasis using the <strong>PET</strong>-<strong>CT</strong> method are the same as the odds for magnetic resonance:<br />

OR = 1.00 (95% CI: 0.34; 2.95). The result is statistically significant. Based on two trials included in<br />

this <strong>analysis</strong>, sensitivity is 93% and 100% for <strong>PET</strong>-<strong>CT</strong> and 90% and100% for MRI. Specificity is 95% and


98% for <strong>PET</strong>-<strong>CT</strong> and 95% and 100% for MRI.<br />

A review <strong>of</strong> the diagnostic accuracy <strong>of</strong> the methods in total staging <strong>of</strong> cancer (TNM staging)<br />

was conducted by meta-<strong>analysis</strong> <strong>of</strong> two trials, which showed that the accuracy is higher for <strong>PET</strong>-<br />

<strong>CT</strong> than for MRI. The values are 86% (95% CI: 65; 98) and 75% (95% CI: 33; 99) respectively. The<br />

odds <strong>of</strong> obtaining results consistent with reference test are 2.61 times higher for <strong>PET</strong>-<strong>CT</strong> than for<br />

MRI. The result is statistically significant; OR = 2.61 (95% CI: 1.46; 4.67); NNT = 6 (95% CI: 4; 15).<br />

The use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> MRI in the staging <strong>of</strong> cancer was conducive to changes in<br />

therapeutic recommendations in 12% <strong>of</strong> patients suffering from cancer with varied locations,<br />

while the use <strong>of</strong> MRI instead <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was conducive to changes in therapeutic<br />

recommendations in 2% <strong>of</strong> patients<br />

In patients with various cancer locations, the <strong>PET</strong>-<strong>CT</strong> method shows higher diagnostic efficacy<br />

in comparison to MRI in detecting primary and secondary neoplasm, as well as in the staging <strong>of</strong><br />

tumor (T-status), lymph nodes involvement (N-status), and distant metastases (M-status) and<br />

combined staging (TNM staging).<br />

245


246<br />

17.2. Neoplasm in varied locations: disease staging(<strong>PET</strong>-<strong>CT</strong><br />

versus <strong>CT</strong>)<br />

17.2.1. Results <strong>of</strong> trial search<br />

As a result <strong>of</strong> searching medical databases only one primary retrospective study was<br />

found (Antoch 2004;tab. 139, app. 18.1), where <strong>PET</strong>-<strong>CT</strong> scanning was compared directly<br />

with <strong>CT</strong> in staging neoplasm in varied locations.<br />

Both methods were verified based on a reference test (histologic study and/or clinical<br />

observation).<br />

17.2.2. Population characteristics<br />

Included in the trial under <strong>analysis</strong> were patients with suspected malignant tumor or<br />

with confirmed cancer.<br />

Staging was performed for 112 patients (43%), and post-treatment restaging for 148<br />

patients (57%).<br />

Table 118 provides initial characteristics <strong>of</strong> patients included in Antoch 2004.<br />

Table 118.<br />

Initial characteristics <strong>of</strong> patients with neoplasm in varied locations included in Antoch 2004.<br />

Cancer type<br />

(proportion <strong>of</strong> patients)<br />

Parameter<br />

Population number<br />

Median age<br />

[years]<br />

Male proportion<br />

lung cancer<br />

head and neck cancer<br />

esophageal cancer<br />

liver cancer<br />

thyroid cancer<br />

cancer <strong>of</strong> unknown primary origin<br />

genitourinary cancer<br />

s<strong>of</strong>t tissues or bone cancer<br />

breast cancer<br />

adrenal cancer<br />

pleural cancer<br />

Antoch 2004<br />

260<br />

56<br />

26%<br />

22%<br />

18%<br />

17%<br />

9%<br />

9%<br />

8%<br />

7%<br />

4%<br />

2%<br />

2%<br />

2%


Excluded from the trial were patients with no prior disease staging using the TNM system<br />

or clinical data such as histopathological examination <strong>of</strong> primary tumor or at least 6-<br />

months-long clinical observation.<br />

Out <strong>of</strong> 400 patients initially included in the study, 140 patients were excluded for the<br />

reasons above (120 patients for insufficient follow-up, and 20 for unavailability <strong>of</strong> TNM-<br />

staging). Ultimately, 260 patients were included in the trial. 55 were given treatment<br />

during or after <strong>PET</strong>-<strong>CT</strong> scanning. The authors noted that 123 untreated patients included in<br />

the trial had had typical indications (as defined by the National Coverage Analysis, USA) 16<br />

for <strong>PET</strong> scanning, refunded by the Centers <strong>of</strong> Medicare and Medicaid Services, USA.<br />

The median age <strong>of</strong> the participants in the trial was 56, and the male proportion was 26%.<br />

17.2.3. Description <strong>of</strong> intervention<br />

17.2.3.1 <strong>PET</strong>-<strong>CT</strong><br />

In Antoch 2004, the Biograph system produced by Siemens Medical Solutions<br />

(H<strong>of</strong>fman Estates, USA) was used for disease staging. The system consisted <strong>of</strong> a single-<br />

slice spiral <strong>CT</strong> and a full-ring <strong>PET</strong>.<br />

Whole body scans (from head to upper thigh) were taken using <strong>PET</strong>-<strong>CT</strong> for 212 patients<br />

(82%), and for the remaining 48 patients (18%) smaller areas were scanned. FDG (fluoro-<br />

deoxy-glucose) with the activity <strong>of</strong> 350 MBq was given as a radiopharmaceutical 60<br />

minutes prior to <strong>PET</strong>-<strong>CT</strong> tests. Glucose concentration in blood was checked before<br />

administering FDG.<br />

Table 119 contains a detailed description <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong> tests.<br />

Table 119.<br />

Description <strong>of</strong> the intervention in Antoch 2004.<br />

<strong>PET</strong>-<strong>CT</strong> scanner<br />

type<br />

Biograph<br />

<strong>PET</strong>-<strong>CT</strong> scanner<br />

manufacturer<br />

Siemens Medical<br />

Solutions (H<strong>of</strong>fman<br />

Estates, USA)<br />

Radiopharmaceutical<br />

type and<br />

administration method<br />

FDG intravenous<br />

Study range<br />

whole body (82%)<br />

smaller area<br />

(12%)<br />

Radiomarker<br />

activity<br />

[MBq]<br />

In Antoch 2004, FDG was given intravenously to all patients without contraindication<br />

for the purposes <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning.<br />

Two radiologists and two nuclear medicine consultants assessed the <strong>PET</strong>-<strong>CT</strong> images.<br />

<strong>PET</strong>-<strong>CT</strong> as well as <strong>CT</strong> scans were carried out using the same <strong>PET</strong>-<strong>CT</strong> machine, however <strong>PET</strong>-<strong>CT</strong><br />

16 National Coverage Determinations: Positron Emission Tomography (<strong>PET</strong>) Scans, Centers for<br />

Medicare and Medicaid Services. http://cms.hhs.gov/mcd/viewdecisionmemo.asp?id=85<br />

350<br />

247


scans were analyzed 12 weeks after the assessment <strong>of</strong> <strong>CT</strong> images. Identical criteria were used<br />

for the assessment <strong>of</strong> cancer malignancy using <strong>CT</strong> and <strong>PET</strong>-<strong>CT</strong>, and all assessors had the<br />

same information available for all patients.<br />

248<br />

17.2.3.2 Diagnostic trial compared<br />

Antoch 2004 compared <strong>PET</strong>-<strong>CT</strong> with <strong>CT</strong> using a single slice-spiral <strong>CT</strong> that was an<br />

integral part <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> system.<br />

Table 120 provides a detailed description <strong>of</strong> the <strong>CT</strong> tests performed in Antoch 2004.<br />

Table 120.<br />

Description <strong>of</strong> the diagnostic <strong>CT</strong> tests performed in Antoch 2004<br />

<strong>CT</strong> scanner type<br />

no data<br />

<strong>CT</strong> scanner manufacturer<br />

Siemens Medical<br />

Solutions<br />

Contrast medium type<br />

and administration<br />

method<br />

intravenous contrast<br />

medium<br />

Scope <strong>of</strong> test<br />

whole body (82%)<br />

smaller area (18%)<br />

In Antoch 2004, whole body scans that corresponded to the coverage <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong><br />

tests (from head to upper thigh) were performed for 82% <strong>of</strong> patients, and for 18% smaller<br />

areas were scanned.<br />

17.2.3.3 Reference test<br />

In Antoch 2004, histopathological examination and/or clinical observation were the<br />

reference test. In line with the AJCC classification (American Joint Committee on Cancer),<br />

cancer staging was done by a medical board consisting <strong>of</strong> a nuclear medicine consultant<br />

and a radiologist. All clinical and radiological data for all the patients were provided to the<br />

medical board. The medical board members had not participated in disease staging based<br />

on the imaging techniques compared. Cancer staging was based on histopathological tests<br />

and data obtained through observation, including clinical examinations, laboratory tests,<br />

radiological procedures and histopathological tests that followed a surgery carried out later.<br />

For 77 patients (30%), their tumors were resected and T-staged so for these patients<br />

only the diagnostic efficacy <strong>of</strong> the procedures compared was evaluated. The involvement<br />

<strong>of</strong> lymph nodes (N feature) and the presence <strong>of</strong> distant metastases (M feature) were<br />

identified following a surgery for 71 (28%) and 57 (22%) patients respectively. However, the<br />

imaging methods were compared with respect to these two features for 260 trial<br />

participants. The average time observation, which was the reference method for N- and M-<br />

staging, was 311 days (SD = 125 days). This relatively short observation time was caused by<br />

the patient's death due to cancer.


17.2.4. Findings<br />

17.2.4.1 T-staging<br />

T-staging was done based on the TNM staging system designed by the AJCC<br />

(American Joint Committee on Cancer). <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> method results were verified<br />

histopathologicaly. The total <strong>of</strong> 77 patients were analyzed following tumor resections.<br />

The consistency between the T-staging done using the diagnostic methods compared<br />

and the reference test as described in Antoch 2004 is presented in table 121.<br />

Table 121.<br />

Consistency <strong>of</strong> T-staging with the reference test in patients with neoplasm in varied locations in Antoch 2004, <strong>PET</strong>-<strong>CT</strong><br />

vs <strong>CT</strong><br />

N*<br />

Correct staging<br />

(accuracy)<br />

n (%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Overstaged<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

77 63 (82%) 6 (8%**) 8 (10%**)<br />

*Number <strong>of</strong> patients verified using the reference test<br />

**Calculated based on data available<br />

Correct staging<br />

(accuracy)<br />

n (%)<br />

51 (66%)<br />

<strong>CT</strong><br />

Overstaged<br />

11 (14%**)<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

15 (19.5%**)<br />

Based on the results in Antoch 2004, the proportion <strong>of</strong> correctly staged patients was<br />

higher for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong> and the result was statistically significant (p = 0.0018). <strong>PET</strong>-<strong>CT</strong><br />

accuracy is 82% (95% CI: 71; 90), and <strong>CT</strong> accuracy is 66% (95% CI: 55; 77). Overstaged and<br />

understaged results represented 8% and 10% <strong>of</strong> total results respectively for <strong>PET</strong>-<strong>CT</strong>, 14%<br />

and 19,5% respectively for <strong>CT</strong>,.<br />

Calculated based on the results above, the odds ratio for the consistency <strong>of</strong> T-staging<br />

with the reference test for patients with neoplasm in varied locations is 2.29 (95% CI: 1.03;<br />

5.25). It means that the probability <strong>of</strong> consistency between T-staging and the reference<br />

test is 2.29 times higher for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>. The result is statistically significant.<br />

In order to obtain one additional case <strong>of</strong> correct T-staging it is necessary to use <strong>PET</strong>-<strong>CT</strong><br />

instead <strong>of</strong> <strong>CT</strong> with 7 patients, NNT = 7 (95% CI: 4; 59).<br />

17.2.4.2 N-staging<br />

In Antoch 2004, N-staging was based on the TNM lung cancer staging system designed<br />

by AJCC (American Joint Committee on Cancer).<br />

Antoch 2004 evaluated the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in comparison with <strong>CT</strong> in 260<br />

patients; in 72 <strong>of</strong> them the results were verified histopathologically, and for the remaining<br />

participants, clinical observation was reference test.<br />

The consistency between the N-staging results <strong>of</strong> the diagnostic methods compared<br />

and the reference test in Antoch 2004 is presented in table 122.<br />

249


Table 122.<br />

Consistency between the reference test and N-staging results in patients with carcinomas in varied locations, <strong>PET</strong>-<strong>CT</strong><br />

vs <strong>CT</strong><br />

N*<br />

250<br />

Correct staging<br />

(accuracy)<br />

n (%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Overstaged<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

260 240 (92%) 12 (5%**) 8 (3%**)<br />

*Number <strong>of</strong> patients verified using the reference test<br />

**Calculated based on data available<br />

Correct staging<br />

(accuracy)<br />

n (%)<br />

197 (76%)<br />

<strong>CT</strong><br />

Overstaged<br />

27 (10%**)<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

36 (14%**)<br />

Based on the data above, the proportion <strong>of</strong> correctly staged patients was higher for<br />

<strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>. The diagnostic accuracy for <strong>PET</strong>-<strong>CT</strong> is 92% (95% CI: 88; 95), and the<br />

diagnostic accuracy for <strong>CT</strong> is 76% (95% CI: 70; 81). Overstaged and understaged results<br />

represented 5% and 3% <strong>of</strong> results total respectively for <strong>PET</strong>-<strong>CT</strong>, and 10% and 14%<br />

respectively for <strong>CT</strong>.<br />

The authors <strong>of</strong> Antoch 2004 noted that the differences in accuracy between the<br />

diagnostic methods compared was statistically significant (p < 0,0001).<br />

The odds ratio calculated based on the results in Antoch 2004 is 3.84 (95% CI: 2.19;<br />

6.93) so the probability <strong>of</strong> obtaining consistency between the reference test and N-<br />

staging using <strong>PET</strong>-<strong>CT</strong> is almost four times higher than the odds ratio for <strong>CT</strong>.<br />

In order to obtain one additional N-staging consistent with the reference test it is necessary<br />

to use <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> <strong>CT</strong> with 7 patients with neoplasm in varied locations; NNT = 7 (95% CI:<br />

5; 10).<br />

17.2.4.3 Assessment <strong>of</strong> distant metastasis<br />

Antoch 2004 cancer staging was done based on the TNM system with respect to distant<br />

metastasis.<br />

The authors <strong>of</strong> the trial evaluated the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> as compared to <strong>CT</strong> in<br />

260 patients, and histopathological tests were done for 57 patients, while for the remaining<br />

patients clinical observation was used as reference test.<br />

The consistency between the reference test and the M-staging results obtained using the<br />

diagnostic methods compared as described by the authors <strong>of</strong> the trial are given in table<br />

123.


Table 123.<br />

Consistency between the reference test and M-staging in patients with neoplasm in varied locations, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong><br />

N*<br />

Correct staging<br />

(accuracy)<br />

n (%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Overstaged<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

260 248 (95%) 4 (1,5%**) 8 (3%**)<br />

*Number <strong>of</strong> patients verified using the reference test<br />

**Calculated based on data available<br />

Correct staging<br />

(accuracy)<br />

n (%)<br />

230 (88%)<br />

<strong>CT</strong><br />

Overstaged<br />

7 (3%**)<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

23 (9%**)<br />

In Antoch 2004, a higher diagnostic accuracy was observed for <strong>PET</strong>-<strong>CT</strong> study as<br />

compared to <strong>CT</strong>, and the differences were statistically significant (p = 0.0001). Based on<br />

the data above, <strong>PET</strong>-<strong>CT</strong> accuracy is 95% (95% CI: 92; 98), and <strong>CT</strong> accuracy is 88% (95%<br />

CI: 84; 92). The proportions <strong>of</strong> overstaged and understaged results represented 1.5% and<br />

3% <strong>of</strong> results total respectively for <strong>PET</strong>-<strong>CT</strong>, and 3% and 9% respectively for <strong>CT</strong>.<br />

The odds ratio calculated based on the results <strong>of</strong> a single trial is 2.70 (95% CI: 1,30;<br />

5,92), which means that the probability <strong>of</strong> obtaining M-staging results consistent with<br />

the reference tests is 2.7 times higher for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>, and the result is statistically<br />

significant.<br />

In order to obtain one additional case <strong>of</strong> correct M-staging it is necessary to use <strong>PET</strong>-<strong>CT</strong><br />

scanning instead <strong>of</strong> <strong>CT</strong> with 15 patients, NNT = 15 (95% CI: 7; 44).<br />

The parameters <strong>of</strong> the diagnostic efficacy <strong>of</strong> the methods compared (accuracy,<br />

specificity and sensitivity) presented in the trial are listed in table 124. In Antoch 2004, the<br />

authors additionally provided data on the efficacy <strong>of</strong> both methods in detecting metastasis in<br />

individual organs.<br />

Table 124.<br />

Evaluation <strong>of</strong> the efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in detecting metastasis in patients with neoplasm in varied locations<br />

Parameter<br />

Se (95% CI)<br />

Sp (95% CI)<br />

Acc (95% CI)<br />

Location <strong>of</strong> metastases<br />

lungs<br />

liver<br />

bones<br />

total<br />

lungs<br />

liver<br />

bones<br />

total<br />

lungs<br />

liver<br />

<strong>PET</strong>-<strong>CT</strong><br />

92% (83; 97)<br />

95% (86; 99)<br />

81% (62; 94)<br />

94% (88; 97)<br />

93% (88; 96)<br />

97% (93; 99)<br />

99% (97; 100)<br />

97% (92; 99)<br />

93% (no data)<br />

97% (no data)<br />

<strong>CT</strong><br />

83% (72; 91)<br />

86% (75; 94)<br />

48% (29; 68)<br />

82% (74; 88)<br />

91% (85; 95)<br />

95% (90; 98)<br />

98% (95; 99)<br />

95% (89; 98)<br />

89% (no data)<br />

93% (no data)<br />

251


252<br />

bones<br />

total<br />

97% (no data)<br />

95% (92; 98)<br />

93% (no data)<br />

88% (84; 92)<br />

The sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> for individual organs was 94%, and that <strong>of</strong> <strong>CT</strong> was 82%. <strong>PET</strong>-<strong>CT</strong><br />

specificity is within the range <strong>of</strong> 97%, and MRI specificity was 95%.<br />

Based on Antoch 2004, the diagnostic specificity, sensitivity and accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

scanning were higher compared to <strong>CT</strong> in detecting metastasis to lungs, liver and bones, as<br />

well as to all organs total.<br />

Antoch 2004 presented additional diagnostic parameters calculated for a population<br />

<strong>of</strong> untreated patients (n = 123) with typical indications for <strong>PET</strong> scanning refunded by the<br />

Centers <strong>of</strong> Medicare and Medicaid Services, USA.<br />

Table 125.<br />

Parameters <strong>of</strong> the diagnostic efficacy <strong>of</strong> M-staging using <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in patients with typical indications for <strong>PET</strong> in<br />

Antoch 2004.<br />

Parameter<br />

Se (95% CI)<br />

Sp (95% CI)<br />

Acc (95% CI)<br />

<strong>PET</strong>-<strong>CT</strong><br />

97% (90; 100)<br />

96% (87; 100)<br />

97% (no data)<br />

<strong>CT</strong><br />

84% (73; 92)<br />

95% (85; 99)<br />

89% (no data)<br />

Statistical<br />

significance<br />

(<strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong>)<br />

no data<br />

no data<br />

no data<br />

With respect to patients with typical indications for <strong>PET</strong>, the sensitivity, specificity and<br />

diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> were higher compared to <strong>CT</strong> and stood at 97% vs. 84%,<br />

96% vs. 95%, 97% vs. 89% respectively. The trial provides no information on the statistical<br />

significance <strong>of</strong> the differences between the methods compared.<br />

17.2.4.4 TNM staging system<br />

In Antoch 2004, disease staging was done based on the TNM system designed by the<br />

AJCC (American Joint Committee on Cancer).<br />

Detailed data on the consistency between the reference (index) test and the diagnostic<br />

technologies under assessment as described by the authors are given in table 126.


Table 126.<br />

Consistency between the reference test and cancer staging in patients with neoplasm in varied locations, <strong>PET</strong>-<strong>CT</strong> vs.<br />

<strong>CT</strong><br />

N<br />

Correct staging<br />

(accuracy)<br />

n (%)<br />

<strong>PET</strong>-<strong>CT</strong><br />

Overstaged<br />

260 218 (84%) 18 (7%)<br />

*Calculated based on data available<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

24 (9%)<br />

Correct staging<br />

(accuracy)<br />

n (%)<br />

163 (63%)<br />

<strong>CT</strong><br />

Overstaged<br />

36 (14%)<br />

Incorrect staging<br />

n (%)<br />

Understaged<br />

61 (23,5%)<br />

Based on the data above, the proportion <strong>of</strong> correctly staged patients was higher for<br />

<strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>. The accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was 84% (95% CI: 79; 88), and the accuracy<br />

<strong>of</strong> <strong>CT</strong> was 63% (95% CI: 57; 69). Antoch 2004 reports that the differences in diagnostic<br />

accuracy between the scanning technologies were statistically significant (p < 0.0001).<br />

In Antoch 2004, the proportions <strong>of</strong> overstaged and understaged results were 7% and 9%<br />

respectively for <strong>PET</strong>-<strong>CT</strong>, and 14% and 23.5% respectively for <strong>CT</strong>.<br />

As calculated based on the data above, the odds ratio is 3.09 (95% CI: 2.00; 4.80),<br />

so the probability <strong>of</strong> cancer staging consistent with the reference test is more than<br />

three times higher for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>. The difference is statistically significant.<br />

In order to obtain one additional case <strong>of</strong> correct cancer staging it is necessary to use <strong>PET</strong>-<br />

<strong>CT</strong> instead <strong>of</strong> <strong>CT</strong> with 5 patients, NNT = 5 (95% CI: 4; 8).<br />

17.2.4.5 Impact on therapy<br />

Antoch 2004 reports on how the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning instead <strong>of</strong> <strong>CT</strong> caused the<br />

therapeutic procedure to be changed.<br />

Clinically significant impact on treatment was defined in the trial as a change <strong>of</strong><br />

recommendation from surgery to palliative treatment, from neoadjuvant to surgery, and<br />

from palliative to neoadjuvant treatment.<br />

The results reported in Antoch 2004 are given in table 127.<br />

Table 127.<br />

Influence <strong>of</strong> the diagnostic methods compared on therapy in patients with neoplasm in varied locations, <strong>PET</strong>-<strong>CT</strong> vs.<br />

<strong>CT</strong><br />

Therapeutic decision<br />

Other therapeutic decisions; total<br />

*Calculated based on data available<br />

<strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> <strong>CT</strong><br />

n/N (%)<br />

39/260 (15%*)<br />

<strong>CT</strong> instead <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

n/N (%)<br />

2/260 (0,8%*)<br />

Staging the disease using <strong>PET</strong>-<strong>CT</strong> scanning instead <strong>of</strong> <strong>CT</strong> led to different<br />

recommendations for further treatment in the case <strong>of</strong> 39 patients (15%), and the use <strong>of</strong> <strong>CT</strong><br />

instead <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> resulted in changing the therapy for 2 patients (0.8%).<br />

253


254<br />

17.2.4.6 Safety<br />

No information on the safety <strong>of</strong> the diagnostic procedures used was found in the trial<br />

under <strong>analysis</strong>.<br />

17.2.5. Results<br />

One primary, retrospective clinical trial (Antoch 2004) was identified, where the diagnostic<br />

efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in the staging <strong>of</strong> neoplasm with varied locations was compared directly with<br />

<strong>CT</strong> (N=260). Both testing methods were verified against the reference methods, i.e.<br />

histopathological examination and/or clinical follow-up.<br />

For T-staging, the diagnostic accuracy was 82% (95% CI: 71; 90) for <strong>PET</strong>-<strong>CT</strong>, and 66% (95% CI:<br />

55; 77) for <strong>CT</strong>. The differences between the groups under discussion are statistically significant (p<br />

= 0.0018). The odds <strong>of</strong> obtaining a T-status consistent with the reference method is 2.29 times<br />

higher for <strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong>. The result is statistically significant; OR = 2.29 (95% CI: 1.03; 5.25), NNT =<br />

7 (95% CI: 4; 59).<br />

Analysis showed that the <strong>PET</strong>-<strong>CT</strong> method is statistically significantly more accurate than <strong>CT</strong> in<br />

the staging <strong>of</strong> the involvement <strong>of</strong> lymph nodes (N status). The diagnostic accuracy is 92% (95%<br />

CI: 88; 95) for <strong>PET</strong>-<strong>CT</strong> and 76% (95% CI: 70; 81) for <strong>CT</strong>. The odds <strong>of</strong> obtaining lymph nodes<br />

involvement staging results consistent with the reference test is nearly four times higher for the<br />

<strong>PET</strong>-<strong>CT</strong> procedure versus <strong>CT</strong>. The result is statistically significant; OR = 3.84 (95% CI: 2.19; 6.93), NNT<br />

= 7 (95% CI: 5; 10).<br />

The diagnostic accuracy <strong>of</strong> the methods compared in the staging <strong>of</strong> distant metastases (M<br />

status) is 95% (95% CI: 92; 98) for <strong>PET</strong>-<strong>CT</strong> and 88% (95% CI: 84; 92) for <strong>CT</strong>, and the differences<br />

between the methods are statistically significant (p=0.0001). The odds <strong>of</strong> M-staging results being<br />

consistent with the reference method is 2.7 times higher for <strong>PET</strong>-<strong>CT</strong> versus <strong>CT</strong>. The result is<br />

statistically significant; OR = 2.70 (95% CI: 1.30; 5.92), NNT = 15 (95% CI: 7; 44).<br />

Sensitivity, specificity and accuracy are higher for <strong>PET</strong>-<strong>CT</strong> compared to <strong>CT</strong>, including in staging<br />

metastases to lungs, liver and bones, as well as organs total.<br />

Analysis showed that the diagnostic accuracy <strong>of</strong> the methods under assessment in cancer<br />

staging (TNM staging) is higher when the <strong>PET</strong>-<strong>CT</strong> method is used compared to <strong>CT</strong>. The accuracy<br />

<strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is 84% (95% CI: 79; 88), and for <strong>CT</strong> it is 63% (95% CI: 57; 69). The odds <strong>of</strong> obtaining staging<br />

results consistent with the reference standard is more than three times higher for <strong>PET</strong>-<strong>CT</strong><br />

compared to <strong>CT</strong>. The result is statistically significant, OR = 3,09 (95% CI 2.00; 4.80), NNT = 5 (95% CI:<br />

4; 8).<br />

Using <strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> <strong>CT</strong> in disease staging led to a different recommendation for further<br />

treatment in 15% patients. Using <strong>CT</strong> instead <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> caused therapeutic decisions to be


changed for 0.8% patients<br />

17.3. Cancers <strong>of</strong> unknown primary origin: diagnostic efficacy<br />

17.3.1. Results <strong>of</strong> trial search<br />

Two primary clinical trials were found: Freudenberg 2005 (tab. 142, app. 18.1), Gutzeit<br />

2005 (tab. 143, app. 18.1) that compared directly the diagnostic efficacy <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong><br />

method with <strong>CT</strong> in locating focuses <strong>of</strong> primary carcinoma in metastatic patients.<br />

The scanning methods compared were verified based on reference tests:<br />

histopathological examination and/or clinical observation in Freudenberg 2005, and<br />

histopathological examination in Gutzeit 2005.<br />

As the trials were executed at the same health center (University Hospital Essen), and the<br />

patients recruitment periods coincided, the fact that the population <strong>of</strong> Freudenberg 2005<br />

represented a subgroup <strong>of</strong> Gutzeit 2005 population was taken into account. However, in view<br />

<strong>of</strong> differences in cancer scanning methodology as reported in the trials, and differences in<br />

final trial results, the populations in the two trials included in this <strong>analysis</strong> are deemed<br />

disparate.<br />

17.3.2. Population characteristics<br />

Patients with metastasis <strong>of</strong> unknown origin participated in the two trials.<br />

In Freudenberg 2005, patients with metastasis to neck lymph nodes participated, while in<br />

Gutzeit 2005, and patients with metastasis both to neck and outside neck participated.<br />

Patients with metastasis confirmed histologically or cytologically as well as patients<br />

with the disease staged clinically, endoscopically, sonographically or radiologically<br />

were included in Freudenberg 2005. <strong>CT</strong> <strong>of</strong> the neck or head had not been done for any<br />

<strong>of</strong> the patients.<br />

In Gutzeit 2005 study, metastases to neck lymph nodes were detected in 40% <strong>of</strong><br />

patients and metastasis to other body parts in 60% <strong>of</strong> patients. A histological test <strong>of</strong> at<br />

least one metastasis spot was done for each patient. All patients were interviewed, and<br />

results <strong>of</strong> detailed medical examinations, laboratory tests, imaging tests and endoscopic<br />

procedures were collected.<br />

Neither <strong>of</strong> the trials provided detailed patient inclusion or exclusion criteria.<br />

Table 128 contains initial characteristics <strong>of</strong> patients included in each trial.<br />

255


Table 128.<br />

Initial characteristics <strong>of</strong> patients included in trials.<br />

256<br />

Histology<br />

(patient proportion)<br />

Parameter<br />

Size <strong>of</strong> population<br />

Mean age [years]<br />

Male proportion<br />

adenocarcinoma<br />

squamous cell<br />

carcinoma<br />

Freudenberg<br />

2005<br />

21<br />

64<br />

76%<br />

19%<br />

67%<br />

anaplastic carcinoma<br />

14%<br />

* average weighted by the number <strong>of</strong> patients participating in the two trials<br />

Gutzeit<br />

2005<br />

21 and 45 patients were included in Freudenberg 2005 and Gutzeit 2005 trials<br />

respectively. The total <strong>of</strong> 66 patients were included in the <strong>analysis</strong> with the mean age <strong>of</strong> 59-<br />

64 and the male proportion <strong>of</strong> 58–76%. In Freudenberg 2005, squamous cell carcinoma was<br />

identified in most patients, while in Gutzeit 2005, metastases had the form <strong>of</strong><br />

adenocarcinoma in most patients.<br />

17.3.3. Description <strong>of</strong> intervention<br />

17.3.3.1 <strong>PET</strong>-<strong>CT</strong><br />

The Biograph system produced by Siemens Medical Solutions (H<strong>of</strong>fman Estates, USA)<br />

was used for disease staging in both trials included in <strong>analysis</strong>. The system consisted <strong>of</strong> a<br />

dual-slice spiral computed tomograph (<strong>CT</strong>) and full-ring <strong>PET</strong> tomograph with BGO<br />

detectors.<br />

FDG (fluoro-deoxy-glucose) was used as radiopharmaceutical in both trials, its activity<br />

ranging from 350 MBq in Gutzeit 2005, to 360 MBq in Freudenberg 2005. The<br />

radiopharmaceutical was administered 60 minutes prior to <strong>PET</strong>-<strong>CT</strong> scanning in both studies.<br />

Patients could not eat for at least 4 hours before the radiopharmaceutical was given in<br />

Gutzeit 2005, and for 10 hours in Freudenberg 2005. In both studies, the level <strong>of</strong> glucose<br />

concentration was tested.<br />

In Gutzeit 2005, whole body scans were done, and in Freudenberg 2005, scans <strong>of</strong> head<br />

and neck were taken, followed after 70-75 minutes by additional whole body scans (from<br />

thorax to pelvis).<br />

Table 129 contains a detailed description <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong> tests.<br />

45<br />

59<br />

58%<br />

56%<br />

33%<br />

11%<br />

Total<br />

66<br />

61*<br />

64%<br />

44%<br />

44%<br />

12%


Table 129.<br />

Description <strong>of</strong> intervention<br />

Study<br />

Freudenberg<br />

2005<br />

Gutzeit<br />

2005<br />

<strong>PET</strong>-<strong>CT</strong><br />

scanner<br />

type<br />

Biograph<br />

Biograph<br />

<strong>PET</strong>-<strong>CT</strong> scanner<br />

manufacturer<br />

Siemens Medical<br />

Solutions (H<strong>of</strong>fman<br />

Estates, USA)<br />

Siemens Medical<br />

Solutions (H<strong>of</strong>fman<br />

Estates, USA)<br />

Radiopharmaceu<br />

tical type and<br />

administration<br />

method<br />

FDG<br />

intravenous<br />

FDG<br />

intravenous<br />

Study range<br />

head and<br />

neck; whole<br />

body<br />

whole body<br />

Radiomarker<br />

activity [MBq]<br />

<strong>PET</strong>-<strong>CT</strong> images included in the <strong>analysis</strong> were assessed by two nuclear medicine<br />

consultants and two radiologists blinded to the results <strong>of</strong> other imaging studies.<br />

I Gutzeit 2005 noted that the medical assessors had 2 years’ experience with <strong>PET</strong>-<strong>CT</strong>. In<br />

order to avoid mistakes in locating primary tumors, the time interval between scan reading<br />

sessions for particular imaging methods was 4 weeks, and the images were analyzed in a<br />

random order.<br />

17.3.3.2 Diagnostic test compared<br />

In the two trials under <strong>analysis</strong> (Freudenberg 2005, Gutzeit 2005), <strong>PET</strong>-<strong>CT</strong> scanning was<br />

compared with <strong>CT</strong>.<br />

In both clinical trials, <strong>CT</strong> imaging was done as part <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanning, and the<br />

computed tomograph was an integral component <strong>of</strong> the hybrid scanner. For both studies<br />

the spiral computed tomograph Somatom Emotion produced by Siemens Medical<br />

Solutions was used.<br />

Table 130 contains a detailed description <strong>of</strong> Ct tests in each study.<br />

Table 130.<br />

Description <strong>of</strong> the diagnostic tests compared<br />

Study<br />

Freudenberg<br />

2005<br />

<strong>CT</strong> scanner type<br />

Somatom Emotion<br />

<strong>CT</strong> scanner<br />

manufacturer<br />

Siemens Medical<br />

Solutions<br />

Contrast medium type<br />

and administration<br />

method<br />

Xenetix 300<br />

contrasts medium,<br />

intravenous<br />

360<br />

350<br />

Study range<br />

head and neck<br />

whole body<br />

257


258<br />

Gutzeit 2005<br />

Somatom Emotion<br />

Siemens Medical<br />

Solutions<br />

Xenetix 300<br />

contrasts medium,<br />

intravenous<br />

whole body<br />

In order to perform <strong>CT</strong> as part <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> testing, iodinated Xenetix 300 (Guerbet GmbH,<br />

Sulzbach, Germany) was given intravenously as contrast medium with 300 mg/ml <strong>of</strong> iodine<br />

in both trials. 140 ml was given in Gutzeit 2005 while in Freudenberg 2005, 60 ml was given<br />

for head and neck scanning, and 90 ml for whole body scanning.<br />

Intestine outline was achieved by administering 1000 ml <strong>of</strong> barium sulfate or 1500 ml <strong>of</strong><br />

0.2% solution <strong>of</strong> locust bean gum, and 2.5% solution <strong>of</strong> mannitol.<br />

A blind <strong>analysis</strong> <strong>of</strong> <strong>CT</strong> images was done by two radiologists.<br />

17.3.3.3 Reference test<br />

The reference method in Freudenberg 2005 was histopathological examination and<br />

clinical follow-up, while in Gutzeit 2005, results were verified histologically.<br />

In Freudenberg 2005, histopathological examination was performed for 67% patients, and<br />

results verification based on clinical follow-up (at least 9 months) was performed for 33% <strong>of</strong> the<br />

patients included in <strong>analysis</strong>. During the follow-up, panendoscopy with biopsy <strong>of</strong> the most<br />

probable locations <strong>of</strong> primary tumor and USG was performed for 33% patients; <strong>CT</strong> and/or MRI<br />

was performed for 29% patients; and diagnostic removal <strong>of</strong> tonsils (tonsillectomy) with<br />

additional biopsy were performed for 19% patients.<br />

Gutzeit 2005 provides no detailed description <strong>of</strong> the reference test.<br />

17.3.4. Findings<br />

17.3.4.1 Diagnostic accuracy in detecting primary cancer <strong>of</strong> unknown origin<br />

In both studies included in this <strong>analysis</strong> (Freudenberg 2005, Gutzeit 2005), the efficacy <strong>of</strong><br />

detecting carcinomas <strong>of</strong> unknown location using <strong>PET</strong>-<strong>CT</strong> scans compared to <strong>CT</strong> for metastasis<br />

patients was evaluated.<br />

In Freudenberg 2005, the location <strong>of</strong> primary carcinoma was histologically confirmed in<br />

14 out <strong>of</strong> 21 patients with metastasis to neck. Out <strong>of</strong> that number, 4 patients were<br />

diagnosed with oral cavity cancer, 3 with bronchial cancer, 2 with laryngeal cancer and<br />

non-Hodgkin lymphoma, and single patients were diagnosed with esophageal, thyroid and<br />

salivary gland cancers. In 7 patients no primary tumor was detected using any <strong>of</strong> the<br />

diagnostic methods or during the follow-up period since they took combined radio-<br />

chemotherapy.


In Gutzeit 2005, the total <strong>of</strong> 17 primary tumors were diagnosed: 8 lung cancer cases, 3<br />

breast cancers, 2 gastric cancers, 2 mandible adenocarcinomas, one tongue base<br />

cancer and 1 esophageal cancer.<br />

The number <strong>of</strong> patients with true positive (TP), false positive (FP), false negative (FN)<br />

results <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> is given in table 131. Patients without primary tumor located using<br />

either <strong>PET</strong>-<strong>CT</strong> or the reference method are included under false negative.<br />

Table 131.<br />

Number <strong>of</strong> metastasis patients with TP, FP, FN and TN results in the diagnostics <strong>of</strong> primary tumor using <strong>PET</strong>-<strong>CT</strong><br />

and <strong>CT</strong><br />

Study<br />

Freudenberg 2005<br />

Gutzeit 2005<br />

Population<br />

neck area<br />

metastasis<br />

neck area<br />

metastasis<br />

metastasis<br />

outside neck<br />

total metastasis<br />

*Calculated based on data available<br />

TP<br />

12<br />

6<br />

9<br />

15<br />

<strong>PET</strong>-<strong>CT</strong><br />

Gutzeit 2005 noted that the reason for three false positive results for <strong>PET</strong>-<strong>CT</strong> scans were<br />

colitis, inflammation <strong>of</strong> esophagus and lung infarction.<br />

<strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> sensitivity eating for each study are given in table 132.<br />

Table 132.<br />

<strong>PET</strong>-<strong>CT</strong> sensitivity vs. <strong>CT</strong> sensitivity in detecting primary carcinomas <strong>of</strong> unknown locations<br />

Study<br />

Freudenberg<br />

2005<br />

Gutzeit 2005<br />

Population<br />

neck area<br />

metastasis<br />

neck area<br />

metastasis<br />

metastasis<br />

outside neck<br />

total<br />

*Calculated based on data available<br />

<strong>PET</strong>-<strong>CT</strong><br />

Se (95% CI)<br />

n/N<br />

57% (34; 78)*<br />

12/21<br />

35% (14; 62)*<br />

6/17<br />

36% (18; 57)*<br />

9/25<br />

36% (22; 52)*<br />

15/42<br />

FP<br />

0<br />

1<br />

2<br />

3<br />

FN<br />

9*<br />

11<br />

16<br />

27<br />

<strong>CT</strong><br />

TP<br />

5<br />

4<br />

4<br />

8<br />

Se (95% CI)<br />

n/N<br />

28% (10; 53)*<br />

5/18<br />

25% (7; 52)*<br />

4/16<br />

15% (4; 35)*<br />

4/26<br />

19% (9; 34)*<br />

8/42<br />

The authors <strong>of</strong> Freudenberg 2005 found that the sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is significantly different<br />

<strong>CT</strong><br />

FP<br />

3<br />

2<br />

1<br />

3<br />

FN<br />

13*<br />

12<br />

22<br />

34<br />

Statistical<br />

significance<br />

(<strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong>)<br />

p = 0,03<br />

than that on <strong>CT</strong> (p=0.03). In Gutzeit 2005, <strong>PET</strong>-<strong>CT</strong> sensitivity was higher compared to <strong>CT</strong>, but the<br />

NS<br />

NS<br />

NS<br />

259


260<br />

differences were not statistically significant.<br />

Graph 40 illustrates <strong>PET</strong> /<strong>CT</strong> diagnostic accuracy for patients with distant metastasis to the<br />

neck area as calculated by meta-<strong>analysis</strong> <strong>of</strong> the two trials discussed.<br />

Graph 40.<br />

Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostic sensitivity in patients with metastasis to neck area<br />

<strong>PET</strong>-<strong>CT</strong> study sensitivity as calculated by meta-<strong>analysis</strong> <strong>of</strong> two trials for patients with<br />

metastasis to the neck area is 47% (95% CI: 31; 64), which means that if hybrid <strong>PET</strong>-<strong>CT</strong> is used<br />

the probability <strong>of</strong> primary carcinoma detection in patients with metastasis to the neck area<br />

is 47%.<br />

Freudenberg 2005 0,57 (0,34, 0,78)<br />

Meta-<strong>analysis</strong> result<br />

[fixed]<br />

Gutzeit 2005 0,35 (0,14, 0,62)<br />

Graph 41 illustrates a meta-<strong>analysis</strong> <strong>of</strong> the diagnostic sensitivity <strong>of</strong> <strong>CT</strong> in patients with<br />

metastasis to the neck area.<br />

0,0 0,2 0,4 0,6 0,8<br />

Diagnostic sensitivity (95% confidence<br />

interval)<br />

0,47 (0,31, 0,64)


Graph 41.<br />

Meta-<strong>analysis</strong> <strong>of</strong> <strong>CT</strong> diagnostic sensitivity in patients with metastasis to the neck area.<br />

The average diagnostic sensitivity <strong>of</strong> <strong>CT</strong> is 26% (95% CI: 13; 44), which means that if <strong>CT</strong> is<br />

used the probability <strong>of</strong> primary carcinoma detection in patients with metastasis to the<br />

neck area is 26%.<br />

The average values <strong>of</strong> diagnostic sensitivity for <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> are listed in table 133.<br />

Table 133.<br />

Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> sensitivity in primary carcinoma detection in patients with metastasis to the neck<br />

area.<br />

Parameter<br />

Se (95% CI)<br />

<strong>PET</strong>-<strong>CT</strong><br />

47% (31; 64)<br />

<strong>CT</strong><br />

26% (13; 44)<br />

Based on the data above, the diagnostic sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in primary carcinoma<br />

detection in patients with metastasis to the neck area is almost two times higher than that <strong>of</strong><br />

<strong>CT</strong>.<br />

Freudenberg 2005 0,28 (0,10, 0,53)<br />

Gutzeit 2005 0,25 (0,07, 0,52)<br />

Meta-<strong>analysis</strong> result {fixed] 0,26 (0,13, 0,44)<br />

0,0 0,2 0,4 0,6<br />

Diagnostic sensitivity (95% confidence interval)<br />

The odds ratio for primary carcinoma detection in patients with metastasis to the neck<br />

area as calculated by meta-<strong>analysis</strong> using <strong>PET</strong>-<strong>CT</strong> or <strong>CT</strong> is presented in graph 42.<br />

261


Graph 42.<br />

Odds ratio for primary carcinoma detection in patients with metastasis to the neck area<br />

262<br />

Freudenberg 2005 4,27 (0,96, 20,24)<br />

The odds ratio as calculated by meta-<strong>analysis</strong> <strong>of</strong> the two trials is 2.87 (95% CI: 1.08; 7.63),<br />

so the probability <strong>of</strong> locating primary carcinoma in patients with metastasis to the neck area<br />

is almost three times higher for <strong>PET</strong>-<strong>CT</strong> than for <strong>CT</strong>, and the result is statistically significant.<br />

In order to obtain one additional case <strong>of</strong> primary carcinoma detection it is necessary to use<br />

<strong>PET</strong>-<strong>CT</strong> instead <strong>of</strong> <strong>CT</strong> with 5 patients with metastasis to the neck area, NNT = 5 (95% CI: 3; 35).<br />

Based on Gutzeit 2005 results, in patients with metastasis to the neck area, the sensitivity <strong>of</strong><br />

<strong>PET</strong>-<strong>CT</strong> is 36% (95% CI: 18; 57), and the sensitivity <strong>of</strong> <strong>CT</strong> is 15% (95% CI: 4; 35).<br />

The odds ratio for primary carcinoma detection in patients with metastasis to the neck<br />

area as calculated based on a single study is 3.09 (95% CI: 0.69; 15.93). It means that for<br />

<strong>PET</strong>-<strong>CT</strong>, the probability <strong>of</strong> primary carcinoma detection is over three times higher than for<br />

<strong>CT</strong>, but the result is not statistically significant.<br />

Based on the results <strong>of</strong> a single trial, the diagnostic sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> was higher as<br />

compared to <strong>CT</strong> in locating primary carcinoma in the total population with metastasis to the<br />

neck area and outside the neck area, and was 36% (95% CI: 22; 52) vs. 19% (95% CI: 9; 34).<br />

used.<br />

Gutzeit 2005 1,75 (0,32, 10,43)<br />

Meta-<strong>analysis</strong> result [fixed] 2,87 (1,08, 7,63)<br />

17.3.4.2 Safety<br />

0,2 0,5 1 2 5 10 100<br />

The trials under <strong>analysis</strong> provide no information on the safety <strong>of</strong> the diagnostic procedures<br />

17.3.5. Results<br />

Odds ratio (95% confidence interval)<br />

Two primary clinical trials were found that compared directly the diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>


with that <strong>of</strong> <strong>CT</strong> in locating primary neoplasm in metastasis patients. Both testing methods were<br />

verified against the reference test (histopathological examination and/or clinical observation).<br />

Based on a meta-<strong>analysis</strong>, for patients with metastasis to the neck area the diagnostic<br />

sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> is higher than the values established for <strong>CT</strong>: 47% (95% CI: 31; 64) vs. 26% (95%<br />

CI: 13; 44). Another meta-<strong>analysis</strong> showed that the odds <strong>of</strong> determining the location <strong>of</strong> primary<br />

neoplasm in a population <strong>of</strong> patients with metastasis in the neck area using <strong>PET</strong>-<strong>CT</strong> was almost<br />

three times higher compared to <strong>CT</strong>, and the result was statistically significant; OR = 2.87 (95% CI:<br />

1.08; 7.63), NNT = 5 (95% CI: 3; 35).<br />

A single-trial <strong>analysis</strong> <strong>of</strong> the diagnostic efficacy <strong>of</strong> the technologies compared in locating<br />

primary tumor in patients with cancer metastases outside the neck indicated no statistically<br />

significant differences between <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong>. The diagnostic sensitivity <strong>of</strong> the <strong>PET</strong>-<strong>CT</strong> method<br />

was higher in versus <strong>CT</strong>, and stood at 36% (95% CI: 18; 57) vs. 15% (95% CI: 4; 35).<br />

The results <strong>of</strong> a single trial also warranted the claim that the diagnostic sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong><br />

proved higher than the sensitivity <strong>of</strong> <strong>CT</strong> in locating cancer focuses in the total population <strong>of</strong><br />

patients with cancer metastases both in and outside the neck, and was 36% (95% CI: 22; 52) for<br />

<strong>PET</strong>-<strong>CT</strong> vs. 19% (95% CI: 9; 34) for <strong>CT</strong>.<br />

263


18. APPENDICES<br />

264<br />

18.1. Tool for quality assessment <strong>of</strong> studies<br />

Table 134.<br />

Antoch 2003 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e. the<br />

index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail to<br />

permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without knowledge<br />

<strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 135.<br />

Lardinois 2003 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e. the<br />

index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail to<br />

permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without knowledge<br />

<strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

14 Were withdrawals from the study explained? +<br />

Answer<br />

Yes No Unclear<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

265


Table 136.<br />

Cerfolio 2005 quality assessment: QUADAS checklist<br />

1<br />

266<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail to<br />

permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without knowledge<br />

<strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

14 Were withdrawals from the study explained? +<br />

Answer<br />

Yes No Unclear<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 137.<br />

Shim 2005 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without knowledge<br />

<strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

267


Table 138.<br />

Antoch 2004 quality assessment: QUADAS checklist (mixed)<br />

1<br />

268<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

14 Were withdrawals from the study explained? +<br />

Answer<br />

Yes No Unclear<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 139.<br />

Antoch 2003 quality assessment: QUADAS checklist (mixed)<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

14 Were withdrawals from the study explained? +<br />

Answer<br />

Yes No Unclear<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

269


Table 140.<br />

Schmidt 2005 quality assessment: QUADAS checklist<br />

1<br />

270<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

14 Were withdrawals from the study explained? +<br />

Answer<br />

Yes No Unclear<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 141.<br />

Freudenberg 2005 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

271


Table 142.<br />

Gutzeit 2005 quality assessment: QUADAS checklist<br />

1<br />

272<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 143.<br />

Freudenberg 2003 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported?<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

273


Table 144.<br />

Schaefer 2004 quality assessment: QUADAS checklist<br />

1<br />

274<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported?<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 145.<br />

Cerfolio 2005 quality assessment: QUADAS checklist (esopaegal and gastric cancer)<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported?<br />

14 Were withdrawals from the study explained? +<br />

Answer<br />

Yes No Unclear<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

275


Table 146.<br />

Kula 2005 quality assessment: QUADAS checklist<br />

1<br />

276<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported?<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 147.<br />

Grisaru 2004 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported?<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

277


Table 148.<br />

Hauth 2005 quality assessment: QUADAS checklist<br />

1<br />

278<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e. the<br />

index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail to<br />

permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without knowledge<br />

<strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 149.<br />

Makhija 2001 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e. the<br />

index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail to<br />

permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without knowledge<br />

<strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

279


Table 150.<br />

Nahas 2005 quality assessment: QUADAS checklist<br />

1<br />

280<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported?<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 151.<br />

Freudenberg 2004 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

281


Table 152.<br />

Zimmer 2003 quality assessment: QUADAS checklist<br />

1<br />

282<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 153.<br />

Wild 2006 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

283


Table 154.<br />

Branstetter 2005 quality assessment: QUADAS checklist<br />

1<br />

284<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 155.<br />

Goerres 2005 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

285


Table 156.<br />

Koshy 2005 quality assessment: QUADAS checklist<br />

1<br />

286<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 157.<br />

Heinrich 2005 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

287


Table 158.<br />

Antoch 2004 quality assessment: QUADAS checklist<br />

1<br />

288<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

Answer<br />

Yes No Unclear<br />

14 Were withdrawals from the study explained? +<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+


Table 159.<br />

Veit 2006 quality assessment: QUADAS checklist<br />

1<br />

Quality criteria<br />

Was the spectrum <strong>of</strong> patients representative <strong>of</strong> the patients who<br />

will receive the test in practice?<br />

2 Were selection criteria clearly described? +<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

Is the reference standard likely to correctly classify the target<br />

condition?<br />

Is the time period between reference standard and index test<br />

short enough to be reasonably sure that the target condition did<br />

not change between the two tests?<br />

Did the whole sample or a random selection <strong>of</strong> the sample,<br />

receive verification using a reference standard?<br />

Did patients receive the same reference standard regardless <strong>of</strong><br />

the index test result?<br />

Was the reference standard independent <strong>of</strong> the index test (i.e.<br />

the index test did not form part <strong>of</strong> the reference standard)?<br />

Was the execution <strong>of</strong> the index test described in sufficient detail<br />

to permit replication <strong>of</strong> the test?<br />

Was the execution <strong>of</strong> the reference standard described in<br />

sufficient detail to permit its replication?<br />

Were the index test results interpreted without knowledge <strong>of</strong> the<br />

results <strong>of</strong> the reference standard?<br />

Were the reference standard results interpreted without<br />

knowledge <strong>of</strong> the results <strong>of</strong> the index test?<br />

Were the same clinical data available when test results were<br />

interpreted as would be available when the test is used in<br />

practice?<br />

13 Were uninterpretable/ intermediate test results reported? +<br />

14 Were withdrawals from the study explained? +<br />

Answer<br />

Yes No Unclear<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

289


290<br />

18.2. Detailed search results<br />

Table 160.<br />

Result <strong>of</strong> the serach in Cochrane database<br />

ID Strategy<br />

Serach<br />

result<br />

#1 MeSH descriptor Positron-Emission Tomography, this term only in MeSH products 122<br />

#2 MeSH descriptor Tomography, Emission-Computed, this term only in MeSH products 673<br />

#3 <strong>PET</strong> OR <strong>positron</strong> <strong>emission</strong> tomography in All Fields in all products 1288<br />

#4 (#1 OR #2 OR #3) 1407<br />

#5 MeSH descriptor Tomography, X-Ray Computed, this term only in MeSH products 1571<br />

#6 MeSH descriptor Tomography, Spiral Computed, this term only in MeSH products 66<br />

#7 <strong>CT</strong> OR computed tomography in All Fields in all products 17330<br />

#8 (#5 OR #6 OR #7) 17330<br />

#9<br />

dual OR integral OR integration OR combination OR combined OR fusion OR fused<br />

OR hybrid OR coincidental OR combining OR coincidence OR coregistered in All<br />

Fields in all products<br />

#10 (#4 AND #8) 271<br />

#11 (#4 AND #9) 257<br />

#12 (#10 OR #11) 421<br />

83610<br />

#13 MeSH descriptor Neoplasms, this term only in MeSH products 2707<br />

#14 MeSH descriptor Medical Oncology, this term only in MeSH products 74<br />

#15 (#13 OR #14) 2750<br />

#16 (#12 AND #15) 22


Table 161.<br />

Results <strong>of</strong> the serach in Pubmed data basis<br />

ID Strategy<br />

Serach<br />

result<br />

#1 "Positron-Emission Tomography"[MeSH] OR "Tomography, Emission-Computed"[MeSH] 39914<br />

#2 <strong>PET</strong> OR <strong>positron</strong> <strong>emission</strong> tomography 29430<br />

#3 (#1 OR #2) 50482<br />

#4 ("Tomography, Spiral Computed"[MeSH] OR "Tomography, X-Ray Computed"[MeSH]) 167516<br />

#5 <strong>CT</strong> OR computed tomography 243809<br />

#6 (#4 OR #5) 243809<br />

#7<br />

dual OR integral OR integration OR combination OR combined OR fusion OR fused<br />

OR hybrid OR coincidental OR combining OR coincidence OR coregistered<br />

1061412<br />

#8 (#3 AND #6) 12708<br />

#9 (#3 AND #7) 6428<br />

#10 (#8 OR #9) 16819<br />

#11 #8 OR #9 Limits: Humans 15079<br />

#12 #8 OR #9 Limits: English, Polish, Humans 12889<br />

#13 #8 OR #9 Limits: English, Polish, Publication Date from 1998/01/01, Humans 7786<br />

#14 "Neoplasms"[MeSH] Limits: English, Polish, Publication Date from 1998/01/01, Humans 418359<br />

#15 "Neoplasms"[MeSH] 1744479<br />

#16 "Medical Oncology"[MeSH] 6670<br />

#17 (#15 OR #16) 1747280<br />

#18 (#10 AND #17) 5240<br />

#19 #10 AND #17 Limits: English, Polish, Publication Date from 1998/01/01, Humans 3233<br />

291


Table 162.<br />

Results <strong>of</strong> serach in Embase database<br />

ID Strategy Serach result<br />

1 <strong>positron</strong> <strong>emission</strong> tomography.mp. OR exp Positron Emission Tomography 27340<br />

2<br />

292<br />

Emission Computed Tomography.mp. OR exp Computer Assisted Emission<br />

Tomography<br />

3 <strong>PET</strong>.mp. 19613<br />

4 (1 OR 2 OR 3) 36988<br />

5<br />

(dual OR integral OR integration OR combination OR combined OR fusion OR<br />

fused OR hybrid OR coincidental OR combining OR coincidence OR<br />

coregistered).mp. [mp=title, abstract, subject headings, heading word, drug<br />

trade name, original title, device manufacturer, drug manufacturer name]<br />

6301<br />

615862<br />

6 (4 AND 5) 4268<br />

7 exp Computer Assisted Tomography 183410<br />

8 <strong>CT</strong>.mp 82030<br />

9 (7 OR 8) 205988<br />

10 (4 AND 9) 13380<br />

11 (6 OR 10) 15661<br />

12 limit 11 to human 13700<br />

13 limit 12 to yr="1999 - 2006" 9119<br />

14 exp NEOPLASM 990525<br />

15 (11 AND 14) 5231<br />

16 limit 15 to (human and yr="1998 - 2006") 4213<br />

Table 163.<br />

MeSH terms used for the search strategy<br />

Term [MeSH]<br />

Positron-Emission Tomography<br />

Tomography, Emission-Computed<br />

Synonyms<br />

Positron Emission Tomography<br />

<strong>PET</strong> Scan<br />

<strong>PET</strong> Scans<br />

Scan, <strong>PET</strong><br />

Scans, <strong>PET</strong><br />

Tomography, Positron-Emission<br />

Tomography, Positron Emission<br />

Tomography, Emission Computed<br />

CAT Scan, Radionuclide<br />

CAT Scans, Radionuclide<br />

Radionuclide CAT Scan<br />

Radionuclide CAT Scans<br />

Scan, Radionuclide CAT<br />

Scans, Radionuclide CAT<br />

Computed Tomographic Scintigraphy<br />

Tomographic Scintigraphy, Computed<br />

Computerized Emission Tomography


Emission-Computed Tomography<br />

Emission Computed Tomography<br />

Scintigraphy, Computed Tomographic<br />

Tomography, Computerized Emission<br />

Emission Tomography, Computerized<br />

Radionuclide-Computed Tomography<br />

Radionuclide Computed Tomography<br />

Radionuclide Tomography, Computed<br />

Computed Radionuclide Tomography<br />

Tomography, Computed Radionuclide<br />

Tomography, Radionuclide-Computed<br />

Tomography, Radionuclide Computed<br />

Tomography, Spiral Computed Computed Tomographies, Spiral<br />

Computed Tomography, Spiral<br />

Spiral Computed Tomographies<br />

Tomographies, Spiral Computed<br />

Helical Computed Tomography<br />

Computed Tomographies, Helical<br />

Computed Tomography, Helical<br />

Helical Computed Tomographies<br />

Tomographies, Helical Computed<br />

Helical <strong>CT</strong><br />

<strong>CT</strong>, Helical<br />

<strong>CT</strong>s, Helical<br />

Helical <strong>CT</strong>s<br />

Spiral Computed Tomography<br />

Spiral <strong>CT</strong><br />

<strong>CT</strong>, Spiral<br />

<strong>CT</strong>s, Spiral<br />

Spiral <strong>CT</strong>s<br />

Spiral Volumetric <strong>CT</strong><br />

<strong>CT</strong>, Spiral Volumetric<br />

<strong>CT</strong>s, Spiral Volumetric<br />

Spiral Volumetric <strong>CT</strong>s<br />

Volumetric <strong>CT</strong>, Spiral<br />

Volumetric <strong>CT</strong>s, Spiral<br />

Tomography, Helical Computed<br />

Tomography, Spiral Volumetric Computed<br />

Tomography, X-Ray Computed • Computed Tomographies, X-Ray<br />

• Computed Tomography, X-Ray<br />

• Tomographies, X-Ray Computed<br />

• X-Ray Computed Tomographies<br />

• X-Ray Computed Tomography<br />

• Computerized Tomography, X Ray<br />

• Computerized Tomography, X-Ray<br />

• Computerized Tomographies, X-Ray<br />

• Tomographies, X-Ray Computerized<br />

• Tomography, X-Ray Computerized<br />

• X-Ray Computerized Tomographies<br />

• X-Ray Computerized Tomography<br />

293


294<br />

• <strong>CT</strong> X Ray<br />

• <strong>CT</strong> X Rays<br />

• X Ray, <strong>CT</strong><br />

• X Rays, <strong>CT</strong><br />

• Tomodensitometry<br />

• Tomodensitometries<br />

• CAT Scan, X Ray<br />

• CAT Scan, X-Ray<br />

• CAT Scans, X-Ray<br />

• Scan, X-Ray CAT<br />

• Scans, X-Ray CAT<br />

• X-Ray CAT Scan<br />

• X-Ray CAT Scans<br />

• Tomography, Xray Computed<br />

• Computed Tomographies, Xray<br />

• Computed Tomography, Xray<br />

• Tomographies, Xray Computed<br />

• Xray Computed Tomographies<br />

• Xray Computed Tomography<br />

• Tomography, X Ray Computed<br />

• X Ray Tomography, Computed<br />

• X-Ray Tomography, Computed<br />

• Computed X-Ray Tomographies<br />

• Computed X-Ray Tomography<br />

• Tomographies, Computed X-Ray<br />

• Tomography, Computed X-Ray<br />

• X-Ray Tomographies, Computed<br />

• Cine-<strong>CT</strong><br />

• Cine <strong>CT</strong><br />

• Tomography, Transmission Computed<br />

• Computed Tomographies, Transmission<br />

• Computed Tomography, Transmission<br />

• Tomographies, Transmission Computed<br />

• Transmission Computed Tomographies<br />

• Transmission Computed Tomography<br />

• Electron Beam Computed Tomography<br />

• Electron Beam Tomography<br />

• Beam Tomographies, Electron<br />

• Beam Tomography, Electron<br />

• Electron Beam Tomographies<br />

• Tomographies, Electron Beam<br />

• Tomography, Electron Beam


Neoplasms Neoplasm<br />

Tumors<br />

Tumor<br />

Benign Neoplasms<br />

Neoplasms, Benign<br />

Benign Neoplasm<br />

Neoplasm, Benign<br />

Carcinoma<br />

Carcinomas<br />

Medical Oncology Oncology, Medical<br />

295


19. LITERATURE USED IN THE ANALYSIS<br />

296<br />

19.1. Lung cancer<br />

1. Antoch G, Stattaus J, Nemat AT, Marnitz S, Beyer T, Kuehl H, Bockisch A, Debatin JF,<br />

Freudenberg LS. Non-Small Cell Lung Cancer: Dual-Modality <strong>PET</strong>-<strong>CT</strong> in Preoperative Staging.<br />

Radiology 2003; Vol. 229 (2): pp 526–533.<br />

2. Lardinois D, Weder W, Hany TF, Kamel EM, Korom S, Seifert B, von Schulthess GK, Steinert HC.<br />

Staging <strong>of</strong> non-small-cell lung cancer with integrated <strong>positron</strong>-<strong>emission</strong> tomography and<br />

computed tomography. N Engl J Med 2003; 348: pp 2500–2507.<br />

3. Cerfolio RJ, Bryant AS, Ojha B, Eloubeidi M. Improving the inaccuracies <strong>of</strong> clinical staging <strong>of</strong><br />

patients with NSCLC: A prospective trial. Ann Thora Surg 2005; Vol. 80 (4): pp 1207–1214.<br />

4. Shim SS, Lee KS, Kim BT, Chung MJ, Lee EJ, Han J, Choi JY, Kwon OJ, Shim YM, Kim S. Non-small<br />

cell lung cancer: prospective comparison <strong>of</strong> integrated FDG <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> alone for<br />

preoperative staging. Radiology 2005 Sep; 236 (3): pp 1011–1019. Epub 2005 Jul 12.<br />

19.2. Neoplasm in varied locations<br />

1. Antoch G, Saoudi N, Kuehl H, Dahmen G, Mueller SP, Beyer T, Bockisch A, Debatin JF,<br />

Freudenberg LS. Accuracy <strong>of</strong> whole-body dual-modality fluorine-18-2-fluoro-2-deoxy-D-glucose<br />

<strong>positron</strong> <strong>emission</strong> tomography and computed tomography (FDG-<strong>PET</strong>-<strong>CT</strong>) for tumor staging in<br />

solid tumors: comparison with <strong>CT</strong> and <strong>PET</strong>. J Clin Oncol 2004; Vol. 1; 22 (21): pp 4357–4368.<br />

2. Antoch G, Vogt FM, Freudenberg LS, Nazaradeh F, Goehde SC, Barkhausen J, Dahmen G,<br />

Bockisch A, Debatin JF, Ruehm SG. Whole-body dual-modality <strong>PET</strong>-<strong>CT</strong> and whole-body MRI for<br />

tumor staging in oncology. JAMA 2003; Vol. 24; 290 (24): pp 3199–3206.<br />

3. Schmidt GP, Baur-Melnyk A, Herzog P, Schmid R, Tiling R, Schmidt M, Reiser MF, Schoenberg SO.<br />

High-resolution whole-body magnetic resonance image tumor staging with the use <strong>of</strong> parallel<br />

imaging versus dual-modality <strong>positron</strong> <strong>emission</strong> tomography-computed tomography:<br />

experience on a 32-channel system. Invest Radiol 2005; Vol. 40 (12): pp 743–753.<br />

4. Freudenberg LS, Fischer M, Antoch G, Jentzen W, Gutzeit A, Rosenbaum SJ, Bockisch A, Egelh<strong>of</strong><br />

T. Dual modality <strong>of</strong> 18F-fluorodeoxyglucose-<strong>positron</strong> <strong>emission</strong> tomography/computed<br />

tomography in patients with cervical carcinoma <strong>of</strong> unknown primary. Med Princ Pract 2005;<br />

Vol. 14 (3): pp 155–160.<br />

5. Gutzeit A, Antoch G, Kuhl H, Egelh<strong>of</strong> T, Fischer M, Hauth E, Goehde S, Bockisch A, Debatin J,<br />

Freudenberg L. Unknown primary tumors: detection with dual-modality <strong>PET</strong>-<strong>CT</strong> – initial<br />

experience. Radiology 2005; Vol. 234 (1): pp 227–234. Epub 2004 Nov 24.<br />

19.3. Lymphomas<br />

1. Freudenberg LS, Antoch G, Schütt P, Beyer T, Jentzen W, Müller SP, Görges R, Nowrousian MR,<br />

Bockisch A, Debatin JF. FDG-<strong>PET</strong>-<strong>CT</strong> in re-staging <strong>of</strong> patients with lymphoma. Eur J Nucl Med Mol<br />

Imaging 2004; 31 (3): pp 325–329.<br />

2. Schaefer NG, Hany TF, Taverna C, Seifert B, Stumpe KD, von Schulthess GK, Goerres GW. Non-<br />

Hodgkin lymphoma and Hodgkin disease: coregistered FDG <strong>PET</strong> and <strong>CT</strong> at staging and<br />

restaging – do we need contrast-enhanced <strong>CT</strong>? Radiology 2004 Sep; 232 (3): pp 823–829.


19.4. Esophageal cancer<br />

1. Cerfolio RJ, Bryant AS, Ohja B, Bartolucci AA, Eloubeidi MA. The accuracy <strong>of</strong> endoscopic<br />

ultrasonography with fine-needle aspiration, integrated <strong>positron</strong> <strong>emission</strong> tomography with<br />

computed tomography, and computed tomography in restaging patients with esophageal<br />

cancer after neoadjuvant chemoradiotherapy. J Thorac Cardiovasc Surg 2005; Vol. 129 (6): pp<br />

1232–1241.<br />

2. Kula Z, Pietrzak T, Kobus-Błachnio K, Zuchora Z. Combined <strong>positron</strong> <strong>emission</strong> tomography and<br />

computed tomography (<strong>PET</strong>-<strong>CT</strong>) imaging in staging esophageal cancer – <strong>analysis</strong> <strong>of</strong> 12 casas.<br />

Współczesna Onkologia 2005; vol. 9 (8): pp 336–341.<br />

19.5. Cancer in the female genitals<br />

1. Grisaru D, Almog B, Levine C, Metser U, Fishman A, Lerman H, Lessing JB, Even-Sapir E. The<br />

diagnostic accuracy <strong>of</strong> 18F-fluorodeoxyglucose <strong>PET</strong>-<strong>CT</strong> in patients with gynecological<br />

malignancies. Gynecol Oncol 2004 Sep; 94 (3): pp 680–684.<br />

19.6. Ovarian cancer<br />

1. Hauth EA, Antoch G, Stattaus J, Kuehl H, Veit P. Evaulation <strong>of</strong> integrated whole-body <strong>PET</strong>/ <strong>CT</strong> in<br />

detection <strong>of</strong> recurrent ovarian cancer. Eur J Radiol 2005 Nov; 56 (2): pp 263–268.<br />

2. Makhija S, Howden N, Edwards R, Kelley J, Townsed DW, Meltzer CC. Positron <strong>emission</strong><br />

tomography/computed tomography imaging for the detection <strong>of</strong> recurrent ovarian and<br />

fallopian tube carcinoma; a retrospective review. Gynecol Oncol 2002 Apr; 85 (1): pp 53–58.<br />

19.7. Thyroid gland cancer<br />

1. Nahas Z, Goldenberg D, Fakhry C, Ewertz M, Zeiger M, Ladenson PW, Wahl R, Tufano RP. The role<br />

<strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography/computed tomography in the management <strong>of</strong> recurrent<br />

papillary thyroid carcinoma. Laryngoscope 2005 Feb; 115 (2): pp 237–243.<br />

2. Freudenberg LS, Antoch G, Jentzen W, Pink R, Knust J, Gorges R, Muller SP, Bockisch A, Debatin<br />

JF, Brandau W. Value <strong>of</strong> 124I -<strong>PET</strong>-<strong>CT</strong> in staging <strong>of</strong> patients with differentiated thyroid cancer. Eur<br />

Radiol 2004; Vol. 14 (11): pp 2092–2098.<br />

3. Zimmer LA, McCook B, Meltzer C, Fukui M, Bascom D, Snyderman C, Townsend DW, Johnson JT.<br />

Combined <strong>positron</strong> <strong>emission</strong> tomography/computed tomography imaging <strong>of</strong> recurrent thyroid<br />

cancer. Otolaryngology – Head & Neck Surgery 2003; Vol. 128 (2): pp 178–184. Date <strong>of</strong><br />

Publication: 01 FEB 2003.<br />

19.8. Head and neck cancer<br />

1. Wild D, Eyrich GK, Ciernik IF, Stoeckli SJ, Schuknecht B, Goerres GW. In-line 18Ffluorodeoxyglucose<br />

<strong>positron</strong> <strong>emission</strong> tomography with computed tomography (<strong>PET</strong>-<strong>CT</strong>) in<br />

patients with carcinoma <strong>of</strong> the sinus/nasal area and orbit. Journal <strong>of</strong> Cranio-Maxillo-Facial<br />

Surgery 2006; Vol. 34 (1): pp 9–16.<br />

2. Branstetter IV BF, Blodgett TM, Zimmer LA, Snyderman CH, Johnson JT, Raman S, Meltzer CC.<br />

Head and neck malignancy: Is <strong>PET</strong>-<strong>CT</strong> more accurate than <strong>PET</strong> or <strong>CT</strong> alone?. Radiology 2005;<br />

Vol. 235 (2): pp 580–586.<br />

297


298<br />

3. Goerres GW, Schmid DT, Schuknecht B, Eyrich GK. Bone invasion in patients with oral cavity<br />

cancer: Comparison <strong>of</strong> conventional <strong>CT</strong> with <strong>PET</strong>-<strong>CT</strong> and SPE<strong>CT</strong>/<strong>CT</strong>. Radiology 2005; Vol. 237 (1):<br />

pp 281–287.<br />

4. Koshy M, Paulino AC, Howell R, Schuster D, Halkar R, Davis LW. F-18 FDG <strong>PET</strong>-<strong>CT</strong> fusion in<br />

radiotherapy treatment planning for head and neck cancer. Head & Neck 2005; Vol. 27 (6): pp<br />

494–502.<br />

19.9. Pancreatic cancer<br />

1. Heinrich S, Goerres GW, Schafer M, Sagmeister M, Bauerfeind P, Pestalozzi BC, Hany TF, von<br />

Schulthess GK, Clavien P. Positron <strong>emission</strong> tomography/computed tomography influences on<br />

the management <strong>of</strong> resectable pancreatic cancer and its cost-<strong>effectiveness</strong>. Ann Surg 2005;<br />

Vol. 242 (2): pp 235-243.<br />

19.10. Sarcomas<br />

1. Antoch G, Kanja J, Bauer S, Kuehl H, Renzing-Koehler K, Schuette J, Bockisch A, Debatin JF,<br />

Freudenberg LS. Comparison <strong>of</strong> <strong>PET</strong>, <strong>CT</strong>, and dual-modality <strong>PET</strong>-<strong>CT</strong> imaging for monitoring <strong>of</strong><br />

imatinib (STI571) therapy in patients with gastrointestinal stromal tumors. J Nucl Med 2004 Mar;<br />

45 (3): pp 357–365.<br />

19.11. Colon cancer<br />

1. Veit P, Antoch G, Stergar H, Bockisch A, Forsting M, Kuehl H. Detection <strong>of</strong> residual tumor after<br />

radi<strong>of</strong>requency ablation <strong>of</strong> liver metastasis with dual-modality <strong>PET</strong>-<strong>CT</strong>: Initial results. Eur Radiol<br />

2006; Vol. 16 (1): pp 80–87.


299


20. TRIALS EXCLUDED FROM THE ANALYSIS<br />

1. Abe K, Sasaki M, Kuwabara Y, Koga H, Baba S, Hayashi K, Takahashi N, Honda H. Comparison <strong>of</strong><br />

18 FDG-<strong>PET</strong> with 99m Tc-HMDP scintigraphy for the detection <strong>of</strong> bone metastases in patients with breast<br />

cancer. Ann Nucl Med 2005; Vol. 19 (7): pp 573–579. (absence <strong>of</strong> assessed intervention)<br />

2. Aberle DR, Chiles C, Gatsonis C, Hillman BJ, Johnson CD, McClennan BL, Mitchell DG, Pisano ED,<br />

Schnall MD, Sorensen AG. Imaging and cancer: research strategy <strong>of</strong> the American College <strong>of</strong><br />

Radiology Imaging Network. Radiology 2005; Vol. 235 (3): pp 741–751. (secondary trial)<br />

3. Acker MR, Burrell SC. Utility <strong>of</strong> 18F-FDG <strong>PET</strong> in evaluating cancers <strong>of</strong> lung. J Nucl Med Technol 2005<br />

Jun; 33 (2): pp 69–74; quiz 75–77. (secondary trial)<br />

4. Aigner RM, Schultes G, Wolf G, Yamashita Y, Sorantin E, Karcher H. F-18-FDG <strong>PET</strong>: early postoperative<br />

period <strong>of</strong> oro-maxillo-facial flaps. Nuclearmedizin 2003; Vol. (42): pp 210–214. (inadequate<br />

population)<br />

5. Aizer-Dannon A, Bar-Am A, Ron IG, Flusser G, Even-Sapir E. Fused functional-anatomic images <strong>of</strong><br />

metastatic cancer <strong>of</strong> cervix obtained by a combined gamma camera and an X-ray tube hybrid<br />

system with an illustrative case and review <strong>of</strong> the 18 F-fluorodeoxyglukose literature. Gynecol Oncol<br />

2003 Aug; 90 (2): pp 453–457. (casuistic description)<br />

6. Akeboshi M, Yamakado K, Nakatsuka A, Hataji O, Taguchi O, Takao M, Takeda K. Percutaneous<br />

radi<strong>of</strong>requency ablation <strong>of</strong> lung neoplasms: initial therapeutic response. J Vasc Interv Radiol 2004<br />

May; 15 (5): pp 463–470. (absence <strong>of</strong> assessed intervention)<br />

7. Alavi A, Lakhani P, Mavi A, Kung JW, Zhuang H. <strong>PET</strong>: A revolution in medical imaging. Radiol Clin<br />

N Am 2004; Vol. 42 (6): pp 983–1001. (secondary trial)<br />

8. Allen-Auerbach M, Quon A, Weber WA, Obrzut S, Crawford T, Silverman DH, Ratib O, Phelps ME,<br />

Czernin J. Comparison between 2-deoxy-2-[18F]fluoro-D-glucose <strong>positron</strong> <strong>emission</strong> tomography and<br />

<strong>positron</strong> <strong>emission</strong> tomography/computed tomography hardware fusion for staging <strong>of</strong> patients with<br />

lymphoma. Mol Imaging Biol 2004 Nov-Dec; 6 (6): pp 411–416. (absence <strong>of</strong> assessed intervention)<br />

9. Ambrosini V, Rubello D, Nanni C, Farsad M, Castellucci P, Franchi R, Fabbri M, Rampin L, Crepaldi G,<br />

Al-Nahhas A, Fanti S. Additional value <strong>of</strong> hybrid <strong>PET</strong>-<strong>CT</strong> fusion imaging vs. conventional <strong>CT</strong> scan<br />

alone in the staging and management <strong>of</strong> patients with malignant pleural mesothelioma. Nucl Med<br />

Rev 2005; Vol. 8 (2): pp 111–115. (absence <strong>of</strong> reference method)<br />

10. Amit A, Beck D, Lowenstein L, Lavie O, Bar Shalom R, Kedar Z, Israel O. The role <strong>of</strong> hybrid <strong>PET</strong>-<strong>CT</strong> in<br />

the evaluation <strong>of</strong> patients with cervical cancer. Gynecol Oncol 2006; Vol. 100 (1): pp 65–69.<br />

(absence <strong>of</strong> the classical comparative method)<br />

11. An Y-S, Yoon J-K, Lee M-H, Joh C-W, Yoon S-N. False negative F-18 FDG <strong>PET</strong>-<strong>CT</strong> in nonsmall cell lung<br />

cancer bone metastases. J Nucl Med 2005; Vol. 30 (3): pp 203–204. (casuistic description)<br />

12. Anderson GS, Brinkmann F, Soulen MC, Alavi A, Zhuang H. FDG <strong>positron</strong> <strong>emission</strong> tomography in the<br />

surveillance <strong>of</strong> hepatic tumors treated with radi<strong>of</strong>requency ablation. Clin Nucl Med 2003 Mar; 28<br />

(3): pp 192–197. (absence <strong>of</strong> assessed intervention)<br />

13. Annovazzi A, Peeters M, Maenhout A, Signore A, Dierckx R, Van de Wiele C. 18-Fluorodeoxyglucose<br />

<strong>positron</strong> <strong>emission</strong> tomography in nonendocrine neoplastic disorders <strong>of</strong> the gastrointestinal tract.<br />

Gastroenterology 2003; Vol. 125 (4): pp 1235–1245. (secondary trial)<br />

14. Anonymous. Recent advances and future perspectives in the management <strong>of</strong> lung cancer. Curr<br />

Probl Surg 2005; Vol. 42 (8): pp 548–610. (secondart trial)<br />

15. Antoch G, Freudenberg LS, Beyer T, Bockisch A, Debatin JF. To enhance or not to enhance? 18F-<br />

FDG and <strong>CT</strong> contrast agents in dual-modality. J Nucl Med 2004 Jan; 45 (Suppl 1): pp 56S–65S.<br />

(secondary trial)<br />

16. Aquino SL, Asmuth JC, Moore RH, Weise SB, Fischman AJ. Improved image interpretation with<br />

registered thoracic <strong>CT</strong> and <strong>positron</strong> <strong>emission</strong> tomography data sets. AJR Am J Roentgenol 2002 Apr;<br />

178 (4): pp 939–944. (absence <strong>of</strong> assessed intervention)<br />

300


17. Bachaud JM, Marre D, Dygai I, Caselles O, Hamelin D, Begue M, Laprie A, Zerdoud S, Gancel M,<br />

Courbon F. The impact <strong>of</strong> 18F-fluorodeoxyglucose <strong>positron</strong> <strong>emission</strong> tomography on the 3D<br />

conformal radiotherapy planning in patients with non-small cell lung cancer. Cancer Radiotherapie<br />

2005; Vol. 9 (8): pp 602–609. (article in French)<br />

18. Bagheri B, Maurer AH, Cone L, Doss M, Adler L. Characterization <strong>of</strong> the normal adrenal gland with<br />

18F-FDG <strong>PET</strong>-<strong>CT</strong>. J Nucl Med 2004 Aug; 45 (8): pp 1340–1343. (absence <strong>of</strong> assessed intervention)<br />

19. Bar-Shalom R, Gaitini D, Keidar Z, Israel O. Non-malignant FDG uptake in infradiaphragmatic<br />

adipose tissue: a new site <strong>of</strong> physiological tracer biodistribution characterised by <strong>PET</strong>-<strong>CT</strong>. Eur J Nucl<br />

Med Mol Imaging 2004 Aug; 31 (8): pp 1105–1113. Epub 2004 Mar 9. (absence <strong>of</strong> assessed<br />

intervention)<br />

20. Bar-Shalom R, Guralnik L, Tsalic M, Leiderman M, Frenkel A, Gaitini D, Ben-Nun A, Keidar Z, Israel O.<br />

The additional value <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> over <strong>PET</strong> in FDG imaging <strong>of</strong> oesophageal cancer. European Eur<br />

J Nucl Med Mol Imaging 2005; Vol. 32 (8): pp 918–924. (absence <strong>of</strong> assessed intervention)<br />

21. Bar-Shalom R, Yefremov N, Guralnik L, Gaitini D, Frenkel A, Kuten A, Altman H, Keidar Z, Israel O.<br />

Clinical performance <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in evaluation <strong>of</strong> cancer: additional value for diagnostic imaging and<br />

patient management. J Nucl Med 2003; Vol. 44 (8): pp 1200–1209. (absence <strong>of</strong> comparative<br />

diagnostic test)<br />

22. Belohlavek O, Klener J, Vymazal J, Dbaly V, Tovarys F. The diagnostics <strong>of</strong> recurrent gliomas using<br />

FDG-<strong>PET</strong>: Still questionable. Nuclear Medicine Review 2002; Vol. 5 (2): pp 127–130. (absence <strong>of</strong><br />

assessed intervention)<br />

23. Benchaou M, Lehmann W, Slosman DO, Becker M, Lemoine R, Rufenacht D, Donath A. The role <strong>of</strong><br />

FDG-<strong>PET</strong> in the preoperative assessment <strong>of</strong> N-staging in head and neck cancer. Acta Otoaryngol<br />

1996; Vol. 116 (2): pp 332–335. (absence <strong>of</strong> assessed intervention)<br />

24. Berlangieri SU, Brizel DM. Scher RL, Schifter T, Hawk TC, Hamblen S, Coleman RE, H<strong>of</strong>fman JM. Pilot<br />

study <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography in patients with advanced head and neck cancer receiving<br />

radiotherapy and chemotherapy. Head & Neck 1994; Vol. 16 (4): pp 340–346. (absence <strong>of</strong> assessed<br />

intervention)<br />

25. Berthelsen AK, Holm S, L<strong>of</strong>t A, Klausen TL, Andersen F, Hojgaard L. <strong>PET</strong>-<strong>CT</strong> with intravenous contrast<br />

can be used for <strong>PET</strong> attenuation correction in cancer patients. Eur J Nucl Med Mol Imaging 2005;<br />

Vol. 32 (10): pp 1167–1175. (absence <strong>of</strong> reference method)<br />

26. Beyer T, Townsend DW, Brun T, Kinahan PE, Charron M, Roddy R, Jerin J, Young J, Byars L, Nutt R.<br />

A combined <strong>PET</strong>-<strong>CT</strong> scanner for clinical oncology. J Nucl Med 2000; Vol. 41 (8): pp 1369–1379.<br />

(absence <strong>of</strong> assessed intervention)<br />

27. Bienert M, McCook B, Carr BI, Geller DA, Sheetz M, Tutor C, Amesur N, Avril N. 90 Y microsphere<br />

treatment <strong>of</strong> unresectable liver metastases: Changes in 18 F-FDG uptake and tumour size on <strong>PET</strong>-<strong>CT</strong>.<br />

Eur J Nucl Med Mol Imaging 2005; Vol. 32 (7): pp 778–787. (absence <strong>of</strong> end-points)<br />

28. Blake MA, Slattery JMA, Kalra MK, Halpern EF, Fischman AJ, Mueller PR, Boland GW. Adrenal lesions:<br />

Characterization with fused <strong>PET</strong>-<strong>CT</strong> image in patients with proved or suspected malignancy – Initial<br />

experience. Radiology 2006; Vol. 238 (3): pp 970–977. (absence <strong>of</strong> comparative method)<br />

29. Blake MA, Sweeney AT, Kalra MK, Maher MM. Collision adrenal tumors on <strong>PET</strong>/ <strong>CT</strong>. AJR Am J<br />

Roentgenol 2004; Vol. 183 (3): pp 864–865. (casuistic description)<br />

30. Blomqvist L, Torkzad MR. Whole-Body Imaging with MRI or <strong>PET</strong>-<strong>CT</strong>: The Future for Single-Modality<br />

Imaging in Oncology. J Am Med Association 2003; Vol. 290 (24): pp 3248–3249. Date <strong>of</strong> Publication:<br />

24 DEC 2003. (secondary trial)<br />

31. Bockisch A, Brandt-Mainz K, Gorges R, Muller St, Stattaus J, Antoch G. Diagnosis in medullary thyroid<br />

cancer with [ 18 F] FDG-<strong>PET</strong> and improvement using a combined <strong>PET</strong>-<strong>CT</strong> scanner. Acta Medica<br />

Austriaca 2003; Vol. 30 (1): pp 22–25. (secondary trial)<br />

32. Bradley JD, Dehdashti F, Mintun MA, Govindan R, Trinkaus K, Siegel BA. Positron <strong>emission</strong><br />

tomography in limited-stage small-cell lung cancer: a prospective study. J Clin Oncol 2004 Aug 15;<br />

22 (16): pp 3248–3254. (absence <strong>of</strong> assessed intervention)<br />

33. Bradley JD, Perez CA, Dehdashti F, Siegel BA. Implementing biologic target volumes in radiation<br />

treatment planning for non-small cell lung cancer. J Nucl Med 2004 Jan; 45 Suppl 1: pp 96S–101S.<br />

(secondary trial)<br />

301


34. Braun V, Dempf S, Weller R, Reske S-N, Schachenmayr W, Richter HP. Cranial neuronavigation with<br />

direct integration <strong>of</strong> 11 C methionine <strong>positron</strong> <strong>emission</strong> tomography (<strong>PET</strong>) data – Results <strong>of</strong> a pilot<br />

study in 32 surgical cases. Acta Neurochirurgica 2002; Vol. 144 (8): pp 777–782. (absence <strong>of</strong><br />

assessed intervention)<br />

35. Brianzoni E, Rossi G, Ancidei S, Berbellini A, Capoccetti F, Cidda C, D'Avenia P, Fattori S, Montini GC,<br />

Valentini G, Proietti A. Radiotherapy planning: <strong>PET</strong>-<strong>CT</strong> scanner performances in the definition <strong>of</strong><br />

gross tumour volume and clinical target volume. Eur J Nucl Med Mol Imaging 2005; Vol. 32 (12): pp<br />

1392–1399. (absence <strong>of</strong> comparative diagnostic test)<br />

36. Brink I, Klenzner Th, Krause Th, Mix M, Ross UH, Moser E, Nitzsche EU. Lymph node staging in<br />

extracranial head and neck cancer with FDG <strong>PET</strong> – Appropriate uptake period and sizedependence<br />

<strong>of</strong> the results. Nuklearmedizin 2002; Vol. 41 (2): pp 108–113. (absence <strong>of</strong> assessed<br />

diagnostic method)<br />

37. Bristow RE, del Carmen MG, Pannu HYK, Cohade C, Zhurak ML, Fishman EK, Wahl RL, Montz FJ.<br />

Clinically occult recurrent ovarian cancer: patient selection for secondary cytoreductive surgery<br />

using combined <strong>PET</strong>-<strong>CT</strong>. Gynecol Oncol 2003 Sep; 90 (3): pp 519–528. (absence <strong>of</strong> comparative<br />

diagnostic method)<br />

38. Brun E, Kjellen E, Tennvall J, Ohlsson T, Sandell A, Perfekt R, Wennerberg J, Strand SE. FDG <strong>PET</strong> studies<br />

during treatment: Prediction <strong>of</strong> therapy outcome in head and neck squamous cell carcinoma.<br />

Head & Neck 2002; Vol. 24 (2): pp 127–135. (absence <strong>of</strong> assessed intervention)<br />

39. Buell U, Wieres FJ, Schneider W, Reinartz P. 18 FDG-<strong>PET</strong> in 733 consecutive patients with or without<br />

side-by-side <strong>CT</strong> evaluation: Analysis <strong>of</strong> 921 lesions. Nuklearmedizin 2004; Vol. 43 (6): pp 210–216.<br />

(absence <strong>of</strong> assessed intervention)<br />

40. Caldwell CB, Mah K, Skinner M, Danjoux CE. Can <strong>PET</strong> provide the 3D extent <strong>of</strong> tumor motion for<br />

individualized internal target volumes? A phantom study <strong>of</strong> the limitations <strong>of</strong> <strong>CT</strong> and the promise <strong>of</strong><br />

<strong>PET</strong>. Int J Radiat Oncol Biol Phys 2003 Apr 1; 55 (5): pp 1381–1393. (inadequate population)<br />

41. Calvo FA, Domper M, Matute R, Martinez-Lazaro R, Arranz JA, Desco M, Alvarez E, Carreras JL. 18F-<br />

FDG <strong>positron</strong> <strong>emission</strong> tomography staging and restaging in rectal cancer treated with<br />

preoperative chemoradiation. Int J Radiat Oncol Biol Phys. 2004 Feb 1; 58 (2): pp 528–535. (absence<br />

<strong>of</strong> assessed intervention)<br />

42. Castillo E, Lawler LP. Diagnostic radiology and nuclear medicine. J Surg Oncol 2005 Dec 1; 92 (3): pp<br />

191–202. (secondary trial)<br />

43. Cerfolio RJ, Ojha B, Bryant AS, Raghuveer V, Mountz JM, Bartolucci AA. The accuracy <strong>of</strong> integrated<br />

<strong>PET</strong>-<strong>CT</strong> compared with dedicated <strong>PET</strong> alone for the staging <strong>of</strong> patients with nonsmall cell lung<br />

cancer. Ann Thorac Surg 2004 Sep; 78 (3): pp 1017–1023. (absence <strong>of</strong> assessed intervention)<br />

44. Chaiken L, Rege S, Hoh C, Choi Y, Jabour B, Juillard G, Hawkins R, Parker R. Positron <strong>emission</strong><br />

tomography with fluorodeoxyglucose to evaluate tumor response and control after radiation<br />

therapy. Int J Radiat Oncol Biol Phys 1993; Vol. 27 (2): pp 455–464. (absence <strong>of</strong> assessed diagnostic<br />

method)<br />

45. Chander S, Ergun EL. Positron Emission Tomographic-Computed Tomographic imaging <strong>of</strong> uterine<br />

sarcoma. Clin Nucl Med 2003; Vol. 28 (5): pp 443–444. (secondary trial)<br />

46. Charron M, Beyer T, Bohnen NN, Kinahan PE, Dachille M. Jerin J, Nutt R, Meltzer CC, Villemagne V,<br />

Townsend DW. Image <strong>analysis</strong> in patients with cancer studied with a combined <strong>PET</strong> and <strong>CT</strong> scanner.<br />

Clin Nucl Med 2000; Vol. 25 (11): pp 905–910. (absence <strong>of</strong> assessed intervention)<br />

47. Chen J, Cheong JH, Yun MJ, Kim J, Lim JS, Hyung WJ, Noh SH. Improvement in preoperative staging<br />

<strong>of</strong> gastric adenocarcinoma with <strong>positron</strong> <strong>emission</strong> tomography. Cancer 2005 Jun 1; 103 (11): pp<br />

2383–2390. (absence <strong>of</strong> assessed intervention)<br />

48. Chen Y-K, Ding H-J, Su C-T, Shen Y-Y, Chen L-K, Liao AC, Hung T-Z, Hu F-L, Kao C-H. Application <strong>of</strong><br />

<strong>PET</strong> and <strong>PET</strong>-<strong>CT</strong> imaging for cancer screening. Anticancer Research 2004; Vol. 24 (6): pp 4103–4108.<br />

(only slight perecentage <strong>of</strong> patients had <strong>PET</strong>-<strong>CT</strong> test and there was no reference test)<br />

49. Cheng EY. Surgical management <strong>of</strong> sarcomas. Hematol Oncol Clin N Am 2005; Vol. 19 (3): pp 451–<br />

470. (secondary trial)<br />

50. Chessin DB, Kiran RP, Akhurst T, Guillem JG. The emerging role <strong>of</strong> 18F-fluorodeoxyglucose <strong>positron</strong><br />

<strong>emission</strong> tomography in the management <strong>of</strong> primary and recurrent rectal cancer. J Am Coll Surg<br />

2005 Dec; 201 (6): pp 948–956. (secondary trial)<br />

302


51. Chin BB, Chang PPL. Gastrointestinal malignancies evaluated with 18 F-fluoro-2-deoxyglucose<br />

<strong>positron</strong> <strong>emission</strong> tomography. Best Practice & Research in Clinical Gastroenterology 2006; Vol. 20<br />

(1): pp 3–21. (secondary trial)<br />

52. Cho D, Kocher O, Parker JA, Halmos B. Multiple <strong>positron</strong>-<strong>emission</strong> tomography false positives in<br />

a patient with malignant melanoma. Clinical Oncology (Royal College <strong>of</strong> Radiologists) 2006; Vol. 18<br />

(1): pp 87–88. (casuistic description)<br />

53. Cho Sm, Ha HK, Byun JY, Lee JM. Usefulness <strong>of</strong> FDG <strong>PET</strong> for assessemant <strong>of</strong> early recurrent eithelial<br />

ovarian cancer. AJR Am J Roentgenol 2002 Aug; 179 (2): pp 391–395. (absence <strong>of</strong> assessed<br />

intervention)<br />

54. Choi JY, Lee KS, Kwon OJ, Shim YM, Baek CH, Park K, Lee KH, Kim BT. Improved detection <strong>of</strong> second<br />

primary cancer using integrated [18F] fluorodeoxyglucose <strong>positron</strong> <strong>emission</strong> tomography and<br />

computed tomography for initial tumor staging. J Clin Oncol 2005; Vol. 20; 23 (30): pp 7654–7659.<br />

(absence <strong>of</strong> assessed diagnostic test)<br />

55. Choi MY, Lee KM, Chung JK, Lee DS, Jeong JM, Park JG, Kim JH, Lee MC. Correlation between<br />

serum CEA level and metabolic volume as determined by FDG <strong>PET</strong> in postoperative patients with<br />

recurrent colorectal cancer. Ann Nucl Med 2005 Apr; 19 (2): pp 123–129. (absence <strong>of</strong> assessed<br />

intervention)<br />

56. Ciernik IF, Dizendorf E, Baumert BG, Reiner B, Burger C, Davis JB, Lutolf UM, Steinert HC, Von<br />

Schulthess GK. Radiation treatment planning with an integrated <strong>positron</strong> <strong>emission</strong> and computer<br />

tomography (<strong>PET</strong>-<strong>CT</strong>): a feasibility study. Int J Radiat Oncol Biol Phys 2003; Vol. 1; 57 (3): pp 853–863.<br />

(absence <strong>of</strong> reference method)<br />

57. Civantos FJ, Gomez C, Duque C, Pedroso F, Goodwin WJ, Weed DT, Arnold D, M<strong>of</strong>fat F. Sentinel<br />

node biopsy in oral cavity cancer: correlation with <strong>PET</strong> scan and immunohistochemistry. Head Neck<br />

2003 Jan; 25 (1): pp 1–9. (absence <strong>of</strong> assessed intervention)<br />

58. Cohade C, Osman M, Leal J, Wahl RL. Direct comparison <strong>of</strong> 18 F-FDG <strong>PET</strong> and <strong>PET</strong>-<strong>CT</strong> in patients with<br />

colorectal carcinoma. J Nucl Med 2003; Vol. 44 (11): pp 1797–1803. (absence <strong>of</strong> the classical<br />

comparative method)<br />

59. Cohade C, Osman M, Marshall L, Wahl R. <strong>PET</strong>-<strong>CT</strong>: accuracy <strong>of</strong> <strong>PET</strong> and <strong>CT</strong> spatial registration <strong>of</strong> lung<br />

lesions. Eur J Nucl Med Mol Imaging 2003; Vol. 30 (5): pp 721–726. Epub 2003 Mar 1. (absence <strong>of</strong><br />

refrence test)<br />

60. Cohade C, Osman M, Nakamoto Y, Marshall L, Links J, Fishman E, Wahl R. Initial experience with oral<br />

contrast in <strong>PET</strong>-<strong>CT</strong>: phantom and clinical studies. J Nucl Med 2003; Vol. 44: pp 412–416. (inadequate<br />

population)<br />

61. Cohade C, Wahl RL. Applications <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography/computed tomography image<br />

fusion in clinical <strong>positron</strong> <strong>emission</strong> tomography-clinical use, interpretation methods, diagnostic<br />

improvements. Semin Nucl Med 2003 Jul; 33 (3): pp 228–237. (secondary trial)<br />

62. Collins BT, Gardner LJ, Verma AK, Lowe VJ, Dunphy FR, Boyd JH. Correlation <strong>of</strong> fine needle<br />

aspiration biopsy and fluoride-18 fluorodeoxyglucose <strong>positron</strong> <strong>emission</strong> tomography in the<br />

assessment <strong>of</strong> locally recurrent and metastatic head and neck neoplasia. Acta Cytologica 1998;<br />

Vol. 42 (6): pp 1325–1329. (absence <strong>of</strong> assessed intervention)<br />

63. Comans EF, Miles KA, Schirrmeister H. Staging <strong>of</strong> non-small-cell lung cancer with integrated <strong>PET</strong> and<br />

<strong>CT</strong>. N Engl J Med 2003 Sep 18; 349 (12): pp 1188–1190; author reply pp 1188–1190. (letters)<br />

64. D'Amico TA, Wong TZ, Harpole DH, Brown SD, Coleman RE. Impact <strong>of</strong> computed tomography<strong>positron</strong><br />

<strong>emission</strong> tomography fusion in staging patients with thoracic malignancies. Ann Thorac<br />

Surg 2002; Vol. 74 (1): pp 160–163. (absence <strong>of</strong> the classical comparative method)<br />

65. Dehdashti F, Siegel BA. Neoplasms <strong>of</strong> the esophagus and stomach. Semin Nucl Med 2004; Vol. 34<br />

(3): pp 198–208. (secondary trial)<br />

66. Delbeke D, Martin WH. <strong>PET</strong> and <strong>PET</strong>-<strong>CT</strong> for evaluation <strong>of</strong> colorectal carcinoma. Semin Nucl Med<br />

2004; Vol. 34 (3): pp 209–223. (secondary trial)<br />

67. Deniaud-Alexandre E, Touboul E, Lerouge D, Grahek D, Foulquier JN, Petegnief Y, Gres B, El Balaa H,<br />

Keraudy K, Kerrou K, Montravers F, Milleron B, Lebeau B, Talbot JN. Impact <strong>of</strong> computed<br />

tomography and 18F-deoxyglucose coincidence detection <strong>emission</strong> tomography image fusion for<br />

optimization <strong>of</strong> conformal radiotherapy in non-small-cell lung cancer. Int J Radiat Oncol Biol Phys<br />

2005 Dec 1; 63 (5): pp 1432–1441. Epub 2005 Aug 25. (absence <strong>of</strong> assessed intervention)<br />

303


68. Desai DC, Zervos EE, Arnold MW, Burak WE Jr, Mantil J, Martin EW Jr. Positron <strong>emission</strong> tomography<br />

affects surgical management in recurrent colorectal cancer patients. Ann Surg Oncol 2003 Jan-<br />

Feb; 10 (1): pp 59–64. (absence <strong>of</strong> assessed intervention)<br />

69. Di Martino E, Nowak B, Krombach GA, Sellhaus B, Hausmann R, Cremerius U, Bull U, Westh<strong>of</strong>en M.<br />

Results <strong>of</strong> pretherapeutic lymph node diagnosis in head and neck cancer: Clinical value <strong>of</strong> 18fluorodeoxyglucose<br />

<strong>positron</strong> <strong>emission</strong> tomography (<strong>PET</strong>). Laryngo- Rhino- Otol 2000; Vol. 79 (4): pp<br />

201–206. (article in German)<br />

70. Diaz-Montes TP, Jacene HA, Wahl RL, Bristow RE. Combined FDG-<strong>positron</strong> <strong>emission</strong> tomography and<br />

computed tomography for the detection <strong>of</strong> ovarian cancer recurrence in an inguinal hernia sac.<br />

Gynecol Oncol 2005; Vol. 98 (3): pp 510–512. (casuistic description)<br />

71. Dos Santos DT, Pereira Lima EN, Chojniak R, Cavalcanti MG. Topographic metabolic map <strong>of</strong> head<br />

and neck squamous cell carcinoma using 18F-FDG <strong>PET</strong> and <strong>CT</strong> image fusion. Oral Surg Oral Med<br />

Oral Pathol Oral Radiol Endod 2005; Vol. 100 (5): pp 619–625. (absence <strong>of</strong> the classical comparative<br />

method)<br />

72. Dresel S, Grammerstorff J, Schwenzer K, Brinkbaumer K, Schmid R, Pfluger T, Hahn K. [18F] FDG<br />

imaging <strong>of</strong> head and neck tumours: comparison <strong>of</strong> hybrid <strong>PET</strong> and morphological methods. Eur J<br />

Nucl Med Mol Imaging 2003 Jul; 30 (7): pp 995–1003. Epub 2003 May 9. (absence <strong>of</strong> assessed<br />

diagnostic method)<br />

73. Dresell S, Schwenzer K, Brinkbaumer K, Schmid R, Szeimies U, Popperl G, Hahn K. [F-18] FDG imaging<br />

<strong>of</strong> head and neck tumors: Comparison <strong>of</strong> Hybrid <strong>PET</strong>, dedicated <strong>PET</strong> and <strong>CT</strong>. Nuklearmedizin 2001;<br />

Vol. 40 (5): pp 172–178. (article in German)<br />

74. Eary JF, O'Sullivan F, Powitan Y, Chandhury KR, Vernon C, Bruckner JD, Conrad EU. Sarcoma tumor<br />

FDG uptake measured by <strong>PET</strong> and patient outcome: a retrospective <strong>analysis</strong>. Eur J Nucl Med Mol<br />

Imaging 2002 Sep; 29 (9): pp 1149–1154. Epub 2002 Jun 19. (absence <strong>of</strong> assessed intervention)<br />

75. Eifel PJ. Accurate diagnosis <strong>of</strong> regional metastasis from cervical cancer: Is <strong>PET</strong> the answer?. Int<br />

J Radiat Oncol Biol Phys 2004; Vol. 59 (3): pp 637–638. Date <strong>of</strong> Publication: 01 JUL 2004. (secondary<br />

trial)<br />

76. Ell PJ. <strong>PET</strong>-<strong>CT</strong> in oncology: A major technology for cancer care. Chang Gung Med J 2005; Vol. 28<br />

(5): pp 274–283. (secondary trial)<br />

77. Erasmus JJ, Truong MT, Smythe WR, Munden RF, Marom EM, Rice DC, Vaporciyan AA, Walsh GL,<br />

Sabl<strong>of</strong>f BS, Broemeling LD, Stevens CW, Pisters KM, Podol<strong>of</strong>f DA, Macapinlac HA. Integrated<br />

computed tomography-<strong>positron</strong> <strong>emission</strong> tomography in patients with potentially resectable<br />

malignant pleural mesothelioma: Staging implications. J Thorac Cardiovasc Surg 2005; Vol. 129 (6):<br />

pp 1364–1370. (absence <strong>of</strong> comparative diagnostic method)<br />

78. Erdi YE, Nehmeh SA, Pan T, Pevsner A, Rosenzweig KE, Mageras G, Yorke ED, Schoder H, Hsiao W,<br />

Squire OD, Vernon P, Ashman JB, Mostafavi H, Larson SM, Humm JL. The <strong>CT</strong> motion quantitation <strong>of</strong><br />

lung lesions and its impact on <strong>PET</strong>-measured SUVs. J Nucl Med 2004 Aug; 45 (8): pp 1287–1292.<br />

(absence <strong>of</strong> the end-points for assessment <strong>of</strong> diagnostic efficacy)<br />

79. Erdi YE, Rosenzweig K, Erdi AK, Macapinlac HA, Hu Y-C, Braban LE, Humm JL, Squire OD, Chui C-S,<br />

Larson SM, Yorke ED. Radiotherapy treatment planning for patients with non-small cell lung<br />

cancer using <strong>positron</strong> <strong>emission</strong> tomography (<strong>PET</strong>). Radiother Oncol 2002 Jan; 62 (1): pp 51–60.<br />

(fusion <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> images)<br />

80. Eschmann SM, Bitzer M, Paulsen F, Friedel G, Besenfelder H, Horger M, Reimold M, Dittmann H,<br />

Pfannenberg AC, Bares R. The benefit <strong>of</strong> functional-anatomical imaging with [18F]<br />

fluorodeoxyglucose utilizing a dual-head coincidence gamma camera with an integrated Xray<br />

transmission system in non-small cell lung cancer. Nucl Med Commun 2004 Sep; 25 (9): pp 909–<br />

915. (absence <strong>of</strong> assessed intervention)<br />

81. Eubank WB, Mank<strong>of</strong>f DA, Schmiedl UP, Winter TC 3rd, Fisher ER, Olshen AB, Graham MM, Eary JF.<br />

Imaging <strong>of</strong> oncologic patients: benefit <strong>of</strong> combined <strong>CT</strong> and FDG <strong>PET</strong> in the diagnosis <strong>of</strong><br />

malignancy. AJR Am J Roentgenol 1998; Vol. 171 (4): pp 1103–1110. (absence <strong>of</strong> assessed<br />

intervention)<br />

82. Even-Sapir E, Metser U, Flusser G, Zuriel L, Kollender Y, Lerman H, Lievshitz G, Ron I, Mishani E.<br />

Assessment <strong>of</strong> malignant skeletal disease: initial experience with 18F-fluoride <strong>PET</strong>-<strong>CT</strong> and comparison<br />

between 18F-fluoride <strong>PET</strong> and 18F-fluoride <strong>PET</strong>-<strong>CT</strong>. J Nucl Med 2004; Vol. 45 (2): pp 272–278.<br />

(absence <strong>of</strong> the classical comparative method)<br />

304


83. Even-Sapir E, Parag Y, Lerman H, Gutman M, Levine C, Rabau M, Figer A, Metser U. Detection <strong>of</strong><br />

recurrence in patients with rectal cancer: <strong>PET</strong>-<strong>CT</strong> after abdominoperineal or anterior resection.<br />

Radiology 2004; Vol. 232 (3): pp 815–822. Epub 2004 Jul 23. (absence <strong>of</strong> the classical comparative<br />

method)<br />

84. Farsad M, Schiavina R, Castellucci P, Nanni C, Corti B, Martorana G, Canini R, Grigioni W, Boschi S,<br />

Marengo M, Pettinato C, Salizzoni E, Monetti N, Franchi R, Fanti S. Detection and localization <strong>of</strong><br />

prostate cancer: correlation <strong>of</strong> (11)C-choline <strong>PET</strong>-<strong>CT</strong> with histopathologic step-section <strong>analysis</strong>. J<br />

Nucl Med 2005; Vol. 46 (10): pp 1642–1649. (absence <strong>of</strong> comparative diagnostic test)<br />

85. Ferris RL, Branstetter BF, Nayak JV. Diagnostic utility <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography-computed<br />

tomography for predicting malignancy in cystic neck masses in adults. Laryngoscope 2005; Vol. 115<br />

(11): pp 1979–1982. (casuistic description)<br />

86. Finger PT, Kuril M, Reddy S, Tena LB, Pavlick AC. Whole body <strong>PET</strong>-<strong>CT</strong> for initial staging <strong>of</strong> choroidal<br />

melanoma. Br J Ophthalmol 2005; Vol. 89 (10): pp 1270–1274. (absence <strong>of</strong> the classical comparative<br />

method)<br />

87. Finger PT, Kurli M, Wesley P, Tena LB, Kerr KR, Pavlick AC. Whole body <strong>PET</strong>-<strong>CT</strong> imaging for detection<br />

<strong>of</strong> metastatic choroidal melanoma. Br J Ophthalmol 2004; Vol. 88 (8): pp 1095–1097. (casuistic<br />

description)<br />

88. Fogarty GB, Peters LJ, Stewart J, Scott C, Rischin D, Hicks RJ. The usefulness <strong>of</strong> fluorine 18-labelled<br />

deoxyglucose <strong>positron</strong> <strong>emission</strong> tomography in the investigation <strong>of</strong> patients with cervical<br />

lymphadenopathy from an unknown primary tumor. Head & Neck 2003; Vol. 25 (2): pp 138–145.<br />

Date <strong>of</strong> Publication: 01 FEB 2003. (absence <strong>of</strong> assessed intervention)<br />

89. Francis DL, Visvikis D, <strong>Cost</strong>a DC, Arulampalam TH, Townsend C, Luthra SK, Taylor I, Ell PJ. Potential<br />

impact <strong>of</strong> [18F]3'-deoxy-3'-fluorothymidine versus [18F] fluoro-2-deoxy-D-glucose in <strong>positron</strong> <strong>emission</strong><br />

tomography for colorectal cancer. Eur J Nucl Med Mol Imaging 2003 Jul; 30 (7): pp 988–994. Epub<br />

2003 May 9. (absence <strong>of</strong> the classical comparative method)<br />

90. Frank SJ, Chao KS, Schwartz DL, Weber RS, Apisarnthanarax S, Macapinlac HA. Technology insight:<br />

<strong>PET</strong> and <strong>PET</strong>-<strong>CT</strong> in head and neck tumor staging and radiation therapy planning. Nat Clin Pract<br />

Oncol 2005; Vol. 2 (10): pp 526–533. (secondary trial)<br />

91. Freudenberg LS, Antoch G, Görges R, Knust EJ, Beyer T, Brandau W, Bockisch A, Debatin JF. 124 I-<strong>PET</strong>-<br />

<strong>CT</strong> in metastatic follicular thyroid carcinoma. Eur J Nucl Med 2002; Vol. 29: p 1106. (casuistic<br />

desription)<br />

92. Freudenberg LS, Antoch G, Görges R, Knust EJ, Beyer T, Debatin JF. Combined <strong>PET</strong>-<strong>CT</strong> with Iodine-<br />

124 in Diagnosis <strong>of</strong> Mediastinal Micrometastases in Thyroid Carcinoma. Int J Radiol 2002; Vol. 2<br />

Number 2. (casuistic desription)<br />

93. Freudenberg LS, Antoch G, Görges R, Knust EJ, Pink R, Jentzen W, Debatin JF, Brandau W, Bockisch<br />

A, Stattaus J. Combined <strong>PET</strong>-<strong>CT</strong> with iodine-124 in diagnosis <strong>of</strong> spread metastatic thyroid carcinoma:<br />

a case report. Eur Radiol 2003 Dec; 13 (Suppl 4): pp L19–23. (casuistic description)<br />

94. Freudenberg LS, Schueler AO, Beyer T, Antoch G, Kuhl H, Bornfeld N, Bockisch A, Egelh<strong>of</strong> T. Wholebody<br />

fluorine-18 fluordeoxyglucose <strong>positron</strong> <strong>emission</strong> tomography/computed tomography (FDG-<br />

<strong>PET</strong>-<strong>CT</strong>) in staging <strong>of</strong> advanced uveal melanoma. Surv Ophthalmol 2004; Vol. 49 (5): pp 537–540.<br />

(casuistic description)<br />

95. Fukui MB, Blodgett TM, Meltzer CC. <strong>PET</strong>-<strong>CT</strong> imaging in recurrent head and neck cancer. Semin<br />

Ultrasound <strong>CT</strong> MR 2003; Vol. 24 (3): pp 157–163. (secondary trial)<br />

96. Fukunaga H, Sekimoto M, Tatsumi M, Ikenaga M, Ohue M, Seshimo I, Higuchi I, Takayama O, Yasui<br />

M, Ikeda M, Yamamoto H, Monden M. Clinical relevance <strong>of</strong> fusion images using (18)F-2-fluoro-2deoxy-D-glucose<br />

<strong>positron</strong> <strong>emission</strong> tomography in local recurrence <strong>of</strong> rectal cancer. Int J Oncol<br />

2002 Apr; 20 (4): pp 691–695. (absence <strong>of</strong> assessed intervention)<br />

97. Goerres GW, Burger C, Kamel E, Seifert B, Kaim AH, Buck A, Buehler TC, von Schulthess GK.<br />

Respiration-induced attenuation artifact at <strong>PET</strong>-<strong>CT</strong>: technical considerations. Radiology 2003 Mar;<br />

226 (3): pp 906–10. (absence <strong>of</strong> end-points)<br />

98. Goerres GW, Haenggeli CA, Allaoua M, Albrecht SR, Dulguerov P, Becker M, Allal AS, Lehmann W,<br />

Slosman DO. Directcomparison <strong>of</strong>f-18-FDG <strong>PET</strong> and ultrasound in the follow-up <strong>of</strong> patients with<br />

squamous cell cancer <strong>of</strong> the head and neck. Nuklearmedizin 2000; Vol. 39 (8): pp 246–250.<br />

(absence <strong>of</strong> assessed diagnostic method)<br />

305


99. Goerres GW, Kamel E, Heidelberg T-N, Schwitter MR, Burger C, von Schulthess GK. <strong>PET</strong>-<strong>CT</strong> image coregistration<br />

in the thorax: influence <strong>of</strong> respiration. Eur J Nucl Med Mol Imaging 2002 Mar; 29 (3): pp<br />

351–360. (absence <strong>of</strong> the end-points for assessment <strong>of</strong> diagnostic efficacy)<br />

100. Goerres GW, Kamel E, Seifert B, Burger C, Buck A, Hany T, von Schulthess GK. Accuracy <strong>of</strong> image<br />

coregistration <strong>of</strong> pulmonary lesions in patients with non-small cell lung cancer using an integrated<br />

<strong>PET</strong>-<strong>CT</strong> system. J Nucl Med 2002 Nov; 43 (11): pp 1469–1575. (absence <strong>of</strong> the end-points for<br />

assessment <strong>of</strong> diagnostic efficacy)<br />

101. Goerres GW, Schmid DT, Gratz KW, von Schulthess GK, Eyrich GK. Impact <strong>of</strong> whole body <strong>positron</strong><br />

<strong>emission</strong> tomography on initial staging and therapy in patients with squamous cell carcinoma <strong>of</strong> the<br />

oral cavity. Oral Oncol 2003 Sep; 39 (6): pp 547–551. (combined assessment <strong>of</strong> the results for <strong>PET</strong> and<br />

<strong>PET</strong>-<strong>CT</strong>)<br />

102. Goerres GW, Stoeckli SJ, von Schulthess GK, Steinert HC. FDG <strong>PET</strong> for mucosal malignant melanoma<br />

<strong>of</strong> the head and neck. Laryngoscope 2002 Feb; 112 (2): pp 381–385. (absence <strong>of</strong> assessed<br />

intervention)<br />

103. Goerres GW, Stupp R, Barghouth G, Hany TF, Pestalozzi B, Dizendorf E, Schnyder P, Luthi F, von<br />

Schulthess GK, Leyvraz S. The value <strong>of</strong> <strong>PET</strong>, <strong>CT</strong> and in-line <strong>PET</strong>-<strong>CT</strong> in patients with gastrointestinal<br />

stromal tumours: long-term outcome <strong>of</strong> treatment with imatinib mesylate. Eur J Nucl Med Mol<br />

Imaging 2005 Feb; 32 (2): pp 153–162. Epub 2004 Sep 4. (absence <strong>of</strong> end-points)<br />

104. Goerres GW, von Schulthess GK, Steinert HC. Why most <strong>PET</strong> <strong>of</strong> lung and head-and-neck cancer will<br />

be <strong>PET</strong>-<strong>CT</strong>. J Nucl Med 2004 Jan; 45 Suppl 1: pp 66S-71S. (review)<br />

105. Green D, Johnson IR. Magnetic resonance imaging for gynaecological masses. Reviews in<br />

Gynaecological Practice 2004; Vol. 4 (2): pp 132–140. (secondary trial)<br />

106. Greven KM, Williams III DW, McGuirt WF Sr, Harkness BA, D'Agostino RB Jr, Keyes JW Jr, Watson NE Jr.<br />

Serial <strong>positron</strong> <strong>emission</strong> tomography scans following radiation therapy <strong>of</strong> patients with head and<br />

neck cancer. Head & Neck 2001; Vol. 23 (11): pp 942–946. (absence <strong>of</strong> assessed intervention)<br />

107. Grigsby PW, Siegel BA, Dehdashti F. Lymph node staging by <strong>positron</strong> <strong>emission</strong> tomography in<br />

patients with carcinoma <strong>of</strong> the cervix. Journal <strong>of</strong> Clinical Oncology 2002; Vol. 19 (17): pp 3745–3749.<br />

Date <strong>of</strong> Publication: 01 SEP 2001. (absence <strong>of</strong> assessed intervention)<br />

108. Grisaru D, Almog B, Levine C, Metser U, Fishman A, Lerman H, Lessing JB, Even-Sapir E. The<br />

diagnostic accuracy <strong>of</strong> 18 F-Fluorodeoxyglucose <strong>PET</strong>-<strong>CT</strong> in patients with gynecological malignancies.<br />

Gynecol Oncol 2004; Vol. 94 (3): pp 680–684.<br />

109. Grosu A-L, Lachner R, Wiedenmann N, Stark S, Thamm R, Kneschaurek P, Schwaiger M, Molls M,<br />

Weber WA. Validation <strong>of</strong> a method for automatic image fusion (BrainLAB System) <strong>of</strong> <strong>CT</strong> data and<br />

11 C-methionine-<strong>PET</strong> data for stereotactic radiotherapy using a LINAC: First clinical experience. Int J<br />

Radiat Oncol Biol Phys 2003; Vol. 56 (5): pp 1450–1463. Date <strong>of</strong> Publication: 01 AUG 2003. (absence<br />

<strong>of</strong> assessed intervention)<br />

110. Gulec SA, Beller M, Edwards K, Lizotte P, O'Day S. Case 3. Positron <strong>emission</strong> tomography-computed<br />

tomography diagnosis <strong>of</strong> metastatic melanoma with intussusception. J Clin Oncol 2004; Vol. 22 (23):<br />

pp 4802–4803. (casuistic description)<br />

111. Gutman F, Alberini JL, Wartski M, Vilain D, Le Stanc E, Sarandi F, Corone C, Tainturier C, Pecking AP.<br />

Incidental colonic focal lesions detected by FDG <strong>PET</strong>-<strong>CT</strong>. AJR Am J Roentgenol 2005 Aug; 185 (2):<br />

pp 495–500. (absence <strong>of</strong> the classical comparative method)<br />

112. Ha PK, Hdeib A, Goldenberg D, Jacene H, Patel P, Koch W, Califano J, Cummings CW, Flint PW,<br />

Wahl R, Tufano RP. The role <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography and computed tomography fusion in<br />

the management <strong>of</strong> early-stage and advanced-stage primary head and neck squamous cell<br />

carcinoma. Arch Otolaryngol Head Neck Surg 2006 Jan; 132 (1): pp 12–16. (absence <strong>of</strong> reference<br />

method)<br />

113. Haberkorn U, Strauss LG, Dimitrakopoulou A, Seiffert E, Oberdorfer F, Ziegler S, Reisser C, Doll J, Helus<br />

F, Van Kaick G. Fluorodeoxyglucose imaging <strong>of</strong> advanced head and neck cancer after<br />

chemotherapy. J Nucl Med 1993; Vol. 34 (1): pp 12–17. (absence <strong>of</strong> assessed intervention)<br />

114. Haberkorn U, Strauss LG, Reisser C, Haag D, Dimitrakopoulou A, Ziegler S, Oberdorfer F, Rudat V, Van<br />

Kaick G. Glucose uptake, perfusion and cell proliferation in head and neck tumors: relation <strong>of</strong><br />

<strong>positron</strong> <strong>emission</strong> tomography to flow cytomery. J Nucl Med 1991; Vol. 32 (8): pp 1548–1555.<br />

(absence <strong>of</strong> assessed intervention)<br />

306


115. Halfpenny W, Hain SF, Biassoni L, Maisey MN, Sherman JA, McGurk M. FDG-<strong>PET</strong>. A possible prognostic<br />

factor in head and neck cancer. Br J Cancer 2002; Vol. 86 (4): pp 512–516. Date <strong>of</strong> Publication: 12<br />

FEB 2002. (absence <strong>of</strong> assessed diagnostic method)<br />

116. Halpern BS, Schiepers C, Weber WA, Crawford TL, Fueger BJ, Phelps ME, Czernin J. Presurgical<br />

staging <strong>of</strong> non-small cell lung cancer: Positron <strong>emission</strong> tomography, integrated <strong>positron</strong> <strong>emission</strong><br />

tomography/<strong>CT</strong>, and s<strong>of</strong>tware image fusion. Chest 2005; Vol. 128 (4): pp 2289–2297. (absence <strong>of</strong><br />

comparative diagnostic method)<br />

117. Hany TF, Heuberger J, von Schulthess GK. Iatrogenic FDG foci in the lungs: a pitfall <strong>of</strong> <strong>PET</strong> image<br />

interpretation. Eur Radiol 2003 Sep; 13 (9): pp 2122–2127. Epub 2002 Oct 17. (casuistic description)<br />

118. Hany TF, Steinert HC, Goerres GW, Buck A, von Schulthess GK. <strong>PET</strong> diagnostic accuracy:<br />

improvement with in-line <strong>PET</strong>-<strong>CT</strong> system: initial results. Radiology 2002 Nov; 225 (2): pp 575–581.<br />

(absence <strong>of</strong> the classical comparative method)<br />

119. Havrilesky LJ, Kulasingam SL, Matchar DB, Myers ER. FDG-<strong>PET</strong> for management <strong>of</strong> cervical and<br />

ovarian cancer. Gynecol Oncol 2005; Vol. 97 (1): pp 183–191. (absence <strong>of</strong> assessed intervention)<br />

120. Hayashi K, Abe K, Yano F, Watanabe S, Iwasaki Y, Kosuda S. Should mediastinoscopy actually be<br />

incorporated into the FDG <strong>PET</strong> strategy for patients with non-small cell lung carcinoma?. Ann Nucl<br />

Med 2005 Jul; 19 (5): pp 393–398. (absence <strong>of</strong> assessed intervention)<br />

121. Heller MT, Meltzer C, Fukui M, Rosen C, Chander S, Martinelli M, Townsend DW. Superphysiologic FDG<br />

uptake in the non-paralyzed vocal cord: resolution <strong>of</strong> a false-positive <strong>PET</strong> result with combined <strong>PET</strong>-<br />

<strong>CT</strong> imaging. Clin Pos Imag 2000; Vol. 3 (5): pp 207–211. (casuistic description)<br />

122. Hensing TA. Clinical evaluation and staging <strong>of</strong> patients who have lung cancer. Hematol Oncol Clin<br />

N Am 2005; Vol. 19 (2): pp 219–235. (secondary trial)<br />

123. Heron DE, Andrade RS, Flickinger J, Johnson J, Agarwala SS, Wu A, Kalnicki S, Avril N. Hybrid <strong>PET</strong>-<strong>CT</strong><br />

simulation for radiation treatment planning in head-and-neck cancers: a brief technical report. Int J<br />

Radiat Oncol Biol Phys 2004 Dec 1; 60 (5): pp 1419–1424. (absence <strong>of</strong> assessed intervention)<br />

124. Hicks RJ, Mac Manus MP, Seymour JF. Initial staging <strong>of</strong> lymphoma with <strong>positron</strong> <strong>emission</strong><br />

tomography and computed tomography. Semin Nucl Med 2005 Jul; 35 (3): pp 165–175. (secondary<br />

trial)<br />

125. Ho CL. Clinical <strong>PET</strong> imaging – An asian perspective. Ann Acad Med Singapore 2004; Vol. 33 (2): pp<br />

155–165. (secondary trial)<br />

126. Ho YH, Ooi LLPJ. Recent advances in the total management <strong>of</strong> colorectal cancer. Ann Acad Med<br />

Singapore 2003; Vol. 32 (2): pp 143–144. (secondary trial)<br />

127. Hoskin PJ. <strong>PET</strong> in lymphoma: what are the oncologist's needs?. Eur J Nucl Med Mol Imaging 2003<br />

Jun; 30 (Suppl 1): pp S37–41. Epub 2003 Apr 4. (secondary trial)<br />

128. Hosten N, Lemke AJ, Wiedenmann B, Bohmig M, Rosewicz S. Combined imaging techniques for<br />

pancreatic cancer. Lancet 2000; Vol. 9; 356 (9233): pp 909–910. (secondary trial)<br />

129. Hustinx R. <strong>PET</strong> imaging in assessing gastrointestinal tumors. Radiol Clin N Am 2004; Vol. 42 (6): pp<br />

1123–1139. (secondary trial)<br />

130. Ioannidis JP, Lau J. 18 F-FDG <strong>PET</strong> for the diagnosis and grading <strong>of</strong> s<strong>of</strong>t-tissue sarcoma: a meta-<strong>analysis</strong>.<br />

J Nucl Med 2003 May; 44 (5): pp 717–724. (absence <strong>of</strong> assessed intervention)<br />

131. Ishimori T, Patel PV, Wahl RL. Detection <strong>of</strong> unexpected additional primary malignancies with <strong>PET</strong>-<strong>CT</strong>.<br />

J Nucl Med 2005; Vol. 46 (5): pp 752–757. (absence <strong>of</strong> comparative diagnostic test)<br />

132. Israel O, Mor M, Gaitini D, Keidar Z, Guralnik L, Engel A, Frenkel A, Bar-Shalom R, Kuten A. Combined<br />

functional and structural evaluation <strong>of</strong> cancer patients with a hybrid camera-based <strong>PET</strong>-<strong>CT</strong> system<br />

using 18 F-FDG. J Nucl Med 2002; Vol. 43 (9): pp 1129–1136. (absence <strong>of</strong> combined resylts for <strong>PET</strong>-<strong>CT</strong>)<br />

133. Israel O, Mor M, Guralnik L, Hermoni N, Gaitini D, Bar-Shalom R, Keidar Z, Epelbaum R. Is 18F-FDG <strong>PET</strong>-<br />

<strong>CT</strong> useful for imaging and management <strong>of</strong> patients with suspected occult recurrence <strong>of</strong> cancer. J<br />

Nucl Med 2004; Vol. 45 (12): pp 2045–2051. (absence <strong>of</strong> the classical comparative method)<br />

134. Israel O, Yefremov N, Bar-Shalom R, Kagana O, Frenkel A, Keidar Z, Fischer D. <strong>PET</strong>-<strong>CT</strong> detection <strong>of</strong><br />

unexpected gastrointestinal foci <strong>of</strong> 18F-FDG uptake: incidence, localization patterns, and clinical<br />

significance. J Nucl Med 2005 Vol. 46 (5): pp 758–762. (absence <strong>of</strong> comparative diagnostic test)<br />

307


135. Jadvar H, Alavi A, Mavi A, Shulkin BL. <strong>PET</strong> in pediatric diseases. Radiol Clin N Am 2005; Vol. 43 (1): pp<br />

135–152. (secondary trial)<br />

136. Jadvar H, Conti PS. The reproductive tract. Semin Nucl Med 2004; Vol. 34 (4): pp 262–273.<br />

(secondary trial)<br />

137. Jadvar H, Gamie S, Ramanna L, Conti PS. Musculoskeletal system. Semin Nucl Med 2004 Oct; 34 (4):<br />

pp 254–261. (secondary trial)<br />

138. Jana S, Zhang T, Milstein DM, Isasi CR, Blaufox MD. FDG-<strong>PET</strong> and <strong>CT</strong> characterization <strong>of</strong> adrenal<br />

lesions in cancer patients. Eur J Nucl Med Mol Imaging 2006; Vol. 33 (1): pp 29–35. (combined<br />

assessment <strong>of</strong> the results for <strong>PET</strong> and <strong>PET</strong>-<strong>CT</strong>)<br />

139. Jaskowiak CJ, Bianco JA, Perlman SB, Fine JP. Influence <strong>of</strong> reconstruction iterations on 18 F-FDG <strong>PET</strong>-<br />

<strong>CT</strong> standardized uptake values. J Nucl Med 2005; Vol. 46 (3): pp 424–428. (absence <strong>of</strong> comparative<br />

diagnostic test)<br />

140. Jerusalem G, Hustinx R, Beguin Y, Fillet G. The value <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography (<strong>PET</strong>) imaging<br />

in disease staging and therapy assessment. Ann Oncol 2002; Vol. 13 (Suppl. 4): pp 227–234.<br />

(secondary trial)<br />

141. Kalff V, Hicks RJ, Ware RE, Hogg A, Binns D, McKenzie AF. The clinical impact <strong>of</strong> (18)F-FDG <strong>PET</strong> in<br />

patients with suspected or confirmed recurrence <strong>of</strong> colorectal cancer: a prospective study. J Nucl<br />

Med 2002 Apr; 43 (4): pp 492–499. (absence <strong>of</strong> assessed intervention)<br />

142. Kalra MK, Maher MM, Boland GW, Saini S, Fischman AJ. Correlation <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography<br />

and <strong>CT</strong> in evaluating pancreatic tumors: Technical and clinical implications. AJR Am J Roentgenol<br />

2003; Vol. 181 (2): pp 387–393. Date <strong>of</strong> Publication: 01 AUG 2003. (secondary trial)<br />

143. Kamel EM, Goerres GW, Burger C, von Schulthess GK, Steinert HC. Recurrent laryngeal nerve palsy in<br />

patients with lung cancer: Detection with <strong>PET</strong>-<strong>CT</strong> image fusion – Report <strong>of</strong> six cases. Radiology 2002;<br />

Vol. 224 (1): 153–156. (fuzja obrazów <strong>PET</strong>-<strong>CT</strong> u 4 pacjentów)<br />

144. Kamel EM, Hany TF, Burger C, Treyer V, Lonn AHR, Von Schulthess GK, Buck A. <strong>CT</strong> vs 68 Ge attenuation<br />

correction in a combined <strong>PET</strong>-<strong>CT</strong> system: Evaluation <strong>of</strong> the effect <strong>of</strong> lowering the <strong>CT</strong> tube current.<br />

Eur J Nucl Med 2002; Vol. 29 (3): pp 346–350. (absence <strong>of</strong> comparative diagnostic test)<br />

145. Kamel EM, Thumshirn M, Truninger K, Schiesser M, Fried M, Padberg B, Schneiter D, Stoeckli SJ, von<br />

Schulthess GK, Stumpe KD. Significance <strong>of</strong> incidental 18F-FDG accumulations in the gastrointestinal<br />

tract in <strong>PET</strong>-<strong>CT</strong>: correlation with endoscopic and histopathologic results. J Nucl Med 2004 Nov; 45<br />

(11): pp 1804–1810. (absence <strong>of</strong> the classical comparative method)<br />

146. Kamel EM, Zwahlen D, Wyss MT, Stumpe KD, von Schulthess GK, Steinert HC. Whole-body 18 F-FDG <strong>PET</strong><br />

improves the management <strong>of</strong> patients with small cell lung cancer. Source. J Nucl Med 2003; Vol. 44<br />

(12): pp 1911–1917. (absence <strong>of</strong> assessed intervention)<br />

147. Kamel IR, Cohade C, Neyman E, Fishman EK, Wahl RL. Incremental value <strong>of</strong> <strong>CT</strong> in <strong>PET</strong>-<strong>CT</strong> <strong>of</strong> patients<br />

with colorectal carcinoma. Abdominal Imaging 2004; Vol. 29 (6): pp 663–668. (absence <strong>of</strong> the<br />

classical comparative method)<br />

148. Kapoor V, Fukui MB, McCook BM. Role <strong>of</strong> 18FFDG <strong>PET</strong>-<strong>CT</strong> in the treatment <strong>of</strong> head and neck<br />

cancers: principles, technique, normal distribution, and initial staging. AJR Am J Roentgenol 2005<br />

Feb; 184 (2): pp 579–587. (secondary trial)<br />

149. Kaste SC. Issues specific to implementing <strong>PET</strong>-<strong>CT</strong> for pediatric oncology: What we have learned<br />

along the way. Pediatr Radiol 2004; Vol. 34 (3): pp 205–213. (secondary trial)<br />

150. Kato H, Miyazaki T, Nakajima M, Fukuchi M, Manda R, Kuwano H. Value <strong>of</strong> <strong>positron</strong> <strong>emission</strong><br />

tomography in the diagnosis <strong>of</strong> recurrent oesophageal carcinoma. Br J Surg 2004; Vol. 91 (8): pp<br />

1004–1009. (absence <strong>of</strong> assessed intervention)<br />

151. Kato H, Miyazaki T, Nakajima M, Takita J, Kimura H, Faried A, Sohda M, Fukai Y, Masuda N, Fukuchi<br />

M, Manda R, Ojima H, Tsukada K, Kuwano H, Oriuchi N, Endo K. The incremental effect <strong>of</strong> <strong>positron</strong><br />

<strong>emission</strong> tomography on diagnostic accuracy in the initial staging <strong>of</strong> esophageal carcinoma.<br />

Cancer 2005; Vol. 103 (1): pp 148–156. (absence <strong>of</strong> assessed intervention)<br />

152. Katzel JA, Heiba SI. <strong>PET</strong>-<strong>CT</strong> F-18 FDG scan accurately identifies osteoporotic fractures in a patient<br />

with known metastatic colorectal cancer. Clin Nucl Med 2005; Vol. 30 (10): pp 651–654. (casuistic<br />

description)<br />

308


153. Kayani I, Groves AM, Ell PJ, George PJ, Bomanji J. Imaging bronchial carcinoma in situ: possible roles<br />

for combined <strong>positron</strong> <strong>emission</strong> tomography (<strong>PET</strong>)-<strong>CT</strong>. Lancet Oncol 2005 Mar; 6 (3): p 190. (casuistic<br />

description)<br />

154. Kayani I, Groves AM, Syed R, Bomanji J. Combined F-18 FDG <strong>positron</strong> <strong>emission</strong> tomography<br />

/computed tomography in the diagnosis <strong>of</strong> colonic polyps: The potential and limitations <strong>of</strong> the<br />

technique. Clin Nucl Med 2005; Vol. 30 (2): pp 116–117. (casuistic description)<br />

155. Keidar Z, Haim N, Guralnik L, Wollner M, Bar-Shalom R, Ben-Nun A, Israel O. <strong>PET</strong>-<strong>CT</strong> using 18F-FDG in<br />

suspected lung cancer recurrence: diagnostic value and impact on patient management. J Nucl<br />

Med 2004 Oct; 45 (10): pp 1640–1646. (absence <strong>of</strong> comparative diagnostic test)<br />

156. Keyes JW, Chen MYM, Watson NE, Greven KM, McGuirt WF, Williams III DW. FDG pet evaluation <strong>of</strong><br />

head and neck cancer: Value <strong>of</strong> imaging the thorax. Head & Neck 2000; Vol. 22 (2): pp 105–110.<br />

(absence <strong>of</strong> assessed intervention)<br />

157. Kim J-H, Czernin J, Allen-Auerbach MS, Halpern BS, Fueger BJ, Hecht JR, Ratib O, Phelps ME, Weber<br />

WA. Comparison between 18 F-FDG <strong>PET</strong>, in-line <strong>PET</strong>-<strong>CT</strong>, and s<strong>of</strong>tware fusion for restaging <strong>of</strong> recurrent<br />

colorectal cancer. J Nucl Med 2005; Vol. 46 (4): pp 587–595. (absence <strong>of</strong> the classical comparative<br />

method)<br />

158. Kinahan P, Townsend D, Beyer T, Sashin D. Attenuation correction for a combined 3D <strong>PET</strong>-<strong>CT</strong><br />

scanner. Med Phys 1998; Vol. 25 (10): pp 2046–2053. (absence <strong>of</strong> assessed intervention)<br />

159. Kluetz PG, Meltzer CC, Villemagne VL, Kinahan PE, Chander S, Martinelli MA, Townsend DW.<br />

Combined <strong>PET</strong>-<strong>CT</strong> Imaging in Oncology: Impact on Patient Management. Clin Positron Imaging<br />

2000; Vol. 3, No. 6: pp 223–230. (casuistic description)<br />

160. Kole AC, Plukker JT, Nieweg OE, Vaalburg W. Positron <strong>emission</strong> tomography for staging <strong>of</strong><br />

oesophageal and gastroesophageal malignancy. Br J Cancer 1998 Aug; 78 (4): pp 521–527.<br />

(absence <strong>of</strong> assessed intervention)<br />

161. Konski A, Doss M, Milestone B, Haluszka O, Hanlon A, Freedman G, Adler L. The integration <strong>of</strong> 18fluoro-deoxy-glucose<br />

<strong>positron</strong> <strong>emission</strong> tomography and endoscopic ultrasound in the treatmentplanning<br />

process for esophageal carcinoma. Int J Radiat Oncol Biol Phys 2005; Vol. 61 (4): pp 1123–<br />

1128. (absence <strong>of</strong> assessed intervention)<br />

162. Kostakoglu L, Goldsmith SJ. 18F-FDG <strong>PET</strong> evaluation <strong>of</strong> the response to therapy for lymphoma and<br />

for breast, lung, and colorectal carcinoma. J Nucl Med 2003 Feb; 44 (2): pp 224–239. (secondary<br />

trial)<br />

163. Koyama K, Okamura T, Kawabe J, Ozawa N, Torii K, Umesaki N, Miyama M, Ochi H, Yamada R.<br />

Evaluation <strong>of</strong> 18 F-FDG <strong>PET</strong> with bladder irrigation in patients with uterine and ovarian tumors. J Nucl<br />

Med 2003; Vol. 44 (3): pp 353–358. (absence <strong>of</strong> assessed intervention)<br />

164. Kresnik E, Kogler D, Gallowitsch H-J, Mikosch P, Wieser S, Igerc I, Gomez I, Kumnig G, Lind P. Head<br />

and neck tumors. Current evaluation with 18 F-FDG-<strong>PET</strong> compared with conventional methods.<br />

Internistische Praxis 2003; Vol. 43 (2): pp 301–309. (article in German)<br />

165. Krishnasetty V, Fischman AJ, Halpern EL, Aquino SL. Comparison <strong>of</strong> alignment <strong>of</strong> computerregistered<br />

data sets: combined <strong>PET</strong>-<strong>CT</strong> versus independent <strong>PET</strong> and <strong>CT</strong> <strong>of</strong> the thorax. Radiology<br />

2005; Vol. 237 (2): pp 635–639. (inadequate population)<br />

166. Kuhnel G, Horn L-C, Fischer U, Hesse S, Seese A, Georgi P, Kluge R. 18 F-FDG-Positron-Emission-<br />

Tomography in patients with uterine cervical cancer: Preliminary results. Zentralblatt fur Gynakologie<br />

2001; Vol. 123 (4): pp 229–235. (article in German)<br />

167. Kula Z, Szefer J, Pietrzak T, Zuchora Z. An attempt at assessing the efficacy <strong>of</strong> combined <strong>positron</strong><br />

emision tomography and computed tomography (<strong>PET</strong>-<strong>CT</strong>) imaging in the diagnosis <strong>of</strong> pancreatic<br />

carcinoma – Own experiences. Nowotwory 2005; Vol. 55 (5): pp 373–379. (absence <strong>of</strong> the<br />

unambiguous results)<br />

168. Kula Z, Szefer J, Pietrzak T, Zuchora Z. Our first experience with combined <strong>positron</strong> <strong>emission</strong><br />

tomography and computed tomography (<strong>PET</strong>-<strong>CT</strong>) in patients with gastric cancer. Współczesna<br />

Onkologia 2005; Vol. 9 (4): pp 213–217. (absence <strong>of</strong> assessed intervention)<br />

169. Kumar R, Alavi A. <strong>PET</strong> imaging in gynecologic malignancies. Radiol Clin N Am 2004; Vol. 42 (6): pp<br />

1155–1167. (secondary trial)<br />

170. Kunkler I. Cure, palliation, and cost in cancer care. Lancet Oncol 2004; Vol. 5 (12): p 709.<br />

(secondary trial)<br />

309


171. Kurli M, Reddy S, Tena LB, Pavlick AC, Finger PT. Whole body <strong>positron</strong> <strong>emission</strong><br />

tomography/computed tomography staging <strong>of</strong> metastatic choroidal melanoma. Am J Ophthalmol<br />

2005 Aug; 140 (2): pp 193–199. (absence <strong>of</strong> the results in application <strong>of</strong> reference method)<br />

172. Kwon RS, Sahani DV, Brugge WR. Gastrointestinal cancer imaging: Deeper than the eye can see.<br />

Gastroenterology 2005; Vol. 128 (6): pp 1538–1553. (secondary trial)<br />

173. Ladabaum U. Positron <strong>emission</strong> tomography: The gastroenterologists's perspective. Gastrointestinal<br />

Endoscopy 2002; Vol. 55 (7 II): pp S64–S66. (secondary trial)<br />

174. Lamoreaux WT, Grigsby PW, Dehdashti FD, Zoberi I, Powell MA, Gibb RK, Rader JS, Mutch DG, Siegel<br />

BA. FDG-<strong>PET</strong> evaluation <strong>of</strong> vaginal carcinoma. Int J Radiat Oncol Biol Phys 2005; Vol. 62 (3): pp 733–<br />

737. (absence <strong>of</strong> separated results for <strong>PET</strong> and <strong>PET</strong>-<strong>CT</strong>)<br />

175. Lardinois D, Weder W, Roudas M, von Schulthess GK, Tutic M, Moch H, Stahel RA, Steinert HC.<br />

Etiology <strong>of</strong> solitary extrapulmonary <strong>positron</strong> <strong>emission</strong> tomography and computed tomography<br />

findings in patients with lung cancer. J Clin Oncol 2005 Oct 1; 23 (28): pp 6846–6853. (absence <strong>of</strong><br />

comparative diagnostic test)<br />

176. Larson SM, Nehmeh SA, Erdi YE, Humm JL. <strong>PET</strong>-<strong>CT</strong> in non-small-cell lung cancer: Value <strong>of</strong> respiratorygated<br />

<strong>PET</strong>. Chang Gung Med J 2005; Vol. 28 (5): pp 306–314. (secondary trial absence <strong>of</strong> assessed<br />

intervention)<br />

177. Lau WF, Binns DS, Ware RE, Ramdave S, Cachin F, Pitman AG, Hicks RJ. Clinical experience with the<br />

first combined <strong>positron</strong> <strong>emission</strong> tomography/computed tomography scanner in Australia. Med J<br />

Aust 2005; Vol. 21; 182 (4): pp172–176. (secondary trial)<br />

178. Lavrenkov K, Partridge M, Cook G, Brada M. Positron <strong>emission</strong> tomography for target volume<br />

definition in the treatment <strong>of</strong> non-small cell lung cancer. Radiother Oncol 2005; Vol. 77 (1): pp 1–4.<br />

(secondary trial)<br />

179. Lejeune C, Bismuth MJ, Conroy T, Zanni C, Bey P, Bedenne L, Faivre J, Arveux P, Guillemin F. Use <strong>of</strong><br />

a decision <strong>analysis</strong> model to assess the cost-<strong>effectiveness</strong> <strong>of</strong> 18F-FDG <strong>PET</strong> in the management <strong>of</strong><br />

metachronous liver metastases <strong>of</strong> colorectal cancer. J Nucl Med 2005 Dec; 46 (12): pp 2020–2028.<br />

(secondary trial)<br />

180. Lerman H, Metser U, Grisaru D, Fishman A, Lievshitz G, Even-Sapir E. Normal and abnormal 18F-FDG<br />

endometrial and ovarian uptake in pre- and postmenopausal patients: assessment by <strong>PET</strong>-<strong>CT</strong>.<br />

J Nucl Med 2004 Feb; 45 (2): pp 266–271. (absence <strong>of</strong> the classical method and end-points <strong>of</strong><br />

diagnostic efficacy assessment)<br />

181. Leskinen-Kallio S, Lindholm P, Lapela M, Joensuu H, Nordman E. Imaging <strong>of</strong> head and neck tumors<br />

with <strong>positron</strong> <strong>emission</strong> tomography and [ 11 C]methionine. Int J Radiat Oncol Biol Phys 1994; Vol. 30<br />

(5): pp 1195–1199. (absence <strong>of</strong> assessed diagnostic method)<br />

182. Lim JWM, Tang CL, Keng GHW. False positive F-18 fluorodeoxyglucose combined <strong>PET</strong>-<strong>CT</strong> scans from<br />

suture granuloma and chronic inflammation: Report <strong>of</strong> two cases and review <strong>of</strong> literature. Ann<br />

Acad Med Singapore 2005; Vol. 34 (7): pp 457–460. (casuistic description)<br />

183. Lind P, Igerc I, Beyer T, Reinprecht P, Hausegger K. Advantages and limitations <strong>of</strong> FDG <strong>PET</strong> in the<br />

follow-up <strong>of</strong> breast cancer. Eur J Nucl Med Mol Imaging 2004 Jun; 31 (Suppl 1): pp S125–134. Epub<br />

2004 Apr 15. (secondary trial)<br />

184. Lind P, Kumnig G, Heinisch M, Gallowitsch HJ, Mikosch P, Kresnik E, Gomez I, Unterweger O. F-18-FDG<br />

<strong>positron</strong> <strong>emission</strong> tomography in oncology. Tagliche Praxis 2003; Vol. 44 (3): pp 503–520. (article in<br />

German)<br />

185. Lowe VJ, Dunphy FR, Varvares M, Kim H, Wittry M, Dunphy CH, Dunleavy T, McDonough E, Minster J,<br />

Fletcher JW, Boyd JH. Evaluation <strong>of</strong> chemotherapy response in patients with advanced head and<br />

neck cancer using [F-18] fluorodeoxyglucose <strong>positron</strong> <strong>emission</strong> tomography. Head & Neck 1997;<br />

Vol. 19 (8): pp 666–674. (absence <strong>of</strong> assessed intervention)<br />

186. Lytras D, Connor S, Bosonnet L, Jayan R, Evans J, Hughes M, Garvey CJ, Ghaneh P, Sutton R,<br />

Vinjamuri S, Neoptolemos JP. Positron <strong>emission</strong> tomography does not add to computed<br />

tomography for the diagnosis and staging <strong>of</strong> pancreatic cancer. Dig Surg 2005; Vol. 22 (1–2): pp 55–<br />

61. (absence <strong>of</strong> assessed intervention)<br />

187. Ma S-Y, See L-C, Lai C-H, Chou H-H, Tsai C-S, Ng K-K, Hsueh S, Lin W-J, Chen J-T, Yen T-C. Delayed 18 F-<br />

FDG <strong>PET</strong> for detection <strong>of</strong> paraaortic lymph node metastases in cervical cancer patients. J Nucl Med<br />

2003; Vol. 44 (11): pp 1775–1783. (absence <strong>of</strong> assessed intervention)<br />

310


188. Macapinlac HA. FDG <strong>PET</strong> and <strong>PET</strong>-<strong>CT</strong> imaging in lymphoma and melanoma. Cancer J 2004 Jul-Aug;<br />

10 (4): pp 262–270. (secondary trial)<br />

189. Macapinlac HA. FDG-<strong>PET</strong> in the evaluation <strong>of</strong> cervical cancer. Gynecol Oncol 2005; Vol. 99<br />

(3 Suppl.): pp S171–S172. (secondary trial)<br />

190. Macapinlac HA. The utility <strong>of</strong> 2-deoxy-2-[18F]fluoro-D-glucose-<strong>positron</strong> <strong>emission</strong> tomography and<br />

combined <strong>positron</strong> <strong>emission</strong> tomography and computed tomography in lymphoma and<br />

melanoma. Mol Imaging Biol 2004 Jul-Aug; 6 (4): pp 200–207. (secondary trial)<br />

191. Magnani P, Carretta A, Rizzo G, Fazio F, Vanzulli A, Lucignani G, Zannini P. FDG/<strong>PET</strong> and spiral <strong>CT</strong><br />

image fusion for medistinal lymph node assessment <strong>of</strong> patients with small cell lung cancer. J<br />

Cardiovasc Surg (Torino) 1999 Oct; 40 (5): pp 741–748. (absence <strong>of</strong> assessed intervention)<br />

192. Mahoney EJ, Spiegel JH. Evaluation and management <strong>of</strong> malignant cervical lymphadenopathy<br />

with an unknown primary tumor. Otolaryngol Clin N Am 2005; Vol. 38 (1): pp 87–97. (secondary trial)<br />

193. Manavis J, Sivridis L, Koukourakis MI. Nasopharyngeal carcinoma: the impact <strong>of</strong> <strong>CT</strong>-scan and <strong>of</strong> MRI<br />

on staging, radiotherapy treatment planning, and outcome <strong>of</strong> the disease. Clin Imaging 2005 Mar-<br />

Apr; 29 (2): pp 128–133. (secondary trial)<br />

194. Martinez-Roman S, Ramirez PT, Oh J, Viciedo MG, MacApinlac HA. Combined <strong>positron</strong> <strong>emission</strong><br />

tomography and computed tomography for the detection <strong>of</strong> recurrent ovarian mucinous<br />

adenocarcinoma. Gynecol Oncol 2005; Vol. 96 (3): pp 888–891. (absence <strong>of</strong> assessed intervention)<br />

195. Mattes D, Haynor DR, Vesselle H, Lewellen TK, Eubank W. <strong>PET</strong>-<strong>CT</strong> image registration in the chest using<br />

free-form deformations. IEEE Trans Med Imaging 2003 Jan; 22 (1): pp 120–128. (absence <strong>of</strong> endpoints)<br />

196. Matthies A, Ezziddin S, Ulrich E-M, Palmedo H, Biersack H-J, Bender H, Guhlke S. Imaging <strong>of</strong> prostate<br />

cancer metastases with 18 F-fluoroacetate using <strong>PET</strong>/ <strong>CT</strong>. Eur J Nucl Med Mol Imaging 2004; Vol. 31<br />

(5): p 797. (absence <strong>of</strong> assessed intervention)<br />

197. McCarville MB, Christie R, Daw NC, Spunt SL, Kaste SC. <strong>PET</strong>-<strong>CT</strong> in the evaluation <strong>of</strong> childhood<br />

sarcomas. AJR Am J Roentgenol 2005 Apr; 184 (4): pp 1293–1304. (casuistic description)<br />

198. McCollum AD, Burrell SC, Haddad RI, Norris CM, Tishler RB, Case MA, Posner MR, Van Den Abbeele<br />

AD. Positron <strong>emission</strong> tomography with 18 F-fluorodeoxyglucose to predict pathologic response after<br />

induction chemotherapy and definitive chemoradiotherapy in head and neck cancer. Head &<br />

Neck 2004; Vol. 26 (10): pp 890–896. (absence <strong>of</strong> assessed intervention)<br />

199. Menda Y, Graham MM. Update on 18F-fluorodeoxyglucose/<strong>positron</strong> <strong>emission</strong> tomography and<br />

<strong>positron</strong> <strong>emission</strong> tomography/computed tomography imaging <strong>of</strong> squamous head and neck<br />

cancers. Semin Nucl Med 2005; Vol. 35 (4): pp 214–219. (secondary trial)<br />

200. Metser U, Golan O, Levine CD, Even-Sapir E. Tumor lesion detection: When is integrated <strong>positron</strong><br />

<strong>emission</strong> tomography/computed tomography more accurate than side-by-side interpretation <strong>of</strong><br />

<strong>positron</strong> <strong>emission</strong> tomography and computed tomography? J Comput Assist Tomogr 2005; Vol. 29<br />

(4): pp 554–559. (<strong>PET</strong>-<strong>CT</strong> compared to <strong>PET</strong>)<br />

201. Metser U, Goor O, Lerman H, Naparstek E, Even-Sapir E. <strong>PET</strong>-<strong>CT</strong> <strong>of</strong> extranodal lymphoma. AJR Am J<br />

Roentgenol 2004 Jun; 182 (6): pp 1579–1586. (secondary trial)<br />

202. Metser U, Lerman H, Blank A, Lievshitz G, Bokstein F, Even-Sapir E. Malignant involvement <strong>of</strong> the<br />

spine: assessment by 18F-FDG <strong>PET</strong>-<strong>CT</strong>. J Nucl Med 2004; Vol. 45 (2): pp 279–284. (absence <strong>of</strong> results<br />

for the reference methods)<br />

203. Metser U, Miller E, Kessler A, Lerman H, Lievshitz G, Oren R, Even-Sapir E. Solid splenic masses:<br />

Evaluation with 18 F-FDG <strong>PET</strong>-<strong>CT</strong>. J Nucl Med 2005; Vol. 46 (1): pp 52–59. (absence <strong>of</strong> comparative<br />

diagnostic test)<br />

204. Miller E, Lerman H, Gutman M, Figer A, Livshitz G, Even-Sapir E. The clinical impact <strong>of</strong> camera-based<br />

<strong>positron</strong> <strong>emission</strong> tomography imaging in patients with recurrent colorectal cancer. Invest Radiol<br />

2004 Jan; 39 (1): pp 8–12. (absence <strong>of</strong> assessed intervention)<br />

205. Mitchell JC, Grant F, Evenson AR, Parker JA, Hasselgren PO, Parangi S. Preoperative evaluation <strong>of</strong><br />

thyroid nodules with 18FDG-<strong>PET</strong>-<strong>CT</strong>. Surgery 2005 Dec; 138 (6): pp 1166–1174; discussion: pp 1174–<br />

1175. (absence <strong>of</strong> the classical comparative method)<br />

206. Moadel RM, Blaufox MD, Freeman LM. The role <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography in gastrointestinal<br />

imaging. Gastroenterol Clin N Am 2002; Vol. 31 (3): pp 841–861. (secondary trial)<br />

311


207. Modi D, Fulham MJ, Mohamed A, McCaughan BC. Intraluminal FDG uptake in a rectal polyp<br />

detected with <strong>PET</strong> <strong>CT</strong>: Identification <strong>of</strong> an unsuspected synchronous primary bowel tumor. Clin Nucl<br />

Med 2005; Vol. 30 (3): pp 180–181. (casuistic description)<br />

208. Moureau-Zabotto L, Touboul E, Lerouge D, Deniaud-Alexandre E, Grahek D, Foulquier J-N, Petegnief<br />

Y, Gres B, El Balaa H, Kerrou K, Montravers F, Keraudy K, Tiret E, Gendre J-P, Grange J-D, Houry S,<br />

Talbot J-N. Impact <strong>of</strong> <strong>CT</strong> and 18 F-deoxyglucose <strong>positron</strong> <strong>emission</strong> tomography image fusion for<br />

conformal radiotherapy in esophageal carcinoma. Int J Radiat Oncol Biol Phys 2005; Vol. 63 (2): pp<br />

340–345. (absence <strong>of</strong> assessed intervention)<br />

209. Mukherji SK, Wolf GT. Evaluation <strong>of</strong> head and neck squamous cell carcinoma after treatment. AJNR<br />

Am J Neuroradiol 2003; Vol. 24: pp 1743–1746. (secondary trial)<br />

210. Mumber M, Greven K, Haygood T. Pelvic insufficiency fractures associated with radiation atrophy:<br />

clinical recognition and diagnostic evaluation. Skeletal Radiol 1997 Feb; 26 (2): pp 94–99. (casuistic<br />

description)<br />

211. Nakahara T, Fujii H, Ide M, Mochizuki Y, Takahashi W, Yasuda S, Shohtsu A, Kubo A. F-18 FDG uptake<br />

in endometrial cancer. Clin Nucl Med 2001 Jan; 26 (1): pp 82–83. (casuistic description)<br />

212. Nakamoto Y, Cohade C, Tatsumi M, Hammoud D, Wahl RL. <strong>CT</strong> appearance <strong>of</strong> bone metastases<br />

detected with FDG <strong>PET</strong> as part <strong>of</strong> the same <strong>PET</strong>-<strong>CT</strong> examination. Radiology 2005; Vol. 237 (2): pp<br />

627–634. (absence <strong>of</strong> assessed intervention)<br />

213. Nakamoto Y, Osman M, Cohade C, Marshall LT, Links JM, Kohlmyer S, Wahl RD. <strong>PET</strong>-<strong>CT</strong>: comparison<br />

<strong>of</strong> quantitative tracer uptake between germanium and <strong>CT</strong> transmission attenuation-corrected<br />

images. J Nucl Med 2002 Sep; 43 (9): pp 1137–1143. (absence <strong>of</strong> the clinical anf diagnostic<br />

assessment, absence <strong>of</strong> reference method)<br />

214. Nanni C, Rubello D, Castellucci P, Farsad M, Franchi R, Toso S, Barile C, Rampin L, Nibale O, Fanti S.<br />

Role <strong>of</strong> 18 F-FDG <strong>PET</strong>-<strong>CT</strong> imaging for the detection <strong>of</strong> an unknown primary tumour: Preliminary results<br />

in 21 patients. Eur J Nucl Med Mol Imaging 2005; Vol. 32 (5): pp 589–592. (absence <strong>of</strong> the classical<br />

comparative method)<br />

215. Nanni C, Rubello D, Farsad M, De Iaco P, Sanovivini M, Erba P, Rampin L, Mariani G, Fanti S. FDG-<strong>PET</strong><br />

<strong>CT</strong> in evaluation <strong>of</strong> recurrent ovarian cancer: a prospective study on forty one patients. Eur J Surg<br />

Oncol 2005 Sep; 31 (7): pp 792–797. (absence <strong>of</strong> comparative diagnostic method)<br />

216. Nehmeh SA, Erdi YE, Pan T, Pevsner A, Rosenzweig KE, Yorke E, Mageras GS, Schoder H, Vernon P,<br />

Squire O, Mostafavi H, Larson SM, Humm JL. Four-dimensional (4D) <strong>PET</strong>-<strong>CT</strong> imaging <strong>of</strong> the thorax.<br />

Medical Physics 2004; Vol. 31 (12): pp 3179–3186. (secondary trial)<br />

217. Nehmeh SA, Erdi YE, Pan T, Yorke E, Mageras GS, Rosenzweig KE, Schoder H, Mostafavi H, Squire O,<br />

Pevsner A, Larson SM, Humm JL. Quantitation <strong>of</strong> respiratory motion during 4D-<strong>PET</strong>-<strong>CT</strong> acquisition.<br />

Med Phys 2004 Jun; 31 (6): pp 1333–1338. (absence <strong>of</strong> end-points for assessment <strong>of</strong> diagnostic<br />

efficacy)<br />

218. Nestle U, Hellwig D, Fleckenstein J, Walter K, Ukena D, Rube C, Kirsch CM, Baumann M. Comparison<br />

<strong>of</strong> early pulmonary changes in 18FDG-<strong>PET</strong> and <strong>CT</strong> after combined radiochemotherapy for<br />

advanced non-small-cell lung cancer: a study in 15 patients. Front Radiat Ther Oncol 2002; 37: pp<br />

26–33. (absence <strong>of</strong> assessed intervention)<br />

219. Nguyen BD, Fletcher GP, Patel AC. <strong>PET</strong>-<strong>CT</strong> imaging <strong>of</strong> conus medullaris metastasis from lung cancer.<br />

Clin Nucl Med 2005; Vol. 30 (4): pp 253–256. (casuistic description)<br />

220. Nomayr A, Romer W, Hothorn T, Pfahlberg A, Hornegger J, Bautz W, Kuweit T. Anatomical accuracy<br />

<strong>of</strong> lesion localization: Retrospective interactive rigid image registration between 18 F-FDG-<strong>PET</strong> and Xray<br />

<strong>CT</strong>. Nuklearmedizin 2005; Vol. 44 (4): pp 149–155. (absence <strong>of</strong> assessed intervention)<br />

221. Okada J, Yoshikawa K, Imazeki K, Minoshima S, Uno K, Itami J, Kuyama J, Maruno H, Arimizu N. The<br />

use <strong>of</strong> FDG-<strong>PET</strong> in the detection and management <strong>of</strong> malignant lymphoma: Correlation <strong>of</strong> uptake<br />

with prognosis. J Nucl Med 1991; Vol. 32 (4): pp 686–691. (absence <strong>of</strong> assessed intervention)<br />

222. Ollenberger GP. Staging <strong>of</strong> lung cancer with integrated <strong>PET</strong> and <strong>CT</strong>. N Engl J Med 2004 Jan 1; 350<br />

(1): pp 86–87; author reply 86–87. (letter)<br />

223. Ong SC, Ng DC, Sundram FX. Initial experience in use <strong>of</strong> fluorine-18-fluorodeoxyglucose <strong>positron</strong><br />

<strong>emission</strong> tomography/computed tomography in thyroid carcinoma patients with elevated serum<br />

thyroglobulin but negative iodine-131 whole body scans. Singapore Med J 2005 Jun; 46 (6): pp 297–<br />

301. (absence <strong>of</strong> verification <strong>of</strong> study results in application <strong>of</strong> reference test)<br />

312


224. Osman MM, Cohade C, Fishman EK, Wahl RL. Clinically significant incidental findings on the<br />

unenhanced <strong>CT</strong> portion <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> studies: frequency in 250 patients. J Nucl Med 2005; Vol. 46 (8): pp<br />

1352–1355. (absence <strong>of</strong> reference test)<br />

225. Osman MM, Cohade C, Nakamoto Y, Marshall L, Leal J, Wahl R. Clinically significant inaccurate<br />

localization <strong>of</strong> lesions with <strong>PET</strong>-<strong>CT</strong>: frequency in 300 patients. J Nucl Med 2003 Feb; 44 (2): pp 240–<br />

243. (absence <strong>of</strong> the classical comparative method)<br />

226. Osman MM, Cohade C, Nakamoto Y, Wahl RL. Respiratory motion artifacts on <strong>PET</strong> <strong>emission</strong> images<br />

obtained using <strong>CT</strong> attenuation correction on <strong>PET</strong>-<strong>CT</strong>. Eur J Nucl Med Mol Imaging 2003 Apr; 30 (4):<br />

pp 603–606. Epub 2003 Jan 21. (absence <strong>of</strong> end-points for assessment <strong>of</strong> diagnostic efficacy)<br />

227. Otsuka H, Graham MM, Kogame M, Nishitani H. The impact <strong>of</strong> FDG-<strong>PET</strong> in the management <strong>of</strong><br />

patients with salivary gland malignancy. Ann Nucl Med 2005; Vol. 19 (8): pp 691–694. (absence <strong>of</strong><br />

assessed diagnostic method)<br />

228. Oudoux A, Rousseau T, Bridji B, Resche I, Rousseau C. Interest <strong>of</strong> F-18 fluorodeoxyglucose <strong>positron</strong><br />

<strong>emission</strong> tomography in the evaluation <strong>of</strong> vaginal malignant melanoma. Gynecol Oncol 2004; Vol.<br />

95 (3): pp 765–768. (casuistic description)<br />

229. Pacak K, Ilias I, Chen CC, Carrasquillo JA, Whatley M, Nieman LK. The Role <strong>of</strong> [ 18 F]<br />

Fluorodeoxyglucose Positron Emission Tomography and [ 11 In]-Diethylenetriaminepentaacetate-D-<br />

Phe-Pentetreotide Scintigraphy in the Localization <strong>of</strong> Ectopic Adrenocorticotropin-Secreting Tumors<br />

Causing Cushing's Syndrome. J Clin Endocrinol Metab 2004; Vol. 89 (5): pp 2214–2221. (absence <strong>of</strong><br />

assessed intervention)<br />

230. Pannu HK, Cohade C, Bristow RE, Fshman EK, Wahl RL. <strong>PET</strong>-<strong>CT</strong> detection <strong>of</strong> abdominal recurence <strong>of</strong><br />

ovarian cancer: radiologic-surgical correlation. Abdom Imaging 2004 May-Jun; 29 (3): pp 398–403.<br />

(absence <strong>of</strong> comparative diagnostic method)<br />

231. Pardo FS, Aronen HJ, Fitzek M, Kennedy DN, Efird J, Rosen BR, Fischman AJ. Correlation <strong>of</strong> FDG-<strong>PET</strong><br />

interpretation with survival in a cohort <strong>of</strong> glioma patients. Anticancer Research 2004; Vol. 24 (4): pp<br />

2359–2365. (absence <strong>of</strong> assessed intervention)<br />

232. Park D, Kim K, Park S, Lee B, Choi C, Chin S. Diagnosis <strong>of</strong> recurrent uterine cervical cancer:<br />

computed tomography versus <strong>positron</strong> <strong>emission</strong> tomography. Korean J Radiol 2000 Jan-Mar; 1 (1):<br />

pp 51–55. (absence <strong>of</strong> assessed intervention)<br />

233. Patel PV, Cohade C, Chin BB. <strong>PET</strong>-<strong>CT</strong> localizes previously undetectable metastatic lesions in<br />

recurrent fallopian tube carcinoma. Gynecol Oncol 2002; Vol. 87: pp 323–326. (casuistic<br />

description)<br />

234. Paulino AC, Koshy M, Howell R, Schuster D, Davis LW. Comparison <strong>of</strong> <strong>CT</strong>- and FDG-<strong>PET</strong>-defined gross<br />

tumor volume in intensity-modulated radiotherapy for head-and-neck cancer. Int J Radiat Oncol<br />

Biol Phys 2005 Apr 1; 61 (5): pp 1385–1392. (absence <strong>of</strong> the classical comparative method)<br />

235. Pelosi E, Messa C, Sironi S, Picchio M, Landoni C, Bettinardi V, Gianolli L, Del Maschio A, Gilardi MC,<br />

Fazio F. Value <strong>of</strong> integrated <strong>PET</strong>-<strong>CT</strong> for lesion localisation in cancer patients: A comparative study.<br />

Eur J Nucl Med Mol Imaging 2004; Vol. 31 (7): pp 932–939. (absence <strong>of</strong> the classical comparative<br />

method)<br />

236. Perrotin C, Lemeunier P, Grahek D, Molina T, Petino A, Alifano M, Bellenot F, Magdeleinat P, Talbot J-<br />

N, Regnard J-F. Results <strong>of</strong> FDG-<strong>PET</strong> scanning in the pre-operative staging <strong>of</strong> broncho-pulmonary<br />

tumours. Revue des Maladies Respiratoires 2005; Vol. 22 (4): pp 579–585. (article in French)<br />

237. Phongkitkarun S, Varavithya V, Kazama T, Faria SC, Mar MV, Podol<strong>of</strong>f DA, Macapinlac HA.<br />

Lymphomatous involvement <strong>of</strong> gastrointestinal tract: Evaluation by <strong>positron</strong> <strong>emission</strong> tomography<br />

with 18 F-fluorodeoxyglucose. World J Gastroenterol 2005; Vol. 11 (46): pp 7284–7289. Date <strong>of</strong><br />

Publication: 14 DEC 2005. (absence <strong>of</strong> assessed intervention)<br />

238. Picchio M, Sironi S, Messa C, Mangili G, Landoni C, Gianolli L, Zangheri B, Vigano R, Aletti Gde M, De<br />

Cobelli F, Del Maschio A, Ferrari A, Fazio F. Advanced ovarian carcinoma: usefulness <strong>of</strong> [(18)F]FDG-<br />

<strong>PET</strong> in combination with <strong>CT</strong> for lesion detection after primary treatment. Q J Nucl Med 2003 Jun; 47<br />

(2): pp 77–84. (absence <strong>of</strong> assessed intervention)<br />

239. Poöttgen C, Levegrϋn S, Theegarten D, Marnitz S, Grehl S. Value <strong>of</strong> 18 F-fluoro-2-deoxy-D-glucose<strong>positron</strong><br />

<strong>emission</strong> tomography/computed tomography in non-small-cell lung cancer for prediction<br />

<strong>of</strong> pathologic response and times to relapse after neoadjuvant chemoradiotherapy. Clin Cancer<br />

Res 2006; Vol. 12 (1): pp 97–106. (absence <strong>of</strong> end-points for assessment <strong>of</strong> diagnostic efficacy)<br />

313


240. Poppe K, Lahoutte T, Everaert H, Bossuyt A, Velkeniers B. The utility <strong>of</strong> multimodality imaging in<br />

anaplastic thyroid carcinoma. Thyroid 2004 Nov; 14 (11): pp 981–982. (casuistic description)<br />

241. Raanani P, Shasha Y, Perry C, Metser U, Naparstek E, Apter S, Nagler A, Polliack A, Ben-Bassat I,<br />

Even-Sapir E. Is <strong>CT</strong> scan still necessary for staging in Hodgkin and non-Hodgkin lymphoma patients in<br />

the <strong>PET</strong>-<strong>CT</strong> era?. Ann Oncol 2006; Vol. 17 (1): pp 117–122. (absence <strong>of</strong> reference test)<br />

242. Reddy S, Kurli M, Tena LB, Finger PT. <strong>PET</strong>-<strong>CT</strong> imaging: detection <strong>of</strong> choroidal melanoma. Br J<br />

Ophthalmol 2005; Vol. 89 (10): pp 1265–1269. (absence <strong>of</strong> the classical comparative method)<br />

243. Rege S, Maass A, Chaiken L, Hoh CK, Choi Y, Lufkin R, Anzai Y, Juillard G, Maddahi J, Phelps ME. Use<br />

<strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography with fluorodeoxyglucose in patients with extracranial head and<br />

neck cancers. Cancer 1994; Vol. 73 (12): pp 3047–3058. (absence <strong>of</strong> assessed diagnostic method)<br />

244. Reinartz P, Wieres FJ, Schneider W, Schur A, Buell U. Side-by-side reading <strong>of</strong> <strong>PET</strong> and <strong>CT</strong> scans in<br />

oncology: which patients might pr<strong>of</strong>it from integrated <strong>PET</strong>-<strong>CT</strong>?. Eur J Nucl Med Mol Imaging 2004;<br />

Vol. 31 (11): pp 1456–1461. (absence <strong>of</strong> assessed intervention)<br />

245. Reinhardt MJ, Strunk H, Gerhardt T, Roedel R, Jaeger U, Bucerius J, Sauerbruch T, Biersack HJ,<br />

Dumoulin FL. Detection <strong>of</strong> Klatskin's tumor in extrahepatic bile duct strictures using delayed 18F-FDG<br />

<strong>PET</strong>-<strong>CT</strong>: preliminary results for 22 patient studies. J Nucl Med 2005; Vol. 46 (7): pp 1158–1163.<br />

(absence <strong>of</strong> the classical comparative method)<br />

246. Reinhardt MJ, Wiethoelter N, Matthies A, Joe AY, Strunk H, Jaeger U, Biersack H-J. <strong>PET</strong> recognition <strong>of</strong><br />

pulmonary metastases on <strong>PET</strong>-<strong>CT</strong> imaging: Impact <strong>of</strong> attenuation-corrected and non-attenuationcorrected<br />

<strong>PET</strong> images. Eur J Nucl Med Mol Imaging 2006; Vol. 33 (2): pp 134–139. (absence <strong>of</strong> the<br />

classical comparative method)<br />

247. Ribom D, Smits A. Baseline 11 C-methionine <strong>PET</strong> reflects the natural course <strong>of</strong> grade 2<br />

oligodendrogliomas. Neurological Research 2005; Vol. 27 (5): pp 516–521. (absence <strong>of</strong> assessed<br />

intervention)<br />

248. Rizzo G, Cattaneo GM, Castellone P, Castiglioni I, Ceresoli GL, Messa C, Landoni C, Gilardi MC,<br />

Arienti R, Cerutti S, Fazio F. Multi-modal medical image integration to optimize radiotherapy<br />

planning in lung cancer treatment. Ann Biomed Eng 2004 Oct; 32 (10): pp 1399–1408. (absence <strong>of</strong><br />

assessed intervention)<br />

249. Roman CD, Martin WH, Delbeke D. Incremental value <strong>of</strong> fusion imaging with integrated <strong>PET</strong>-<strong>CT</strong> in<br />

oncology. Clin Nucl Med 2005; Vol. 30 (7): pp 470–477. (absence <strong>of</strong> the classical comparative<br />

method)<br />

250. Rosa F, Meimarakis G, Stahl A, Bumm R, Hahn K, Tatsch K, Dresel S. Colorectal cancer patients<br />

before resection <strong>of</strong> hepatic metastases. Impact <strong>of</strong> (18)F-FDG <strong>PET</strong> on detecting extrahepatic<br />

disease. Nuklearmedizin 2004 Aug; 43 (4): pp 135–140. (absence <strong>of</strong> assessed intervention)<br />

251. Rosenbaum SJ, Stergar H, Antoch G, Veit P, Bockisch A, Kuhl H. Staging and follow-up <strong>of</strong><br />

gastrointestinal tumors with <strong>PET</strong>-<strong>CT</strong>. Abdominal Imaging 2006; Vol. 31 (1): pp 25–35. (secondary trial)<br />

252. Ruf J, Hanninen EL, Oettle H, Plotkin M, Pelzer U, Stroszczynski C, Felix R, Amthauer H. Detection <strong>of</strong><br />

recurrent pancreatic cancer: Comparison <strong>of</strong> FDG-<strong>PET</strong> with <strong>CT</strong>/MRI. Pancreatology 2005; Vol. 5 (2–3):<br />

pp 266–272. (absence <strong>of</strong> assessed intervention)<br />

253. Safa AA, Tran LM, Rege S, Brown CV, Mandelkern MA, Wang MB, Sadeghi A, Juillard G. The role <strong>of</strong><br />

<strong>positron</strong> <strong>emission</strong> tomography in occult primary head and neck cancers. Cancer Journal From<br />

Scientific American 1999; Vol. 5 (4): pp 214–218. (absence <strong>of</strong> assessed intervention)<br />

254. Sakamoto H, Nakai Y, Ohashi Y, Okamura T, Ochi H. Positron <strong>emission</strong> tomographic imaging <strong>of</strong> head<br />

and neck lesions. Eur Arch Otorhinolaryngol 1997; Vol. 254 (SUPPL. 1): pp S123-S126. (absence <strong>of</strong><br />

assessed intervention)<br />

255. Sakurai H, Suzuki Y, Nonaka T, Ishikawa H, Shioya M, Kiyohara H, Katoh H, Nakayama Y, Hasegawa<br />

M, Nakano T. FDG-<strong>PET</strong> in the detection <strong>of</strong> recurrence <strong>of</strong> uterine cervical carcinoma following<br />

radiation therapy-tumor volume and FDG uptake value. Gynecol Oncol 2006; Vol. 100 (3): pp 601–<br />

607. (absence <strong>of</strong> assessed intervention)<br />

256. Santos Dellea MM. Malignant Pleural Mesothelioma: Response Evaluation with Integrated <strong>PET</strong>-<strong>CT</strong><br />

Imaging. Radiological Society <strong>of</strong> North America 2004: p 648. (conference report)<br />

257. Sarinas PSA, Chitkara RK. <strong>PET</strong> and SPE<strong>CT</strong> in the management <strong>of</strong> lung cancer. Curr Opin Pulm Med<br />

2002; Vol. 8 (4): pp 257–264. (secondary trial)<br />

314


258. Scarfone C, Lavely WC, Cmelak AJ, Delbeke D, Martin WH, Billheimer D, Hallahan DE. Prospective<br />

feasibility trial <strong>of</strong> radiotherapy target definition for head and neck cancer using 3-dimensional <strong>PET</strong><br />

and <strong>CT</strong> imaging. J Nucl Med 2004 Apr; 45 (4): pp 543–552. (absence <strong>of</strong> assessed intervention)<br />

259. Schaffler GJ, Groell R, Schoellnast H, Kriegl D, Ruppert-Kohlmaier A, Schwarz T, Aigner RM. Digital<br />

image fusion <strong>of</strong> <strong>CT</strong> and <strong>PET</strong> data sets – clinical value in abdominal/pelvic malignancies. J Comput<br />

Assist Tomogr 2000; Vol. 24 (4): pp 644–647. (absence <strong>of</strong> assessed intervention)<br />

260. Scheidhauer K, Walter C, Seemann MD. FDG <strong>PET</strong> and other imaging modalities in the primary<br />

diagnosis <strong>of</strong> suspicious breast lesions. Eur J Nucl Med Mol Imaging 2004 Jun; 31 (Suppl 1): pp S70–79.<br />

Epub 2004 May 6. (secondary trial)<br />

261. Scher B, Seitz M, Reiser M, Hungerhuber E, Hahn K, Tiling R, Herzog P, Reiser M, Schneede P, Dresel S.<br />

18F-FDG <strong>PET</strong>-<strong>CT</strong> for staging <strong>of</strong> penile cancer. J Nucl Med 2005; Vol. 46 (9): pp 1460–1465. (absence<br />

<strong>of</strong> comparative diagnostic test)<br />

262. Schiepers C. <strong>PET</strong>-<strong>CT</strong> in colorectal cancer. J Nucl Med 2003; Vol. 44 (11): pp 1804–1805. (secondary<br />

trial)<br />

263. Schiepers C. Positron <strong>emission</strong> tomography: A coming <strong>of</strong> age?. Eur J Intern Med 2004; Vol. 15 (3): pp<br />

143–146. (secondary trial)<br />

264. Schmid DT, John H, Zweifel R, Cservenyak T, Westera G, Goerres GW, von Schulthess GK, Hany TF.<br />

Fluorocholine <strong>PET</strong>-<strong>CT</strong> in patients with prostate cancer: initial experience. Radiology 2005 May; 235<br />

(2): pp 623–628. (absence <strong>of</strong> comparative diagnostic test)<br />

265. Schmidt M, Schmalenbach M, Jungehulsing M, Theissen P, Dietlein M, Schroder U, Eschner W,<br />

Stennert E, Schicha H. 18 F-FDG <strong>PET</strong> for detecting recurrent head and neck cancer, local lymph node<br />

involvement and distant metastases: Comparison <strong>of</strong> qualitative visual and semiquantitative <strong>analysis</strong>.<br />

Nuklearmedizin 2004; Vol. 43 (3): pp 91–101. (absence <strong>of</strong> assessed intervention)<br />

266. Schmucking M, Baum RP, Bonnet R, Junker K, Müller K-M. Correlation <strong>of</strong> histologic results with <strong>PET</strong><br />

findings for tumor regression and survival in locally advanced non-small cell lung cancer after<br />

neoadjuvant treatment. Pathologe 2005; Vol. 26 (3): pp 178–190. (article in German)<br />

267. Schoder H, Erdi YE, Larson SM, Yeung HWD. <strong>PET</strong>-<strong>CT</strong>: a new imaging technology in nuclear medicine.<br />

Eur J Nucl Med Mol Imaging 2003; Vol. 30 (10): pp 1419–1437. (secondary trial)<br />

268. Schoder H, Herrmann K, Gonen M, Hricak H, Eberhard S, Scardino P, Scher HI, Larson SM. 2-[ 18 F]<br />

fluoro-2-deoxyglucose <strong>positron</strong> <strong>emission</strong> tomography for the detection <strong>of</strong> disease in patients with<br />

prostate-specific antigen relapse after radical prostatectomy. Clin Cancer Res 2005; Vol. 11 (13): pp<br />

4761–4769. (absence <strong>of</strong> separated results for <strong>PET</strong> and <strong>PET</strong>-<strong>CT</strong>)<br />

269. Schoder H, Larson SM, Yeung HW. <strong>PET</strong>-<strong>CT</strong> in oncology: integration into clinical management <strong>of</strong><br />

lymphoma, melanoma, and gastrointestinal malignancies. J Nucl Med 2004 Jan; 45 (Suppl 1): pp<br />

72S–81S. (secondary trial)<br />

270. Schoder H, Yeung HW, Gonen M, Kraus D, Larson SM. Head and neck cancer: clinical usefulness<br />

and accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> image fusion. Radiology 2004 Apr; 231 (1): pp 65–72. Epub 2004 Feb 27.<br />

(absence <strong>of</strong> the classical comparative method)<br />

271. Schrevens L, Lorent N, Dooms C, Vansteenkiste J. The role <strong>of</strong> <strong>PET</strong> scan in diagnosis, staging, and<br />

management <strong>of</strong> non-small cell lung cancer. Oncologist 2004; 9 (6): pp 633–643. (secondary trial)<br />

272. Schreyer AG, Kikinis R. Combined <strong>PET</strong>-<strong>CT</strong> colonography: Is this the way forward?. Gut 2006; Vol. 55<br />

(1): pp 10–12. (secondary trial)<br />

273. Schuetze SM. Imaging and response in s<strong>of</strong>t tissue sarcomas. Hematol Oncol Clin N Am 2005; Vol. 19<br />

(3): pp 471–487. (secondary trial)<br />

274. Schwaiger M, Wieder H. Role <strong>of</strong> <strong>PET</strong> in lymphoma. Chang Gung Med J 2005 May; 28 (5): pp 315–325.<br />

(secondary trial)<br />

275. Schwartz DL, Ford EC, Rajendran J, Yueh B, Coltrera MD, Virgin J, Anzai Y, Haynor D, Lewellyn B,<br />

Mattes D, Meyer J, Phillips M, Leblanc M, Kinahan P. Krohn K. Eary J. Laramore GE. FDG-<strong>PET</strong>-<strong>CT</strong><br />

imaging for preradiotherapy staging <strong>of</strong> head-and-neck squamous cell carcinoma. Int J Radiat<br />

Oncol Biol Phys 2005; Vol. 61 (1): pp 129–136. (absence <strong>of</strong> assessed intervention)<br />

276. Schwartz DL, Ford EC, Rajendran J, Yueh B, Coltrera MD, Virgin J, Anzai Y, Haynor D, Lewellen B,<br />

Mattes D, Kinahan P, Meyer J, Phillips M, Leblanc M, Krohn K, Eary J, Laramore GE. FDG-<strong>PET</strong>-<strong>CT</strong>-<br />

315


316<br />

guided intensity modulated head and neck radiotherapy: a pilot investigation. Head & Neck 2005;<br />

Vol. 27 (6): pp 478–487. (absence <strong>of</strong> assessed intervention)<br />

277. Selzner M, Hany TF, Wildbrett P, McCormack L, Kadry Z, Clavien P-A. Does the novel <strong>PET</strong>-<strong>CT</strong> imaging<br />

modality impact on the treatment <strong>of</strong> patients with metastatic colorectal cancer <strong>of</strong> the liver?. Ann<br />

Surg 2004; Vol. 240 (6): pp 1027–1036. (absence <strong>of</strong> the classical comparative diagnostic test)<br />

278. Shim SS, Lee KS, Kim BT, Choi JY, Shim YM, Chung MJ, Kwon OJ, Lee EJ. Integrated <strong>PET</strong>-<strong>CT</strong> and the<br />

dry pleural dissemination <strong>of</strong> peripheral adenocarcinoma <strong>of</strong> the lung: diagnostic implications.<br />

J Comput Assist Tomogr 2006 Jan-Feb; 30 (1): pp 70–76. (absence <strong>of</strong> independent assessment <strong>of</strong> <strong>CT</strong><br />

from the perspective <strong>of</strong> diagnostics without knowledge about clinical results)<br />

279. Singh AK, Grigsby PW, Dehdashti F, Herzog TJ, Siegel BA. FDG-<strong>PET</strong> lymph node staging and survival <strong>of</strong><br />

patients with FIGO Stage IIIb cervical carcinoma. Int J Radiat Oncol Biol Phys 2003; Vol. 56 (2): pp<br />

489–493. Date <strong>of</strong> Publication: 01 JUN 2003. (absence <strong>of</strong> assessed intervention)<br />

280. Sironi S, Buda A, Picchio M, Perego P, Moreni R, Pellegrino A, Colombo M, Mangioni C, Messa C,<br />

Fazio F. Lymph node metastasis in patients with clinical early-stage cervical cancer: Detection with<br />

integrated FDG <strong>PET</strong>-<strong>CT</strong>. Radiology 2006; Vol. 238 (1): pp 272–279. (absence <strong>of</strong> the classical<br />

comparative method)<br />

281. Sironi S, Messa C, Mangili G, Zangheri B, Aletti G, Garavagalia E, Vigano R, Picchio M, Taccagni G,<br />

Maschio AD, Fazio F. Integrated FDG <strong>PET</strong>-<strong>CT</strong> in patients with persistant ovarian cancer: corelation<br />

with histologic findings. Radiology 2004 Nov; 233 (2): pp 433–440. (absence <strong>of</strong> comparative<br />

diagnostic method)<br />

282. Solberg TD, Agazaryan N, Goss BW, Dahlbom M, Lee SP. A feasibility study <strong>of</strong> 18 F-fluorodeoxyglucose<br />

<strong>positron</strong> <strong>emission</strong> tomography targeting and simultaneous integrated boost for intensity-modulated<br />

radiosurgery and radiotherapy. J Neurosurg 2004; Vol. 101 (Suppl. 3): pp 381–389. (absence <strong>of</strong><br />

comparative diagnostic method)<br />

283. Som P, Urken M, Biller H, Lidov M. Imaging the postoperative neck. Radiology 1993; Vol. 187: pp 593–<br />

603. (absence <strong>of</strong> assessed intervention)<br />

284. Spraul CW, Lang GE, Lang GK. Value <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography in the diagnosis <strong>of</strong> malignant<br />

ocular tumors. Ophthalmologica 2001; Vol. 215 (3): pp 163–168. (absence <strong>of</strong> assessed intervention)<br />

285. Stahl A, Stollfuss J, Ott K, Wieder H, Fink U, Schwaiger M, Weber WA. FDG <strong>PET</strong> and <strong>CT</strong> in locally<br />

advanced adenocarcinomas <strong>of</strong> the distal oesophagus: Clinical relevance <strong>of</strong> a discordant <strong>PET</strong><br />

finding. World J Gastroenterol 2005; Vol. 11 (46): pp 7284–7289. Date <strong>of</strong> Publication: 14 DEC 2005.<br />

(absence <strong>of</strong> assessed intervention)<br />

286. Stahl A, Wieder H, Piert M, Wester HJ, Lordick F, Ott K, Rummeny E, Schwaiger M, Weber WA. <strong>PET</strong>-<strong>CT</strong><br />

molecular imaging in abdominal oncology. Abdom Imaging 2004 Vol. 29: pp 388–397. (secondary<br />

trial)<br />

287. Stahl A, Wieder H, Piert M, Wester HJ, Senekowitsch-Schmidtke R, Schwaiger M. Positron <strong>emission</strong><br />

tomography as a tool for translational research in oncology. Mol Imaging Biol 2004; Vol. 6 (4): pp<br />

214–224. (secondary trial)<br />

288. Steinert HC, Santos Dellea MM, Burger C, Stahel R. Therapy response evaluation in malignant pleural<br />

mesothelioma with integrated <strong>PET</strong>-<strong>CT</strong> imaging. Lung Cancer 2005; Vol. 49S1: pp S33–S35. (absence<br />

<strong>of</strong> reference method)<br />

289. Steinert HC, von Schulthess GK. Initial clinical experience using a new integrated in-line <strong>PET</strong>-<strong>CT</strong><br />

system. Br J Radiol 2002 Nov; 75 Spec No: pp S36–38. (review)<br />

290. Steinert HC. <strong>PET</strong> in lung cancer. Chang Gung Med J 2005 May; 28 (5): pp 296–305. (secondary trial)<br />

291. Strasberg SM, Siegal BA. Annals for Surgery: Letters to the editor [1] (multiple letters). Ann Surg 2002;<br />

Vol. 235 (2): pp 308–310. (letter)<br />

292. Sung J, Espiritu JI, Segall GM, Terris MK. Fluorodeoxyglucose <strong>positron</strong> <strong>emission</strong> tomography studies in<br />

the diagnosis and staging <strong>of</strong> clinically advanced prostate cancer. BJU International 2003; Vol. 92<br />

(1): pp 24–27. (absence <strong>of</strong> assessed intervention)<br />

293. Suzuki A, Kawano T, Takahashi N, Lee J, Nakagami Y, Miyagi E, Hirahara F, Togo S, Shimada H, Inoue<br />

T. Value <strong>of</strong> 18 F-FDG <strong>PET</strong> in the detection <strong>of</strong> peritoneal carcinomatosis. European Journal <strong>of</strong> Nuclear<br />

Medicine & Molecular Imaging 2004; Vol. 31 (10): pp 1413–1420. (absence <strong>of</strong> assessed intervention)


294. Swetter SM, Carroll LA, Johnson DL, Segall GM. Positron <strong>emission</strong> tomography is superior to<br />

computed tomography for metastatic detection in melanoma patients. Ann Surg Oncol 2002 Aug;<br />

9 (7): pp 646–653. (absence <strong>of</strong> assessed intervention)<br />

295. Syed R, Bomanji JB, Nagabhushan N, Hughes S, Kayani I, Groves A, Gacinovic S, Hydes N, Visvikis D,<br />

Copland C, Ell PJ. Impact <strong>of</strong> combined (18)F-FDG <strong>PET</strong>-<strong>CT</strong> in head and neck tumours. Br J Cancer<br />

2005 Mar 28; 92 (6): pp 1046–1050. (absence <strong>of</strong> the classical comparative method)<br />

296. Tanaka T, Kawai Y, Kanai M, Taki Y, Nakamoto Y, Takabayashi A. Usefulness <strong>of</strong> FDG-<strong>positron</strong> <strong>emission</strong><br />

tomography in diagnosing peritoneal recurrence <strong>of</strong> colorectal cancer. Am J Surg 2002 Nov; 184 (5):<br />

pp 433–436. (absence <strong>of</strong> assessed intervention)<br />

297. Tatsumi M, Cohade C, Nakamoto Y, Fishman EK, Wahl RL. Direct comparison <strong>of</strong> FDG <strong>PET</strong> and <strong>CT</strong><br />

findings in patients with lymphoma: initial experience. Radiology 2005 Dec; 237 (3): pp 1038–1045.<br />

(separated assessment <strong>of</strong> hybrid images for <strong>PET</strong> and <strong>CT</strong>, absence <strong>of</strong> the results for combined image)<br />

298. Tatsumi M, Kitayama H, Sugahara H, Tokita N, Nakamura H, Kanakura Y, Nishimura T. Whole-body<br />

hybrid <strong>PET</strong> with 18F-FDG in the staging <strong>of</strong> non-Hodgkin's lymphoma. J Nucl Med 2001 Apr; 42 (4): pp<br />

601–608. (absence <strong>of</strong> assessed intervention)<br />

299. Teknos TN, Rosenthal EL, Lee D, Taylor R, Marn CS. Positron <strong>emission</strong> tomography in the evaluation <strong>of</strong><br />

stage III and IV head and neck cancer. Head & Neck 2001; Vol. 23 (12): pp 1056–1060. (absence <strong>of</strong><br />

assessed intervention)<br />

300. Terhaard CH, Bongers V, Van Rijk PP, Hordijk G-J. F-18-fluoro-deoxy-glucose <strong>positron</strong>-<strong>emission</strong><br />

tomography scanning in detection <strong>of</strong> local recurrence after radiotherapy for laryngeal/pharyngeal<br />

cancer. Head & Neck 2001; Vol. 23 (11): pp 933–941. (absence <strong>of</strong> assessed intervention)<br />

301. Tierney PA, Farag I, Saunders CA. The use <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography and computed<br />

tomography in the assessment <strong>of</strong> trismus associated with head and neck malignancy. J Laryngol<br />

Otol 1998; Vol. 112 (3): pp 303–306. (casuistic description)<br />

302. Tisch M, Hengstermann F, Kraft K, Von Hinuber G, Maier H. Follicular dendritic cell sarcoma <strong>of</strong> the<br />

tonsil: Report <strong>of</strong> a rare case. Ear, Nose, & Throat Journal 2003; Vol. 82 (7): pp 507–509. (absence <strong>of</strong><br />

assessed intervention)<br />

303. Torizuka T, Kanno T, Futatsubashi M, Okada H, Yoshikawa E, Nakamura F, Takekuma M, Maeda M,<br />

Ouchi Y. Imaging <strong>of</strong> gynecologic tumors: Comparison <strong>of</strong> 11 C-choline <strong>PET</strong> with 18 F-FDG <strong>PET</strong>. J Nucl<br />

Med 2003; Vol. 44 (7): pp 1051–1056. (absence <strong>of</strong> assessed intervention)<br />

304. Townsend DW, Beyer T. A combined <strong>PET</strong>-<strong>CT</strong> scanner: the path to true image fusion. Br J Radiol 2002<br />

Nov; 75 Spec No: pp S24–30. (secondary trial)<br />

305. Truong MT, Erasmus JJ, Macapinlac HA, Marom EM, Mawlawi O, Gladish GW, Sabl<strong>of</strong>f BS, Bruzzi JF,<br />

Munden RF. Integrated <strong>positron</strong> <strong>emission</strong> tomography/computed tomography in patients with nonsmall<br />

cell lung cancer: normal variants and pitfalls. J Comput Assist Tomogr 2005 Mar-Apr; 29 (2): pp<br />

205–209. (absence <strong>of</strong> end-points)<br />

306. Truong MT, Erasmus JJ, Macapinlac HA, Podol<strong>of</strong>f DA. Teflon injection for vocal cord paralysis: Falsepositive<br />

finding on FDG <strong>PET</strong>-<strong>CT</strong> in a patient with non-small cell lung cancer. AJR Am J Roentgenol<br />

2004; Vol. 182 (6): pp 1587–1589. (casuistic description)<br />

307. Truong MT, Erasmus JJ, Munden RF, Marom EM, Sabl<strong>of</strong>f BS, Gladish GW, Podol<strong>of</strong>f DA, Macapinlac<br />

HA. Focal FDG uptake in mediastinal brown fat mimicking malignancy: a potential pitfall resolved<br />

on <strong>PET</strong>-<strong>CT</strong>. AJR Am J Roentgenol 2004; Vol. 183 (4): pp 1127–1132. (absence <strong>of</strong> assessed cancers)<br />

308. Turlakow A, Yeung HW, Salmon AS, Macapinlac HA, Larson SM. Peritoneal carcinomatosis: Role <strong>of</strong><br />

18 F-FDG <strong>PET</strong>. J Nucl Med 2003; Vol. 44 (9): pp 1407–1412. (absence <strong>of</strong> assessed intervention)<br />

309. Unger JB, Ivy JJ, Ramaswamy MR, Charrier A, Connor P. Whole-body [ 18 F]fluoro-2-deoxyglucose<br />

<strong>positron</strong> <strong>emission</strong> tomography scan staging prior to planned radical hysterectomy and pelvic<br />

lymphadenectomy. International Journal <strong>of</strong> Gynecological Cancer 2005; Vol. 15 (6): pp 1060–1064.<br />

(absence <strong>of</strong> assessed intervention)<br />

310. Van Eijkeren ME, De Schryver A, Goethals P, Poupeye E, Schelstraete K, Lemahieu I, De Potter CR.<br />

Measurement <strong>of</strong> short-term 11 C-thymidine activity in human head and neck tumours using <strong>positron</strong><br />

<strong>emission</strong> tomography (<strong>PET</strong>). Acta Oncologica 1992; Vol. 31 (5): pp 539–543. (absence <strong>of</strong> assessed<br />

diagnostic method)<br />

317


311. Van Kouwen MCA, Oyen WJG, Nagengast FM, Jansen JBMJ, Drenth JPH. FDG-<strong>PET</strong> scanning in the<br />

diagnosis <strong>of</strong> gastrointestinal cancers. Scand J Gastroenterol – Supplement 2004; Vol. 39 (241): pp<br />

85–92. (secondary trial)<br />

312. Vansteenkiste JF, Stroobants SG, Dupont PJ, De Leyn PR, De Wever WF, Verbeken EK, Nuyts JN,<br />

Maes FP, Bogaert JG. FDG-<strong>PET</strong> scan in potentially operable non-small cell lung cancer: do<br />

anatometabolic <strong>PET</strong>-<strong>CT</strong> fusion images improve the localisation <strong>of</strong> regional lymph node metastses?.<br />

Eur J Nucl Med 1998 Nov; 25 (11): pp 1495–1501. (absence <strong>of</strong> assessed intervention)<br />

313. Veit P, Kuhle C, Beyer T, Kuehl H, Herborn CU, Borsch G, Stergar H, Barkhausen J, Bockisch A, Antoch<br />

G. Whole body <strong>positron</strong> <strong>emission</strong> tomography/computed tomography (<strong>PET</strong>-<strong>CT</strong>) tumour staging with<br />

integrated <strong>PET</strong>-<strong>CT</strong> colonography: Technical feasibility and first experiences in patients with<br />

colorectal cancer. Gut 2006; Vol. 55 (1): pp 68–73. (absence <strong>of</strong> the classical comparative method)<br />

314. Visvikis D, Cheze-Le Rest C, Jarritt P. <strong>PET</strong> technology: Current trends and future developments. Br J<br />

Radiol 2004; Vol. 77 (923): pp 906–910. (review)<br />

315. Visvikis D, <strong>Cost</strong>a DC, Lonn AHR, Bomanji J, Gacinovic S, Ell PJ. <strong>CT</strong>-based attenuation correction in<br />

the calculation <strong>of</strong> semi-quantitative indices <strong>of</strong> [18F]FDG uptake in <strong>PET</strong>. Eur J Nucl Med Mol Imaging<br />

2003 Mar; 30 (3): pp 344–353. Epub 2002 Dec 20. (absence <strong>of</strong> end-points)<br />

316. von Schulthess GK, Steinert HC, Hany TF. Integrated <strong>PET</strong>-<strong>CT</strong>: current applications and future<br />

directions. Radiology 2006 Feb; 238 (2): pp 405–422. (secondary trial)<br />

317. von Schulthess GK. Positron <strong>emission</strong> tomography versus <strong>positron</strong> <strong>emission</strong> tomography<br />

/computed tomography: from “unclear” to “new-clear” medicine. Mol Imaging Biol 2004 Jul-Aug;<br />

6 (4): pp 183–187. (review)<br />

318. Wakabayashi H, Akamoto S, Yachida S, Okano K, Izuishi K, Nishiyama Y, Maeta H. Significance <strong>of</strong><br />

fluorodeoxyglucose <strong>PET</strong> imaging in the diagnosis <strong>of</strong> malignancies in patients with biliary stricture. Eur<br />

J Surg Oncol 2005; Vol. 31 (10): pp 1175–1179. (absence <strong>of</strong> assessed intervention)<br />

319. Wasyliw CW, Caride VJ. Incidental detection <strong>of</strong> bilateral elast<strong>of</strong>ibroma dorsi with F-18 FDG <strong>PET</strong>-<strong>CT</strong>.<br />

J Nucl Med 2005; Vol. 30 (10): pp 700–701. (casuistic description)<br />

320. Wechalekar K, Sharma B, Cook G. <strong>PET</strong>-<strong>CT</strong> in oncology – A major advance. Clinical Radiology 2005;<br />

Vol. 60 (11): pp 1143–1155. (review)<br />

321. Weckesser M, Konemann S, Brinkmann M, Willich N, Schober O. <strong>PET</strong>-<strong>CT</strong> in radiotherapy. Radiologe<br />

2004 Nov; 44 (11): pp 1096–104. (article in German)<br />

322. Wegner EA, Barrington SF, Kingston JE, Robinson RO, Ferner RE, Taj M, Smith MA, O'Doherty MJ. The<br />

impact <strong>of</strong> <strong>PET</strong> scanning on management <strong>of</strong> paediatric oncology patients. Eur J Nucl Med Mol<br />

Imaging 2005 Jan; 32 (1): pp 23–30. Epub 2004 Jul 31. (absence <strong>of</strong> assessed intervention)<br />

323. Weng L-J, Schoder H. Melanoma metastasis to the testis demonstrated with FDG <strong>PET</strong>-<strong>CT</strong>. Clin Nucl<br />

Med 2004; Vol. 29 (12): pp 811–812. (casuistic description)<br />

324. Wieder HA, Beer AJ, Lordick F, Ott K, Fischer M, Rummeny EJ, Ziegler S, Siewer JR, Schwaiger M,<br />

Weber WA. Comparison <strong>of</strong> changes in tumor metabolic activity and tumor size<br />

during chemotherapy <strong>of</strong> adenocarcinomas <strong>of</strong> the esophagogastric junction. J Nucl Med 2005 Dec;<br />

46 (12): pp 2029–2034. (absence <strong>of</strong> assessed intervention)<br />

325. Wiering B, Krabbe PF, Jager GJ, Oyen WJ, Ruers TJ. The impact <strong>of</strong> fluor-18-deoxyglucose-<strong>positron</strong><br />

<strong>emission</strong> tomography in the management <strong>of</strong> colorectal liver metastases. Cancer 2005 Dec 15; 104<br />

(12): pp 2658–2660. (secondary trial)<br />

326. Wiering B, Ruers TJM, Oyen WJG. Role <strong>of</strong> FDG-<strong>PET</strong> in the diagnosis and treatment <strong>of</strong> colorectal liver<br />

metastases. Expert Review <strong>of</strong> Anticancer Therapy 2004; Vol. 4 (4): pp 607–613. (secondary trial)<br />

327. Windorbska W, Lewandowska A. Positron <strong>emission</strong> tomography – The new diagnostic tool in<br />

oncology. Nowotwory 2004; Vol. 54 (1): pp 50–53. (secondary trial)<br />

328. Wolthaus JWH, van Herk M, Muller SH, Belderbos JSA, Lebesque JV, de Bois JA, Rossi MMG, Damen<br />

EMF. Fusion <strong>of</strong> respiration-correlated <strong>PET</strong> and <strong>CT</strong> scans: Correlated lung tumour motion in<br />

anatomical and functional scans. Phys Med Biol 2005; Vol. 50 (7): pp 1569–1583. (absence <strong>of</strong><br />

assessed intervention)<br />

329. Wong W-L, Hussain K, Chevretton E, Hawkes DJ, Baddeley H, Maisey M, McGurk M. Validation and<br />

clinical application <strong>of</strong> computer-combined computed tomography and <strong>positron</strong> <strong>emission</strong><br />

318


tomography with 2-[18F]fluoro-2-deoxy-D- glucose head and neck images. Am J Surg 1996; Vol. 172<br />

(6): pp 628–632. (absence <strong>of</strong> assessed intervention)<br />

330. Wright J, Dehdashti F, Herzog T, Mutch D, Huettner P, Rader J, Gibb R, Powell M, Gao F, Siegel B,<br />

Grigsby P. Preoperative lymph node staging <strong>of</strong> early-stage cervical carcinoma by [ 18 F]-fluoro-2deoxy-D-glucose-<strong>positron</strong><br />

<strong>emission</strong> tomography. Cancer 2005; Vol. 104 (11): pp 2484–2491. Date <strong>of</strong><br />

Publication: 01 DEC 2005. (absence <strong>of</strong> assessed intervention)<br />

331. Yau YY, Chan WS, Tam YM, Vernon P, Wong S, Coel M, Chu SK. Application <strong>of</strong> intravenous contrast<br />

in <strong>PET</strong>-<strong>CT</strong>: does it really introduce significant attenuation correction error?. J Nucl Med 2005; Vol. 46<br />

(2): pp 283–291. (absence <strong>of</strong> end-points)<br />

332. Yau YY, Samman N, Yeung RWK. Positron <strong>emission</strong> tomography/computed tomography true fusion<br />

imaging in clinical head and neck oncology: Early experience. Journal <strong>of</strong> Oral & Maxill<strong>of</strong>acial<br />

Surgery 2005; Vol. 63 (4): pp 479–486. (absence <strong>of</strong> the classical comparative method, casuistic<br />

description)<br />

333. Yen TC, Lai CH. Positron <strong>emission</strong> tomography in gynecologic cancer. Semin Nucl Med 2006 Jan; 36<br />

(1): pp 93–104. (secondary trial)<br />

334. Yeung HWD, Schoder H, Smith A, Gonen M, Larson SM. Clinical value <strong>of</strong> combined <strong>positron</strong> <strong>emission</strong><br />

tomography/computed tomography imaging in the interpretation <strong>of</strong> 2-deoxy-2-[F-18]fluoro-Dglucose-<strong>positron</strong><br />

<strong>emission</strong> tomography studies in cancer patients. Mol Imaging Biol 2005; Vol. 7 (3):<br />

pp 229–235. (absence <strong>of</strong> the classical comparative method)<br />

335. Yi JG, Marom EM, Munden RF, Truong MT, Macapinlac HA, Gladish FW, Sabl<strong>of</strong>f BS, Podol<strong>of</strong>f DA.<br />

Focal uptake <strong>of</strong> fluorodeoxyglucose by the thyroid in patients undergoing initial disease staging with<br />

combined <strong>PET</strong>-<strong>CT</strong> for non-small cell lung cancer. Radiology 2005; Vol. 236 (1): pp 271–275. (absence<br />

<strong>of</strong> verification <strong>of</strong> the study results in application <strong>of</strong> reference test)<br />

336. Yoshida Y, Kurokawa T, Kawahara K, Tsuchida T, Okazawa H, Fujibayashi Y, Yonekura Y, Kotsuji F.<br />

Incremental benefits <strong>of</strong> FDG <strong>positron</strong> <strong>emission</strong> tomography over <strong>CT</strong> alone for thepreperative staging<br />

<strong>of</strong> ovarian ccancer. AJR Am J Roentgenol 2004 Jan; 182 (1): pp 227–233. (absence <strong>of</strong> assessed<br />

intervention)<br />

337. Yun M, Lim JS, Noh SH, Hyung WJ, Cheong JH, Bong JK, Cho A, Lee JD. Lymph node staging <strong>of</strong><br />

gastric cancer using 18 F-FDG <strong>PET</strong>: a comparison study with <strong>CT</strong>. J Nucl Med 2005 Oct; 46 (10): pp<br />

1582–1588. (absence <strong>of</strong> assessed intervention)<br />

338. Zacharias T, Barrier A, Montravers F, Houry S, Lacaine F, Huguier M. Cardio-esophageal cancer. Is<br />

18Fluorodeoxyglucose <strong>positron</strong> <strong>emission</strong> tomography worthwhile?. Hepatogastroenterology 2004<br />

May-Jun; 51 (57): pp 741–743. (absence <strong>of</strong> assessed intervention)<br />

339. Zanation AM, Sutton DK, Couch ME, Weissler MC, Shockley WW, Shores CG. Use, accuracy, and<br />

implications for patient management <strong>of</strong> [18F]-2-fluorodeoxyglucose-<strong>positron</strong> <strong>emission</strong> /<br />

computerized tomography for head and neck tumors. Laryngoscope 2005; Vol. 115 (7): pp 1186–<br />

1190. (absence <strong>of</strong> the classical comparative method)<br />

340. Zangheri B, Messa C, Picchio M, Gianolli L, Landoni C, Fazio F. <strong>PET</strong>-<strong>CT</strong> and breast cancer. Eur J Nucl<br />

Med Mol Imaging 2004 Jun; 31 (Suppl 1): pp S135–142. Epub 2004 May 5. (secondary trial)<br />

341. Zimmer LA, Branstetter BF, Nayak JV, Johnson JT. Current use <strong>of</strong> 18F-fluorodeoxyglucose <strong>positron</strong><br />

<strong>emission</strong> tomography and combined <strong>positron</strong> <strong>emission</strong> tomography and computed tomography in<br />

squamous cell carcinoma <strong>of</strong> the head and neck. Laryngoscope 2005; Vol. 115 (11): pp 2029–2034.<br />

(secondary trial)<br />

342. Zimny M, Hochstenbag M, Lamers R, Reinartz P, Cremerius U, ten Velde G, Buell U. Mediastinal<br />

staging <strong>of</strong> lung cancer with 2-[fluorine-18]-fluoro-2-deoxy-D-glucose <strong>positron</strong> <strong>emission</strong> tomography<br />

and a dual-head coincidence gamma camera. Eur Radiol 2003; Vol. 13 (4): pp 740–747. (absence<br />

<strong>of</strong> assessed intervention)<br />

343. Zimny M, Wildberger JE, Cremerius U, DiMartino E, Jaenicke S, Nowak B, Bull U. Combined image<br />

interpretation <strong>of</strong> computed tomography and hybrid <strong>PET</strong> in head and neck cancer. Nuklearmedizin<br />

2002; Vol. 41 (1): pp 14–21. (absence <strong>of</strong> assessed intervention)<br />

344. Zinzani PL, Chierichetti F, Zompatori M, Tani M, Stefoni V, Garraffa G, Albertini P, Alinari L, Ferlin G,<br />

Baccarani M, Tura S. Advantages <strong>of</strong> <strong>positron</strong> <strong>emission</strong> tomography (<strong>PET</strong>) with respect to<br />

computed tomography in the follow-up <strong>of</strong> lymphoma patients with abdominal presentation. Leuk<br />

Lymphoma 2002 Jun; 43 (6): pp 1239–1243. (absence <strong>of</strong> assessed intervention)<br />

319


345. Zinzani PL, Fanti S, Battista G, Tani M, Castellucci P, Stefoni V, Alinari L, Farsad M, Musuraca G,<br />

Gabriele A, Marchi E, Nanni C, Canini R, Monetti N, Baccarani M. Predictive role <strong>of</strong> <strong>positron</strong> <strong>emission</strong><br />

tomography (<strong>PET</strong>) in the outcome <strong>of</strong> lymphoma patients. Br J Cancer 2004 Aug 31; 91 (5): pp 850–<br />

854. (absence <strong>of</strong> assessed intervention)<br />

346. Zissin R, Metser U, Hain D, Even-Sapir E. Mesenteric panniculitis in oncologic patients: <strong>PET</strong>-<strong>CT</strong> findings.<br />

Br J Radiol 2006; Vol. 79 (937): pp 37–43. (absence <strong>of</strong> clear results)<br />

320


21. TABLE OF ILLUSTRATIONS<br />

Table 1. Morbidity and mortality rates for selected malignant cancer types in Poland in 2002 [4] .........................................31<br />

Table 2. histological malignancy grades <strong>of</strong> lung cancer............................................................................................................40<br />

Table 3. TNM staging system <strong>of</strong> the clinical advancement <strong>of</strong> the lung cancer.........................................................................40<br />

Table 4. Killonian classification <strong>of</strong> non-Hodgkin’s lymphomas ...................................................................................................48<br />

Table 5. Hodgkin’s disease and non-Hodgkin’s lymphomas staging according to Ann Arbor staging system .....................49<br />

Table 6. REAL classification <strong>of</strong> lymphoid tumors...........................................................................................................................49<br />

Table 7. TNM and AJCC staging system <strong>of</strong> esophageal cancer.................................................................................................55<br />

Table 8. FIGO staging for endometrial cancer .............................................................................................................................59<br />

Table 9. FIGO staging for cervical cancer...................................................................................................................................62<br />

Table 10. AJCC and FIGO staging for ovarian cancer ...............................................................................................................65<br />

Table 11. TNM staging system by AJCC........................................................................................................................................68<br />

Table 12. AJCC staging system for exocrine pancreatic cancer ..............................................................................................75<br />

Table 13. 5-year survivals in pancreatic cancer by disease stage ...........................................................................................78<br />

Table 14. TNM staging system and tumor classification for colon cancer .................................................................................82<br />

Table 15. Astler-Coller classification <strong>of</strong> colorectal cancers .......................................................................................................83<br />

Table 16. Search strategy...............................................................................................................................................................93<br />

Table 17. Measurement results <strong>of</strong> assessing diagnostic tests ......................................................................................................96<br />

Table 18. Characteristics <strong>of</strong> parameters <strong>of</strong> diagnostic efficacy .................................................................................................97<br />

Table 19. List <strong>of</strong> the results <strong>of</strong> a search targeted at selected patient populations.....................................................................99<br />

Table 20. Initial characteristics <strong>of</strong> patients by trial .....................................................................................................................101<br />

Table 21. Description <strong>of</strong> intervention ...........................................................................................................................................103<br />

Table 22. Description <strong>of</strong> diagnostic tests compared..................................................................................................................104<br />

Table 23. Consistency between T-staging and reference test, by <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong> trial. .............................................................106<br />

Table 24. Number <strong>of</strong> TP, FP, FN and TN patients in N-staging using <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong>..................................................................110<br />

Table 25. Evaluation <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in N-staging. ...................................................................111<br />

Table 26. Lung cancer staging results for <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> by trial .............................................................................................116<br />

Table 27. Initial characteristics <strong>of</strong> lymphoma patients ..............................................................................................................122<br />

Table 28. Description <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> imaging for both studies...........................................................................................................123<br />

Table 29. List <strong>of</strong> diagnosis reference tests ...................................................................................................................................124<br />

Table 30. Test diagnostic efficacy in detecting cancer in node locations...............................................................................125<br />

Table 31. Test diagnostic efficacy in detecting cancer locations outside <strong>of</strong> nodes ...............................................................126<br />

Table 32. Diagnostic efficacy in lymphomas detection in locations in and outside <strong>of</strong> nodes. ..............................................127<br />

Table 33. Diagnostic accuracy in detecting lymphomas .........................................................................................................138<br />

Table 34. Results <strong>of</strong> lymphoma staging by <strong>PET</strong>-<strong>CT</strong> as compared to <strong>CT</strong>. ...................................................................................138<br />

Table 35. Odds ratio for correct lymphoma staging: <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>...........................................................................................139<br />

Table 36. Patient characteristics in Cerfolio 2005.......................................................................................................................142<br />

Table 37. <strong>PET</strong>-<strong>CT</strong> imaging Cerfolio 2005 ......................................................................................................................................142<br />

Table 38. T-staging results for each <strong>of</strong> the diagnostic methods as compared to the reference test (Cerfolio 2005) ...........144<br />

Table 39. <strong>PET</strong>-<strong>CT</strong>, EUS-FNA and <strong>CT</strong> diagnostic efficacy in tumor restaging ..............................................................................144<br />

Table 40. Proportion <strong>of</strong> cases correctly staged, understaged and overstaged by the diagnostic methods analysed. ......145<br />

321


Table 41. Odds ratio for correct T-staging NNT for diagnostic methods analyzed: <strong>PET</strong>-<strong>CT</strong> vs. EUS and <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>...........146<br />

Table 42. Diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>, EUS-FNA and <strong>CT</strong> in assessing lymph node involvement.........................................146<br />

Table 43. Diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>, EUS-FNA and <strong>CT</strong> in M-staging: stage M1a ...............................................................148<br />

Table 44. Diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in M-staging: stage M1b................................................................................148<br />

Table 45. Diagnostic efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong>, EUS-FNA and <strong>CT</strong> in assessing complete response...................................................150<br />

Table 46. Initial patient characteristics in Kula 2005 ..................................................................................................................152<br />

Table 47. <strong>PET</strong>-<strong>CT</strong> test characteristics............................................................................................................................................153<br />

Table 48. Characteristics <strong>of</strong> patients diagnosed with malignant female genital tumor in Grisaru 2004. ...............................155<br />

Table 49. Information on <strong>PET</strong>-<strong>CT</strong> scanner type and scanning method.....................................................................................156<br />

Table 50. Number <strong>of</strong> patients with TP, FP, FN and TN results in the group with the disease staged: <strong>PET</strong>-<strong>CT</strong> vs.<br />

conventional methods (<strong>CT</strong>, MRI and USG) .................................................................................................................157<br />

Table 51. EBM parameters <strong>of</strong> the diagnostic efficacy in disease staging: <strong>PET</strong>-<strong>CT</strong> vs. conventional methods (<strong>CT</strong>, MRI<br />

and USG) .......................................................................................................................................................................157<br />

Table 52. Patients with TP, FP, FN and TN results in the group with disease recurrence assessed, <strong>PET</strong>-<strong>CT</strong> vs. conventional<br />

methods (<strong>CT</strong>, MRI and USG).........................................................................................................................................159<br />

Table 53. EBM parameters <strong>of</strong> the diagnostic efficacy in assessing disease recurrence: <strong>PET</strong>-<strong>CT</strong> vs. conventional<br />

methods (<strong>CT</strong>, MRI and USG).........................................................................................................................................159<br />

Table 54. Number <strong>of</strong> patients with TP, FP, FN and TN results, <strong>PET</strong>-<strong>CT</strong> vs. conventional methods (<strong>CT</strong>, MRI and USG) .............161<br />

Table 55. EBM parameters <strong>of</strong> diagnostic efficacy in disease staging and disease recurrence assessment; <strong>PET</strong>-<strong>CT</strong> vs.<br />

conventional methods (<strong>CT</strong>, MRI and USG) .................................................................................................................161<br />

Table 56. Characteristics <strong>of</strong> population included in the <strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> efficacy in the assessment <strong>of</strong> ovarian<br />

cancer recurrences......................................................................................................................................................165<br />

Table 57. Number <strong>of</strong> patients with TP, FP, FN and TN results in Hauth 2005 ...............................................................................166<br />

Table 58. <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> diagnostic efficacy in assessing ovarian cancer recurrences. .......................................................167<br />

Table 59. Location <strong>of</strong> lesions detected by <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> scanning, Hauth 2005....................................................................168<br />

Table 60. Number <strong>of</strong> patients with TP, FP, FN and TN results in Makhija 2001 ...........................................................................168<br />

Table 61. Sensitivity <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in Makhija 2001...............................................................................................................169<br />

Table 62. Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> sensitivity in detecting ovarian cancer recurrence .............................................170<br />

Table 63. Initial characteristics <strong>of</strong> subjects <strong>of</strong> each study .........................................................................................................173<br />

Table 64. Description <strong>of</strong> scanner and method used for <strong>PET</strong>-<strong>CT</strong> examination...........................................................................174<br />

Table 65. Number <strong>of</strong> patients with TP, FP, FN and TN results in Zimmer 2003, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>/MRI .............................................175<br />

Table 66. EBM parameters <strong>of</strong> diagnostic efficacy in detecting thyroid cancer recurrence, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>/MRI ...................176<br />

Table 67. Number <strong>of</strong> patients with TP, FP, FN and TN results in both studies, <strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body scintigraphy..........177<br />

Table 68. Sensitivity calculated for each study, <strong>PET</strong>-<strong>CT</strong> vs I 131 whole-body scintigraphy........................................................178<br />

Table 69. Comparison <strong>of</strong> sensitivity in evaluating recurrence, <strong>PET</strong>-<strong>CT</strong> vs I 131 whole-body scintigraphy ................................179<br />

Table 70. Comparison <strong>of</strong> specificity, <strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body scintigraphy .........................................................................179<br />

Table 71. comparison <strong>of</strong> diagnostic accuracy, <strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body scintigraphy.......................................................180<br />

Table 72. Diagnostic accuracy in evaluating disease recurrence, <strong>PET</strong>-<strong>CT</strong> vs. I 131 whole-body scintigraphy .......................181<br />

Table 73. EBM parameters <strong>of</strong> diagnostic efficacy <strong>of</strong> imaging methods compared, <strong>PET</strong>-<strong>CT</strong> vs I 131 whole-body<br />

scintigraphy ..................................................................................................................................................................182<br />

Table 74. Initial characteristics <strong>of</strong> patients examined................................................................................................................184<br />

Table 75. Characteristics <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanner and method. ........................................................................................................185<br />

Table 76. Number <strong>of</strong> lesions detected with the imaging methods compared vs. the reference test for each feature <strong>of</strong><br />

the TNM staging system. ..............................................................................................................................................187<br />

Table 77 Diagnostic sensitivity in lesion staging for each feature <strong>of</strong> the TNM system: I 124 <strong>PET</strong>-<strong>CT</strong> vs. traditional methods<br />

(<strong>CT</strong>, I 131 WBS, USG) .........................................................................................................................................................187<br />

Table 78. Characteristics <strong>of</strong> subject population in Branstetter 2005 .........................................................................................190<br />

Table 79. Test results for each <strong>of</strong> the <strong>of</strong> methods compared vs. reference test: Branstetter 2005; per number <strong>of</strong> lesions....192<br />

322


Table 80. Diagnostic efficacy parameters <strong>of</strong> the methods compared in Branstetter 2005 per number <strong>of</strong> lesions ...............192<br />

Table 81. Population characteristics in Goerres 2005 ................................................................................................................194<br />

Table 82. Test results for each <strong>of</strong> the methods compared vs. the reference test in Goerres 2005; calculated per patient .195<br />

Table 83. Diagnostic efficacy parameters for the technologies compared in Goerres 2005, calculated per patient.........196<br />

Table 84. Population characteristics............................................................................................................................................199<br />

Table 85. Impact <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> on revisions to head and neck cancer therapy ...........................................................................201<br />

Table 86. Revisions in treatment <strong>of</strong> head an neck cancer based on <strong>PET</strong>-<strong>CT</strong> findings .............................................................202<br />

Table 87. Initial characteristics <strong>of</strong> patients in Heinrich 2005. .....................................................................................................204<br />

Table 88. Characteristics <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> scanner and the procedures used in testing. ..................................................................205<br />

Table 89. Number <strong>of</strong> patients with TP, FP, FN and TN results in Heinrich 2005: <strong>PET</strong>-<strong>CT</strong> vs. contrast <strong>CT</strong>.....................................207<br />

Table 90. Diagnostic efficacy parameters <strong>of</strong> the imaging methods compared: <strong>PET</strong>-<strong>CT</strong> vs. contrast <strong>CT</strong> ...............................207<br />

Table 91. Number <strong>of</strong> patients with TP, FP, FN and TN results in M-staging, <strong>PET</strong>-<strong>CT</strong> vs. conventional methods .......................208<br />

Table 92. EBM parameters <strong>of</strong> diagnostic efficacy for the diagnostic methods compared; <strong>PET</strong>-<strong>CT</strong> vs. conventional<br />

methods ........................................................................................................................................................................209<br />

Table 93. Probability <strong>of</strong> revision <strong>of</strong> oncological procedure in patients diagnosed using the methods compared: <strong>PET</strong>-<strong>CT</strong><br />

vs. conventional methods............................................................................................................................................210<br />

Table 94. <strong>PET</strong>-<strong>CT</strong> details ................................................................................................................................................................214<br />

Table 95. <strong>CT</strong> details.......................................................................................................................................................................214<br />

Table 96. Scanning findings vs. reference test............................................................................................................................216<br />

Table 97. Correct response to treatment ....................................................................................................................................216<br />

Table 98. Scanner and <strong>PET</strong>-<strong>CT</strong> procedure...................................................................................................................................219<br />

Table 99. Sensitivity and accuracy <strong>of</strong> the diagnostic methods compared in detecting residual lesions after ablation <strong>of</strong><br />

colorectal liver metastasis, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong> ...................................................................................................................220<br />

Table 100. Initial characteristics <strong>of</strong> patients included in trials ...................................................................................................222<br />

Table 101. Description <strong>of</strong> intervention .........................................................................................................................................224<br />

Table 102. Description <strong>of</strong> diagnostic tests compared................................................................................................................225<br />

Table 103. Number <strong>of</strong> patients with TP, FP, FN and TN results in detecting cancer in patients with varied location <strong>of</strong> the<br />

disease using <strong>PET</strong>-<strong>CT</strong> and MRI. ....................................................................................................................................226<br />

Table 104. Accuracy in detecting cancer in patients with the disease in varied location using <strong>PET</strong>-<strong>CT</strong> vs MRI . .................226<br />

Table 105. Consistency <strong>of</strong> the T feature with the reference test for patients with neoplasm in varied locations, <strong>PET</strong>-<strong>CT</strong> vs<br />

MRI.................................................................................................................................................................................228<br />

Table 106. Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI accuracy in T-staging in patients with neoplasm in varied locations. ........230<br />

Table 107. Consistency <strong>of</strong> N-staging results with the reference test in patients with neoplasm in varied locations, <strong>PET</strong>-<br />

<strong>CT</strong> vs. MRI ......................................................................................................................................................................232<br />

Table 108. Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI accuracy in N-staging in patients with neoplasm in varied locations. .......233<br />

Table 109. The diagnostic accuracy <strong>of</strong> N-staging in patients with neoplasm in varied locations. <strong>PET</strong>-<strong>CT</strong> vs. MRI................234<br />

Table 110. Diagnostic accuracy evaluation parameters for N-staging using <strong>PET</strong>-<strong>CT</strong> vs MRI, as calculated in Schmidt<br />

2005 in relation to the number <strong>of</strong> lymph nodes assessed. ........................................................................................235<br />

Table 111. Consistency <strong>of</strong> distant metastasis staging (M feature) with reference test in patients with neoplasm in<br />

varied locations, <strong>PET</strong>-<strong>CT</strong> vs MRI ...................................................................................................................................236<br />

Table 112. Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI accuracy in M-staging in patients with neoplasm in varied locations........237<br />

Table 113. Evaluation <strong>of</strong> the efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI in detecting distant metastasis in patients with<br />

neoplasm in varied locations...................................................................................................................................238<br />

Table 114. Parameters <strong>of</strong> diagnostic efficacy in M-staging with the use <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI, calculated in Schmidt 2005<br />

based on the number <strong>of</strong> pathological changes........................................................................................................239<br />

Table 115. Consistency between the reference test and cancer staging results (TNM feature) in patients with<br />

neoplasm in varied locations, <strong>PET</strong>-<strong>CT</strong> vs. MRI.............................................................................................................240<br />

Table 116. Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and MRI accuracy in TNM-staging in patients with neoplasm in varied locations....242<br />

323


Table 117. Influence <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> vs MRI scanning results on therapy changes in patients with neoplasm in varied<br />

locations........................................................................................................................................................................243<br />

Table 118. Initial characteristics <strong>of</strong> patients with neoplasm in varied locations included in Antoch 2004............................246<br />

Table 119. Description <strong>of</strong> the intervention in Antoch 2004.........................................................................................................247<br />

Table 120. Description <strong>of</strong> the diagnostic <strong>CT</strong> tests performed in Antoch 2004 ..........................................................................248<br />

Table 121. Consistency <strong>of</strong> T-staging with the reference test in patients with neoplasm in varied locations in Antoch<br />

2004, <strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong>.........................................................................................................................................................249<br />

Table 122. Consistency between the reference test and N-staging results in patients with carcinomas in varied<br />

locations, <strong>PET</strong>-<strong>CT</strong> vs <strong>CT</strong> .................................................................................................................................................250<br />

Table 123. Consistency between the reference test and M-staging in patients with neoplasm in varied locations, <strong>PET</strong>-<br />

<strong>CT</strong> vs. <strong>CT</strong> ........................................................................................................................................................................251<br />

Table 124. Evaluation <strong>of</strong> the efficacy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in detecting metastasis in patients with neoplasm in varied<br />

locations........................................................................................................................................................................251<br />

Table 125. Parameters <strong>of</strong> the diagnostic efficacy <strong>of</strong> M-staging using <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> in patients with typical indications<br />

for <strong>PET</strong> in Antoch 2004. .................................................................................................................................................252<br />

Table 126. Consistency between the reference test and cancer staging in patients with neoplasm in varied locations,<br />

<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>..................................................................................................................................................................253<br />

Table 127. Influence <strong>of</strong> the diagnostic methods compared on therapy in patients with neoplasm in varied locations,<br />

<strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>..................................................................................................................................................................253<br />

Table 128. Initial characteristics <strong>of</strong> patients included in trials. ..................................................................................................256<br />

Table 129. Description <strong>of</strong> intervention .........................................................................................................................................257<br />

Table 130. Description <strong>of</strong> the diagnostic tests compared .........................................................................................................257<br />

Table 131. Number <strong>of</strong> metastasis patients with TP, FP, FN and TN results in the diagnostics <strong>of</strong> primary tumor using <strong>PET</strong>-<strong>CT</strong><br />

and <strong>CT</strong> ...........................................................................................................................................................................259<br />

Table 132. <strong>PET</strong>-<strong>CT</strong> sensitivity vs. <strong>CT</strong> sensitivity in detecting primary carcinomas <strong>of</strong> unknown locations................................259<br />

Table 133. Average values <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> and <strong>CT</strong> sensitivity in primary carcinoma detection in patients with metastasis to<br />

the neck area. ..............................................................................................................................................................261<br />

Table 134. Antoch 2003 quality assessment: QUADAS checklist...............................................................................................264<br />

Table 135. Lardinois 2003 quality assessment: QUADAS checklist ............................................................................................265<br />

Table 136. Cerfolio 2005 quality assessment: QUADAS checklist.............................................................................................266<br />

Table 137. Shim 2005 quality assessment: QUADAS checklist..................................................................................................267<br />

Table 138. Antoch 2004 quality assessment: QUADAS checklist (mixed) ................................................................................268<br />

Table 139. Antoch 2003 quality assessment: QUADAS checklist (mixed) ...............................................................................269<br />

Table 140. Schmidt 2005 quality assessment: QUADAS checklist ............................................................................................270<br />

Table 141. Freudenberg 2005 quality assessment: QUADAS checklist ....................................................................................271<br />

Table 142. Gutzeit 2005 quality assessment: QUADAS checklist...............................................................................................272<br />

Table 143. Freudenberg 2003 quality assessment: QUADAS checklist .....................................................................................273<br />

Table 144. Schaefer 2004 quality assessment: QUADAS checklist............................................................................................274<br />

Table 145. Cerfolio 2005 quality assessment: QUADAS checklist (esopaegal and gastric cancer) .....................................275<br />

Table 146. Kula 2005 quality assessment: QUADAS checklist ...................................................................................................276<br />

Table 147. Grisaru 2004 quality assessment: QUADAS checklist...............................................................................................277<br />

Table 148. Hauth 2005 quality assessment: QUADAS checklist................................................................................................278<br />

Table 149. Makhija 2001 quality assessment: QUADAS checklist ............................................................................................279<br />

Table 150. Nahas 2005 quality assessment: QUADAS checklist................................................................................................280<br />

Table 151. Freudenberg 2004 quality assessment: QUADAS checklist .....................................................................................281<br />

Table 152. Zimmer 2003 quality assessment: QUADAS checklist .............................................................................................282<br />

Table 153. Wild 2006 quality assessment: QUADAS checklist....................................................................................................283<br />

Table 154. Branstetter 2005 quality assessment: QUADAS checklist .........................................................................................284<br />

324


Table 155. Goerres 2005 quality assessment: QUADAS checklist ............................................................................................285<br />

Table 156. Koshy 2005 quality assessment: QUADAS checklist................................................................................................286<br />

Table 157. Heinrich 2005 quality assessment: QUADAS checklist.............................................................................................287<br />

Table 158. Antoch 2004 quality assessment: QUADAS checklist...............................................................................................288<br />

Table 159. Veit 2006 quality assessment: QUADAS checklist ....................................................................................................289<br />

Table 160. Result <strong>of</strong> the serach in Cochrane database.............................................................................................................290<br />

Table 161. Results <strong>of</strong> the serach in Pubmed data basis .............................................................................................................291<br />

Table 162. Results <strong>of</strong> serach in Embase database .....................................................................................................................292<br />

Table 163. MeSH terms used for the search strategy .................................................................................................................292<br />

325


22. TABLE OF GRAPHS<br />

Graph 1. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in T-staging ............................................................................108<br />

Graph 2. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>CT</strong> results in T-staging........................................................................108<br />

Graph 3. Odds ratio for the consistency between the reference test and T-staging results, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>. ..........................109<br />

Graph 4. Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostic accuracy in N-staging......................................................................................112<br />

Graph 5. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>CT</strong> in N-staging ..................................................................................113<br />

Graph 6. Odds ratio for the consistency <strong>of</strong> N-staging with the reference test, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong>. ................................................114<br />

Graph 7. Diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in cancer staging (TNM) .........................................................................................118<br />

Graph 8. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>CT</strong> in cancer staging (TNM)..............................................................119<br />

Graph 9. Odds ratio for the consistency between the reference test and total cancer staging results, <strong>PET</strong>-<strong>CT</strong> vs. <strong>CT</strong> ........119<br />

Graph 10. <strong>PET</strong>-<strong>CT</strong> sensitivity..........................................................................................................................................................127<br />

Graph 11. <strong>CT</strong> sensitivity ................................................................................................................................................................128<br />

Graph 12. <strong>PET</strong>-<strong>CT</strong> specificity.........................................................................................................................................................129<br />

Graph 13. <strong>CT</strong> specificity ...............................................................................................................................................................130<br />

Graph 14. Positive likelihood ratio for <strong>PET</strong>-<strong>CT</strong> scanning .............................................................................................................131<br />

Graph 15. Positive likelihood ratio for..........................................................................................................................................132<br />

Graph 16. Negative likelihood ratio for <strong>PET</strong>-<strong>CT</strong> study.................................................................................................................133<br />

Graph 17. Negative likelihood ratio for <strong>CT</strong> study........................................................................................................................134<br />

Graph 18. Diagnostic odds ratio for <strong>PET</strong>-<strong>CT</strong>.................................................................................................................................135<br />

Graph 19. Diagnostic odds ratio for <strong>CT</strong> .......................................................................................................................................136<br />

Graph 20. <strong>PET</strong>-<strong>CT</strong> accuracy .........................................................................................................................................................137<br />

Graph 21. <strong>CT</strong> accuracy................................................................................................................................................................137<br />

Graph 22. Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> sensitivity in detecting ovarian cancer recurrence. .......................................................169<br />

Graph 23. Meta-<strong>analysis</strong> <strong>of</strong> <strong>CT</strong> sensitivity in detecting ovarian cancer recurrence ...............................................................170<br />

Graph 24. Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> sensitivity in detecting thyroid cancer recurrence..........................................................178<br />

Graph 25. Meta-<strong>analysis</strong> <strong>of</strong> diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> .....................................................................................................180<br />

Graph 26. Meta-<strong>analysis</strong> <strong>of</strong> diagnostic accuracy <strong>of</strong> I 131 whole-body scintigraphy ................................................................181<br />

Graph 27. Meta-<strong>analysis</strong> (fixed effects model) <strong>of</strong> the proportion <strong>of</strong> patients for head and neck cancer therapy was<br />

revised following <strong>PET</strong>-<strong>CT</strong> examination ........................................................................................................................202<br />

Graph 28. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in T-staging in patients with neoplasm in varied<br />

locations........................................................................................................................................................................229<br />

Graph 29. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> MRI in T-staging in patients with neoplasm in varied locations...230<br />

Graph 30. Odds ratio for the consistency between the reference test and T- staging results for patients with neoplasm<br />

in varied locations, <strong>PET</strong>-<strong>CT</strong> vs. MRI ..............................................................................................................................231<br />

Graph 31. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in N-staging in patients with neoplasm in varied<br />

locations........................................................................................................................................................................232<br />

Graph 32. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> MRI in N-staging in patients with neoplasm in varied locations..233<br />

Graph 33. Odds ratio for the consistency between the reference test and N-staging result in patients with neoplasm in<br />

varied locations, <strong>PET</strong>-<strong>CT</strong> vs. MRI ..................................................................................................................................234<br />

Graph 34. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in M-staging in patients with neoplasm in varied<br />

locations........................................................................................................................................................................236<br />

Graph 35. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> MRI in M-staging in patients with neoplasm in varied locations .237<br />

Graph 36. Odds ratio for the consistency <strong>of</strong> M-staging results with the reference test in patients with neoplasm in<br />

varied locations, <strong>PET</strong>-<strong>CT</strong> vs MRI ...................................................................................................................................238<br />

326


Graph 37. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> in cancer staging in patients with neoplasm in varied<br />

locations........................................................................................................................................................................241<br />

Graph 38. Meta-<strong>analysis</strong> <strong>of</strong> the diagnostic accuracy <strong>of</strong> MRI in cancer staging in patients with neoplasm in varied<br />

locations........................................................................................................................................................................242<br />

Graph 39. Odds ratio for the consistency <strong>of</strong> TNM-staging results with the reference test in patients with neoplasm in<br />

varied locations, <strong>PET</strong>-<strong>CT</strong> vs MRI ...................................................................................................................................243<br />

Graph 40. Meta-<strong>analysis</strong> <strong>of</strong> <strong>PET</strong>-<strong>CT</strong> diagnostic sensitivity in patients with metastasis to neck area ......................................260<br />

Graph 41. Meta-<strong>analysis</strong> <strong>of</strong> <strong>CT</strong> diagnostic sensitivity in patients with metastasis to the neck area.......................................261<br />

Graph 42. Odds ratio for primary carcinoma detection in patients with metastasis to the neck area .................................262<br />

327


23. TABLE OF FIGURES<br />

Figure 1. Global incidence and mortality rates for the most common tumors types in developing and developed<br />

countries [1] ....................................................................................................................................................................30<br />

Figure 2. European average relative 5-year survival rates (%) for 42 selected tumor types; adults aged 15–99,<br />

diagnosed in 1990–1994, with the follow-up period till 1999 [5]..................................................................................34<br />

Figure 3. Incidence and mortality rates for ovarian cancer in selected geographic regions [7] ............................................64<br />

Figure 4. Algorithm <strong>of</strong> therapeutic procedure for pancreatic cancer; source: The Polish Oncology Union, Zalecenia<br />

postępowania diagnostyczno-terapeutycznego w nowotworach złośliwych u dorosłych, edited by M.<br />

Krzakowski [6] .................................................................................................................................................................77<br />

Figure 5. Annihilation and detection <strong>of</strong> gamma rays .................................................................................................................87<br />

Figure 6. Dissemination <strong>of</strong> tumor: metastatic lymph nodes in the left groin a) <strong>CT</strong> mage; b) <strong>PET</strong>/<strong>CT</strong> image; c) <strong>PET</strong> image ....88<br />

328

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!