2012 Breast Cancer Symposium Educational Summaries - American ...

2012 Breast Cancer Symposium Educational Summaries - American ... 2012 Breast Cancer Symposium Educational Summaries - American ...

22.12.2012 Views

2012 Breast Cancer Symposium Educational Summaries General Session I: Controversial Topics in Medical Oncology Management Role of Chemotherapy for Small Triple-Negative and HER2-Positive Cancers Robert W. Carlson, MD ....................................................................................................................................4 General Session II: Risk Assessment, Prevention, Detection, and Screening Imaging Techniques for Breast Cancer Detection and Diagnosis: What Works and What Doesn’t David Mankoff, MD, PhD ................................................................................................................................7 Surgical Risk Reduction for Atypical Ductal Hyperplasia, Lobular Carcinoma In Situ, and Ductal Carcinoma In Situ Todd M. Tuttle, MD ..........................................................................................................................................9 Prevention Strategies for Women at High Risk and for Those with Lobular or Ductal Carcinoma In Situ Victor G. Vogel, MD, MHS .............................................................................................................................10 General Session III: Controversies in Diagnosis of Early-Stage Breast Cancer and Management of Sentinel Nodes Magnetic Resonance Imaging Debate: Con Kathryn Evers, MD ........................................................................................................................................14 General Session IV: Controversies in Local/Regional Treatment Debate on the Role of Local Radiation Therapy in Ductal Carcinoma In Situ: Pro Shivani Duggal, DO, and Thomas B. Julian, MD .......................................................................................17 Targeted Intraoperative Radiotherapy: Analysis of a Randomized Trial—The TARGIT-A Trial Jayant S. Vaidya, MBBS, MS, DNB, PhD ....................................................................................................19 Debate on the Role of Radiation Therapy in Ductal Carcinoma In Situ: Con Abram Recht, MD ...........................................................................................................................................22 Intraoperative Radiation Therapy for Breast Cancer Julia White, MD ..............................................................................................................................................25 General Session V: Genomics for the Clinician PI3K in Breast Cancer Toby M. Ward, PhD, Rebecca Olson, MS, Mark D. Pegram, MD, and Charles E. Geyer Jr., MD ........28 Role of Genomic Testing in Patients with Node-Positive Breast Cancer Shaheenah Dawood, MRCP, MPH, and Ana M. Gonzalez-Angulo, MD, MSc ........................................31 Breast Cancer Whole Genome Sequencing Matthew J. Ellis, MB BChir, PhD ................................................................................................................34 General Session VI: Health Care Delivery and Prevention Breast Health Care Delivery in Context: The World Health Organization’s Initiative for Noncommunicable Diseases Benjamin O. Anderson, MD ..........................................................................................................................36 1

<strong>2012</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

<strong>Educational</strong> <strong>Summaries</strong><br />

General Session I: Controversial Topics in Medical Oncology Management<br />

Role of Chemotherapy for Small Triple-Negative and HER2-Positive <strong>Cancer</strong>s<br />

Robert W. Carlson, MD ....................................................................................................................................4<br />

General Session II: Risk Assessment, Prevention, Detection, and Screening<br />

Imaging Techniques for <strong>Breast</strong> <strong>Cancer</strong> Detection and Diagnosis: What Works and What Doesn’t<br />

David Mankoff, MD, PhD ................................................................................................................................7<br />

Surgical Risk Reduction for Atypical Ductal Hyperplasia, Lobular Carcinoma In Situ, and Ductal Carcinoma<br />

In Situ<br />

Todd M. Tuttle, MD ..........................................................................................................................................9<br />

Prevention Strategies for Women at High Risk and for Those with Lobular or Ductal Carcinoma In Situ<br />

Victor G. Vogel, MD, MHS .............................................................................................................................10<br />

General Session III: Controversies in Diagnosis of Early-Stage <strong>Breast</strong> <strong>Cancer</strong> and<br />

Management of Sentinel Nodes<br />

Magnetic Resonance Imaging Debate: Con<br />

Kathryn Evers, MD ........................................................................................................................................14<br />

General Session IV: Controversies in Local/Regional Treatment<br />

Debate on the Role of Local Radiation Therapy in Ductal Carcinoma In Situ: Pro<br />

Shivani Duggal, DO, and Thomas B. Julian, MD .......................................................................................17<br />

Targeted Intraoperative Radiotherapy: Analysis of a Randomized Trial—The TARGIT-A Trial<br />

Jayant S. Vaidya, MBBS, MS, DNB, PhD ....................................................................................................19<br />

Debate on the Role of Radiation Therapy in Ductal Carcinoma In Situ: Con<br />

Abram Recht, MD ...........................................................................................................................................22<br />

Intraoperative Radiation Therapy for <strong>Breast</strong> <strong>Cancer</strong><br />

Julia White, MD ..............................................................................................................................................25<br />

General Session V: Genomics for the Clinician<br />

PI3K in <strong>Breast</strong> <strong>Cancer</strong><br />

Toby M. Ward, PhD, Rebecca Olson, MS, Mark D. Pegram, MD, and Charles E. Geyer Jr., MD ........28<br />

Role of Genomic Testing in Patients with Node-Positive <strong>Breast</strong> <strong>Cancer</strong><br />

Shaheenah Dawood, MRCP, MPH, and Ana M. Gonzalez-Angulo, MD, MSc ........................................31<br />

<strong>Breast</strong> <strong>Cancer</strong> Whole Genome Sequencing<br />

Matthew J. Ellis, MB BChir, PhD ................................................................................................................34<br />

General Session VI: Health Care Delivery and Prevention<br />

<strong>Breast</strong> Health Care Delivery in Context: The World Health Organization’s Initiative for Noncommunicable Diseases<br />

Benjamin O. Anderson, MD ..........................................................................................................................36<br />

1


Reducing <strong>Breast</strong> <strong>Cancer</strong> Risk: Progress toward Resolution<br />

Rowan T. Chlebowski, MD, PhD ..................................................................................................................38<br />

General Session VII: Survivorship<br />

Inflammation in the Pathogenesis and Progression of <strong>Breast</strong> <strong>Cancer</strong><br />

Patrick G. Morris, MD, MSc, Kotha Subbaramaiah, PhD, Andrew J. Dannenberg, MD, and<br />

Clifford A. Hudis, MD ....................................................................................................................................41<br />

Managing Menopausal Symptoms in Patients with <strong>Breast</strong> <strong>Cancer</strong><br />

Patrick Neven, MD, and Anneleen Lintermans, MD ................................................................................44<br />

The Transition from Active Treatment to Normal Life as a Survivor<br />

Denise J. O’Neill, BS ......................................................................................................................................47<br />

General Session VIII: The Future of Health Policy<br />

What Are the Differences between Approving and Using New Drugs and New Devices in the United States?<br />

Gary H. Lyman, MD, MPH ............................................................................................................................50<br />

General Session IX: New Directions in Systemic Therapy for Advanced Disease<br />

Hormone-Responsive Disease<br />

Francisco J. Esteva, MD, PhD ......................................................................................................................53<br />

Therapeutic Strategies Targeting ERBB2: An Update on ERBB2-Positive <strong>Breast</strong> <strong>Cancer</strong><br />

Mark D. Pegram, MD .....................................................................................................................................55<br />

General Session XI: Debates on Current Controversies in Systemic Therapy<br />

Bone Health and the Role of Biphosphonates in Early-Stage <strong>Breast</strong> <strong>Cancer</strong>: A Debate<br />

Julie Gralow, MD, and Catherine Van Poznak, MD .................................................................................60<br />

In Favor of the Use of Anthracyclines in Patients with HER2-Positive and HER2-Negative <strong>Breast</strong> <strong>Cancer</strong><br />

Clifford Hudis, MD .........................................................................................................................................62<br />

Marker-Driven Personalized Therapy for <strong>Breast</strong> <strong>Cancer</strong><br />

Gabriel N. Hortobagyi, MD ...........................................................................................................................64<br />

“Just Say No” to Anthracyclines for Early <strong>Breast</strong> <strong>Cancer</strong><br />

Stephen Jones, MD ........................................................................................................................................66<br />

Reassessing Local Treatment in the Context of Increasingly Effective Systemic Therapy<br />

Jay R. Harris, MD ...........................................................................................................................................68<br />

2


GENERAL SESSION I:<br />

Controversial Topics in Medical Oncology Management<br />

CHAIR<br />

Lori Goldstein, MD<br />

Fox Chase <strong>Cancer</strong> Center<br />

SPEAKERS<br />

William John Gradishar, MD<br />

Northwestern University<br />

Robert W. Carlson, MD<br />

Stanford Comprehensive <strong>Cancer</strong> Center<br />

After this session, attendees should be able to<br />

● Assess the role of mTOR inhibitors in the management of metastatic breast cancer and determine potential patient<br />

benefits from targeting mTOR pathways<br />

● Discuss the potential applications for the use of chemotherapy for small TN and HER2-positive cancers and evaluate how<br />

this technique might be incorporated into treatment plans for specific patients


Role of Chemotherapy for Small Triple-Negative and<br />

HER2-Positive <strong>Cancer</strong>s<br />

<strong>Breast</strong> cancers encompass a diverse group of biologic<br />

subsets of disease that may be identified by biomarker<br />

expression (e.g., estrogen receptor [ER]/progesterone receptor<br />

[PR]/human epidermal growth factor receptor 2<br />

[HER2]) and genetic microarray analysis. The biologic heterogeneity<br />

among invasive breast cancers may be exploited<br />

in the treatment decision-making process. <strong>Breast</strong> cancers<br />

that are HER2-positive or triple-negative (ER-negative, PRnegative,<br />

and HER2-negative) have a worse prognosis than<br />

do the other subtypes. The HER2-positive breast cancers<br />

are highly responsive to HER2-targeted therapy (e.g., trastuzumab)<br />

and to selected chemotherapies (e.g., anthracyclines,<br />

taxanes, platinoids). The triple-negative breast<br />

cancers (TNBCs) also appear highly sensitive to selected<br />

chemotherapies (e.g., anthracyclines, taxanes, and platinoids)<br />

despite their overall relatively poor prognosis. 1<br />

The prognosis in women with T1a/bN0M0 disease has<br />

been the subject of multiple retrospective studies. The results<br />

of these studies consistently demonstrate favorable<br />

outcomes in women with T1a/bN0M0 disease as a group.<br />

Subset analyses to identify favorable or unfavorable biologic<br />

subsets within this cohort of patients have yielded inconsistent<br />

and sometimes contradictory results. 2-6 Some studies<br />

have found age and triple-negative status but not HER2<br />

status to be prognostic; others have found HER2-positive,<br />

endocrine receptor–negative disease or triple-negative status<br />

to be unfavorable; others have found HER2 status to be<br />

prognostic; and still others have found that HER2 status is<br />

not predictive of poor outcome in this anatomic subset.<br />

The activity of adjuvant trastuzumab added to chemotherapy<br />

has been studied in a number of randomized<br />

trials in high-risk women with HER2-positive early<br />

breast cancer. These studies demonstrate an approximate<br />

40% reduction in hazard of recurrence and 34%<br />

reduction in hazard of death with the addition of trastuzumab<br />

to adjuvant therapy. 7 In high-risk women with<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Robert W. Carlson, MD<br />

Stanford University, Stanford, CA<br />

Consultant or<br />

Advisory Role<br />

TNBC, the use of anthracycline- and taxane-containing<br />

adjuvant chemotherapy has demonstrated substantial<br />

improvement in relapse-free and overall survival. Generally,<br />

adjuvant therapy studies have been limited to<br />

patients with either node-positive disease or high-risk<br />

node-negative disease (grade 2 to 3, T � 1 cm). However,<br />

there are no data demonstrating that the relative<br />

risk reductions documented in high-risk populations<br />

should not also apply to patients in lower-risk populations.<br />

In the low-risk populations, the relative risk<br />

reduction in recurrences or deaths from disease may be<br />

large, while the absolute benefits may be small.<br />

The use of adjuvant HER2-targeted therapy and of cytotoxic<br />

chemotherapy is associated with acute and chronic<br />

toxicity, reduced quality of life, and financial expense. Some<br />

of the toxicities, such as second cancers, premature ovarian<br />

failure, and cardiac toxicity may be permanent. While the<br />

absolute efficacy benefits of the adjuvant therapies decrease<br />

with decreasing disease risk, the toxicities of adjuvant therapy<br />

are the same across all risk categories. The challenge is<br />

to assess when the benefits of adjuvant therapy become too<br />

small to justify the toxicity for the individual patient. In the<br />

extreme, do patients with T1a/bN0M0 HER2-positive or<br />

triple-negative disease derive sufficient benefit from adjuvant<br />

therapy to justify adjuvant treatment?<br />

The potential use of chemotherapy plus trastuzumab in<br />

the treatment of HER2-positive disease and of chemotherapy<br />

in the treatment of TNBC in women with T1a/bN0M0<br />

disease must be considered in the context of inconsistent/<br />

heterogeneous data on prognosis, absent clinical trials, and<br />

the known toxicities of treatment. 8 Further investigation to<br />

identify those patients most likely to experience recurrence<br />

of disease and thus have a relatively more acceptable efficacy<br />

to toxicity balance is required before definitive recommendations<br />

regarding adjuvant cytotoxic or HER2-targeted<br />

therapy may be made.<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Carlson, Robert W. AstraZeneca;<br />

Genentech;<br />

Sanofi<br />

References<br />

1. Hayes DF, Thor AD, Dressler LG, et al. HER2 and response to<br />

paclitaxel in node-positive breast cancer. N Engl J Med. 2007;357:<br />

1496-1506.<br />

2. Curigliano G, Viale G, Bagnardi V, et al. Clinical relevance of<br />

HER2 overexpression/amplification in patients with small tumor<br />

size and node-negative breast cancer. J Clin Oncol. 2009;27:<br />

5693-5699.<br />

4<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

3. Fehrenbacher L, Shiraz P, Sattavat M, et al. T1abN0M0 HER2�<br />

invasive breast cancer recurrence: Population-based cohort of<br />

17,000� consecutive breast cancers 2000-2006 at Kaiser Permanente<br />

Northern California (KPNC). J Clin Oncol. 2011;29:57s<br />

(suppl; abstr 551).<br />

4. Gonzalez-Angulo AM, Litton JK, Broglio KR, et al. High risk of<br />

recurrence for patients with breast cancer who have human epidermal<br />

growth factor receptor 2-positive, node-negative tumors 1<br />

cm or smaller. J Clin Oncol. 2009;27:5700-5706.


5. Chia S, Norris B, Speers C, et al. Human epidermal growth factor<br />

receptor 2 overexpression as a prognostic factor in a large tissue<br />

microarray series of node-negative breast cancers. J Clin Oncol.<br />

2008;26:5697-5704.<br />

6. Kwon J, Kim YJ, Lee KW, et al. Triple negativity and young age as<br />

prognostic factors in lymph node-negative invasive ductal carcinoma<br />

of 1 cm or less. BMC <strong>Cancer</strong>. 2010;10:557.<br />

EDUCATIONAL SUMMARIES<br />

7. Moja L, Tagliabue L, Balduzzi, et al. Trastuzumab containing regimens<br />

for early breast cancer. Cochrane Database Syst Rev. http://<br />

onlinelibrary.wiley.com/doi/10.1002/14651858.CD006243.pub2/<br />

pdf. Accessed June 8, <strong>2012</strong>.<br />

8. Carlson RW, Allred DC, Anderson BO, et al. NCCN Clinical<br />

Practice Guidelines: <strong>Breast</strong> <strong>Cancer</strong>. v1.<strong>2012</strong>. www.nccn.org. Accessed<br />

June 8, <strong>2012</strong>.<br />

5


GENERAL SESSION II:<br />

Risk Assessment, Prevention, Detection, and Screening<br />

CHAIR<br />

Banu Arun, MD<br />

University of Texas M. D. Anderson <strong>Cancer</strong> Center<br />

SPEAKERS<br />

Claudine Isaacs, MD<br />

Georgetown University<br />

David A. Mankoff, MD, PhD<br />

University of Pennsylvania<br />

Todd M. Tuttle, MD, MS<br />

University of Minnesota<br />

Victor Vogel, MD, MHS<br />

Geisinger Health System<br />

After this session, attendees should be able to<br />

● Appropriately use genetic testing to identify patients at high risk of cancer and employ strategies to clearly and<br />

effectively communicate results to patients and assist them in making informed decisions about any recommended next<br />

steps<br />

● Compare and contrast imaging techniques used in screening patients for cancer and determine what patient populations<br />

are most likely to benefit from each specific technique currently available<br />

● Examine both positive and negative implications of using risk-reducing surgery for patients with premalignant lesions<br />

and be able to clearly discuss all options with patients to assist them in making an informed decision<br />

● Evaluate different medical management strategies for patients with ductal carcinoma in situ (DCIS) or lobular<br />

carcinoma in situ (LCIS) as well as prevention strategies for women at risk, including pharmacologic interventions, such<br />

as results from MAP.3; determine those strategies’ potential to improve prevention outcomes in breast cancer


Imaging Techniques for <strong>Breast</strong> <strong>Cancer</strong> Detection and<br />

Diagnosis: What Works and What Doesn’t<br />

T<br />

he landscape for breast cancer detection and diagnosis<br />

has become more complex in recent years in part due<br />

to the introduction of new imaging methods. In addition to<br />

mammography and ultrasound, tests that have wellestablished<br />

roles in breast imaging, breast magnetic resonance<br />

imaging (MRI) has become an accepted and widely<br />

used tool for breast imaging. In addition, recent developments<br />

in instrumentation designed for breast imaging have<br />

led to interest in radionuclide breast imaging, namely<br />

breast-specific gamma imaging (BSGI) and positron emission<br />

mammography (PEM). In addition to breast cancer<br />

detection and diagnosis, these modalities have been shown,<br />

to varying degrees, to be helpful for defining the extent of<br />

disease in the breast (local breast staging) and for detecting<br />

regional nodal metastases (locoregional staging). This talk<br />

will provide a brief overview of how the imaging modalities<br />

particularly, the new modalities, work, and where they fit<br />

into the detection, diagnosis, and staging of breast cancer in<br />

current practice.<br />

<strong>Breast</strong> MRI<br />

<strong>Breast</strong> MRI works on the principle of imaging the magnetic<br />

resonance of protons perturbed by radiofrequency signal in<br />

the presence of a strong magnetic field. 1,2 <strong>Breast</strong> MRI uses<br />

dedicate coils to provide high quality, high-resolution images<br />

of the breast. MRI-guided biopsy methods have been<br />

established and are now widely available in practices with<br />

specialized breast imaging radiologists. 1,2 Detection and diagnosis<br />

requires the use of gadolinium-based contrast magnetic<br />

contrast; however, early work in diffusion-weighted<br />

imaging 3 may offer the ability to identify breast tumors<br />

without the need for contrast. <strong>Breast</strong> MRI has been most<br />

widely used for screening in high-risk women 4 and for local<br />

beast staging to guide surgical resection. 1 Some recent studies<br />

have questioned the influence of breast MRI for the<br />

David Mankoff, MD, PhD<br />

University of Pennsylvania, Philadelphia, PA<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Mankoff, David Bristol-Myers<br />

Squibb; Pfizer<br />

References<br />

1. Lehman CD, Schnall MD. Imaging in breast cancer: magnetic<br />

resonance imaging. <strong>Breast</strong> <strong>Cancer</strong> Res. 2005;7:215-219.<br />

2. Morrow M, Waters J, Morris E. MRI for breast cancer screening,<br />

diagnosis, and treatment. Lancet. 2011;378:1804-1811.<br />

3. Partridge SC, Mullins CD, Kurland BF, et al. Apparent diffusion<br />

coefficient values for discriminating benign and malignant<br />

breast MRI lesions: effects of lesion type and size. AJR Am J<br />

Roentgenol. 2010;194:1664-1673.<br />

latter application, 5 and studies under development should<br />

help further define its role in guiding breast surgery. 2<br />

Radiotracer Imaging<br />

Radiotracer imaging, also called nuclear medicine imaging,<br />

provides images of regional radiopharmaceutical concentration.<br />

Radiotracer imaging can use both standard gammaemitting<br />

tracers, single-photon emission computed<br />

tomography (SECT), and positron-emitting compounds<br />

(positron emission tomography [PET]). Radiotracer imaging<br />

plays an important role in the current care for patients<br />

with breast cancer, thus far it has been largely focused on<br />

staging for more advanced breast scans (bone scan and<br />

18<br />

F-fluorodexoyglucose [FDG] PET/computed tomography<br />

[CT]) and for guiding nodal sampling in sentinel lymph<br />

node biopsy. 6,7 Early studies of radiotracer breast imaging<br />

showed that it was useful for imaging mass lesions and<br />

evaluating response to therapy, 8,9 but had lower sensitivity<br />

for smaller, earlier-stage breast cancer. 8 More recently, the<br />

development of dedicated breast imaging devices for SPECT<br />

(breast-specific gamma imaging, BSGI) or PET (positron<br />

emission mammography, PEM) have shown promise detecting<br />

smaller lesions. 10-12 However, unlike breast MRI,<br />

the roles of BSGI and PEM in breast cancer diagnosis<br />

remain unclear. Early data from smaller studies, and some<br />

recent data from multicenter trials, point to reasonable and<br />

high specificity 10,11 ; however, the role of radiotracer breastspecific<br />

imaging methods vis-à-vis established methods such<br />

as mammography, ultrasound, and breast MRI will need to<br />

be defined. In the future, identification of molecular targets<br />

for early breast cancer detection, and translation of these<br />

targets into specific imaging probes, would greatly benefit<br />

radiotracer breast imaging methods and very likely improve<br />

upon the relatively nonspecific probes that are currently<br />

used. 13<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Merck; Pfizer<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

4. Saslow D, Boetes C, Burke W, et al. <strong>American</strong> <strong>Cancer</strong> Society<br />

guidelines for breast screening with MRI as an adjunct to mammography.<br />

CA <strong>Cancer</strong> J Clin. 2007;57:75-89.<br />

5. Turnbull L, Brown S, Harvey I, et al. Comparative effectiveness<br />

of MRI in breast cancer (COMICE) trial: a randomised controlled<br />

trial. Lancet. 2010;375:563-571.<br />

6. Benard F, Turcotte E. Imaging in breast cancer: Single-photon<br />

computed tomography and positron-emission tomography.<br />

<strong>Breast</strong> <strong>Cancer</strong> Res. 2005;7:153-162.<br />

7. Lee JH, Rosen EL, Mankoff DA. The role of radiotracer imaging<br />

7


EDUCATIONAL SUMMARIES<br />

in the diagnosis and management of patients with breast cancer:<br />

part 1—overview, detection, and staging. J Nucl Med. 2009;50:<br />

569-581.<br />

8. Khalkhali I, Villanueva-Meyer J, Edell SL, et al. Diagnostic<br />

accuracy of 99m Tc-sestamibi breast imaging: multicenter trial<br />

results. J Nucl Med. 2000;41:1973-1979.<br />

9. Mankoff DA, Dunnwald LK, Gralow JR, et al. Monitoring the<br />

response of patients with locally advanced breast carcinoma to<br />

neoadjuvant chemotherapy using [technetium 99m]-sestamibi<br />

scintimammography. <strong>Cancer</strong>. 1999;85:2410-2423.<br />

10. Berg WA, Madsen KS, Schilling K, et al. <strong>Breast</strong> cancer: comparative<br />

effectiveness of positron emission mammography and MR<br />

8<br />

imaging in presurgical planning for the ipsilateral breast. Radiology.<br />

2011;258:59-72.<br />

11. Brem RF, Floerke AC, Rapelyea JA, et al. <strong>Breast</strong>-specific gamma<br />

imaging as an adjunct imaging modality for the diagnosis of<br />

breast cancer. Radiology. 2008;247:651-657.<br />

12. Tadwalkar RV, Rapelyea JA, Torrente J, et al. <strong>Breast</strong>-specific<br />

gamma imaging as an adjunct modality for the diagnosis of<br />

invasive breast cancer with correlation to tumour size and grade.<br />

Br J Radiol. <strong>2012</strong>;85:e212–e216.<br />

13. Mankoff DA, Dunnwald LK, Kinahan P. Are we ready for<br />

dedicated breast imaging approaches? J Nucl Med. 2003;44:<br />

594-595.


Surgical Risk Reduction for Atypical Ductal Hyperplasia,<br />

Lobular Carcinoma In Situ, and Ductal Carcinoma In Situ<br />

With wide-spread screening mammography, high-risk<br />

breast lesions including atypical ductal hyperplasia<br />

(ADH) and lobular carcinoma in situ (LCIS) are increasingly<br />

diagnosed. These lesions are usually confirmed histologically<br />

with image-guided–core-needle biopsy. About 15%<br />

to 20% of patients with ADH or LCIS diagnosed using<br />

core-needle biopsy will have ductal carcinoma in situ (DCIS)<br />

or invasive cancer. 1 Because of this, excisional breast biopsy<br />

is usually recommended after core-needle biopsy to exclude<br />

a higher-stage lesion.<br />

Although ADH and LCIS are associated with an increased<br />

risk of breast cancer, surgical risk reduction is not frequently<br />

performed. The most commonly performed riskreduction<br />

procedure is bilateral skin-sparing mastectomy<br />

with immediate reconstruction, which is associated with a<br />

risk reduction of about 90%. 2 Surgical complications from<br />

bilateral mastectomy occur in about 20% of patients. The<br />

most common complications are infection, flap necrosis, and<br />

loss of reconstruction. Survey studies indicate that most<br />

patients are satisfied with their decision to undergo bilateral<br />

mastectomy.<br />

An increasing number of patients are undergoing surgical<br />

risk reduction using bilateral nipple-sparing mastectomies<br />

(NSM). Ideal candidates for NSM are younger patients,<br />

nonsmokers, and nonobese patients. Generally, NSMs are<br />

performed with either a radial or inframammary incision.<br />

The ductal tissue beneath the nipple is excised and submitted<br />

to pathology separately. If DCIS or invasive cancer is<br />

identified in this tissue, the nipple-areolar complex is excised.<br />

The cosmetic outcomes after NSM and reconstruction<br />

are excellent. Nipple necrosis can occur in about 5% of<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Tuttle, Todd M.*<br />

*No relevant relationships to disclose.<br />

Employment or<br />

Leadership<br />

Positions<br />

References<br />

Todd M. Tuttle, MD<br />

University of Minnesota, Minneapolis, MN<br />

Consultant or<br />

Advisory Role<br />

1. Foster MC, Helvie MA, Gregory NE, et al. Lobular carcinoma<br />

in situ or atypical lobular hyperplasia at core-needle biopsy: is<br />

excisional biopsy necessary? Radiology. 2004;231:813-819.<br />

2. Lostumbo L, Carbine NE, Wallace J. Prophylactic mastectomy for<br />

the prevention of breast cancer. Cochrane Database Syst Rev.<br />

http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD002748.<br />

pub3/abstract. Accessed June 25, <strong>2012</strong>.<br />

3. Fisher ER, Dignam J, Tan-Chiu E, et al. Pathologic findings from<br />

the National Surgical Adjuvant <strong>Breast</strong> Project (NSABP) eightyear<br />

update of Protocol B-17: intraductal carcinoma. <strong>Cancer</strong>.<br />

1999;86:429-438.<br />

patients. The occurrence of cancer in the nipple-areolar<br />

complex after NSM is extremely rare.<br />

Today, DCIS is usually detected by screening mammography<br />

and diagnosed by image-guided–core-needle biopsy.<br />

However, approximately 15% of patients with DCIS who are<br />

diagnosed by core-needle biopsy will have invasive cancer<br />

identified in the excision or mastectomy specimen. Local<br />

treatment options for DCIS include breast-conserving surgery<br />

(BCS) alone, BCS plus radiation, or mastectomy. Indications<br />

for mastectomy include multicentric disease, a large<br />

area of microcalcifications, inability to obtain negative surgical<br />

margins, and contraindications to radiation therapy.<br />

BCS without radiation therapy is primarily used for older<br />

patients or those with other significant health problems.<br />

Radiation therapy after breast-conserving surgery markedly<br />

reduces the risk of local recurrence.<br />

In NSABP B17, a negative surgical margin after BCS was<br />

defined as no ink on tumor. 3 Retrospective, single-center<br />

studies have reported lower local recurrence rates with<br />

wider margins. 4 A recent meta-analysis found no advantage<br />

with margins wider than 2 mm. 5 Presently, sentinel lymphnode<br />

biopsy is not routinely recommended for patients with<br />

DCIS; however, sentinel lymph-node biopsy should be considered<br />

for patients with microinvasion, palpable masses, or<br />

mastectomy treatment.<br />

Surgical risk reduction for DCIS is performance of a<br />

contralateral prophylactic mastectomy (CPM). For DCIS,<br />

the annual risk of developing metachronous contralateral<br />

breast cancer is about 0.5% to 0.6% per year. 6 The performance<br />

of CPM will significantly reduce the risk of contralateral<br />

breast cancer. The use of CPM in the United States for<br />

DCIS has markedly increased during the past decade. 7<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

4. Silverstein MJ, Lagios MD, Groshen S, et al. The influence of<br />

margin width on local control of ductal carcinoma in situ of the<br />

breast. N Engl J Med. 1999;340:1455-1461.<br />

5. Dunne C, Burke JP, Morrow M, et al. Effect of margin status on<br />

local recurrence after breast conservation and radiation therapy<br />

for ductal carcinoma in situ. J Clin Oncol. 2009;27:1615-1620.<br />

6. Gao X, Fisher SG, Emami B. Risk of second primary cancer in the<br />

contralateral breast in women treated for early-stage breast cancer:<br />

a population-based study. Int J Radiat Oncol Biol Phys.<br />

2003;56:1038-1045.<br />

7. Tuttle TM, Jarosek S, Habermann EB, et al. Increasing rates of<br />

contralateral prophylactic mastectomy among patients with ductal<br />

carcinoma in situ. J Clin Oncol. 2009;27:1362-1367.<br />

9


Prevention Strategies for Women at High Risk and for Those<br />

with Lobular or Ductal Carcinoma In Situ<br />

Tamoxifen is a selective estrogen-receptor modulator<br />

(SERM) and is approved by the U.S. Food and Drug<br />

Administration for breast cancer risk reduction in both<br />

premenopausal and postmenopausal women. Three phase<br />

III randomized trials prospectively evaluated tamoxifen<br />

compared with placebo for breast cancer risk reduction<br />

(Table 1). The combined reduction in invasive breast cancer<br />

and ductal carcinoma in situ (DCIS) with tamoxifen use<br />

compared with placebo was 34% to 38%. There was no<br />

reduction in the risk of estrogen receptor (ER)-negative<br />

breast cancer, but the incidence of ER-positive breast cancer<br />

decreased by 48%. Age had no apparent effect on the relative<br />

degree of breast cancer risk reduction. For noninvasive<br />

breast cancer occurring in the National Surgical Adjuvant<br />

<strong>Breast</strong> and Bowel Project (NSABP) <strong>Breast</strong> <strong>Cancer</strong> Prevention<br />

Trial (BCPT), the reduction in risk was 50%. Among<br />

women with a history of lobular carcinoma in situ (LCIS),<br />

the reduction in risk was 56%, and among women with a<br />

history of atypical hyperplasia, the reduction in risk was<br />

86%. 1-9<br />

The STAR trial randomly assigned 19,747 postmenopausal<br />

women with a 5-year increased risk of breast cancer<br />

to tamoxifen or raloxifene for 5 years (Fig. 1). The primary<br />

end point was a reduction in breast cancer risk. More than<br />

Table 1. Summary of Tamoxifen Chemoprevention Trials<br />

10<br />

Trial<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Prevention Trial<br />

(BCPT, NSABP<br />

P-1) 1,2<br />

International <strong>Breast</strong><br />

Intervention<br />

Study I 5<br />

Italian Tamoxifen<br />

Prevention<br />

Study 4<br />

All tamoxifen<br />

prevention trials 3<br />

Victor G. Vogel, MD, MHS<br />

Geisinger Health System, Danville, PA<br />

Subject<br />

Characteristics Population<br />

High breast cancer<br />

risk (age � 60<br />

years or a<br />

combination of<br />

risk factors using<br />

the Gail model);<br />

39% � 50 years<br />

Women 35 to 70<br />

years who were<br />

at increased risk<br />

for breast cancer<br />

Women with<br />

hysterectomy<br />

(48% bilateral<br />

oophorectomy);<br />

median age 51<br />

years<br />

� 1.66% 5-year<br />

risk<br />

� two-fold<br />

relative risk<br />

Normal risk,<br />

hysterectomy<br />

32% of STAR participants had a history of breast LCIS or<br />

atypical hyperplasia (AH) compared with 15% of NSABP-P1<br />

participants. The incidence of invasive breast cancer in the<br />

tamoxifen and raloxifene groups was not significantly<br />

different. 10-13 Patients with a history of LCIS or AH have a<br />

four-fold to 10-fold increased risk of subsequent invasive<br />

disease, and in the initial STAR report, tamoxifen and raloxifene<br />

were equally effective in reducing that risk. That is no<br />

longer the case for those with a history of atypical hyperplasia<br />

(relative risk [RR]: 1.48; 95% CI [1.06-2.09], comparing<br />

raloxifene with tamoxifen), although results for the<br />

LCIS group remain similar to those originally reported (RR:<br />

1.13; 95% CI [0.76-1.69]).<br />

In the PEARL trial, 8,556 postmenopausal women with<br />

low-bone density and normal mammograms were randomly<br />

assigned to lasofoxifene (0.25 mg and 0.5 mg) or placebo. 14<br />

The primary endpoints of the PEARL trial were incidence of<br />

ER-positive breast cancer and nonvertebral fractures at<br />

5 years. Compared with placebo, 0.5 mg of lasofoxifene<br />

reduced the risk of total breast cancer by 79% and ERpositive<br />

invasive breast cancer by 83% (hazard ratio [HR]:<br />

0.17; 95% CI [0.05-0.57]). One clinical trial examined the<br />

effect of an aromatase inhibitor on the risk of developing<br />

either invasive or noninvasive breast cancer. In the<br />

Number<br />

Randomized<br />

Intended<br />

Duration<br />

of<br />

Treatment<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Risk Reduction<br />

13,388 5 years Overall: 49%<br />

Ductal carcinoma in<br />

situ: 50%<br />

With prior lobular<br />

carcinoma in situ:<br />

55%<br />

With prior atypical<br />

hyperplasia: 86%<br />

7,152 5 years Overall: 32%<br />

5,408 5 years Overall: no reduction;<br />

in the high-risk<br />

subset, 82%<br />

28,442 Overall: 38%<br />

Estrogen receptorpositive,<br />

invasive:<br />

48%


Fig. 1. Forest plots of the risk of breast cancer from<br />

placebo-controlled trials of tamoxifen or raloxifene,<br />

with pooled estimates overall and for each treatment<br />

separately. 6<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Vogel, Victor G.*<br />

*No relevant relationships to disclose.<br />

Employment or<br />

Leadership<br />

Positions<br />

References<br />

Consultant or<br />

Advisory Role<br />

1. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for<br />

prevention of breast cancer: Report of the National Surgical<br />

Adjuvant <strong>Breast</strong> and Bowel Project P-1 Study. J Natl <strong>Cancer</strong><br />

Inst. 1998;90:1371-1388.<br />

2. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for the<br />

prevention of breast cancer: current status of the National Surgical<br />

Adjuvant <strong>Breast</strong> and Bowel Project P-1 Study. J Natl<br />

<strong>Cancer</strong> Inst. 2005;97:1652-1662.<br />

Canadian-based MAP3 trial, a total of 4,560 postmenopausal<br />

women at an increased risk of developing breast<br />

cancer were randomly assigned to either exemestane or<br />

placebo. 15 At 35 months, there was a 65% relative reduction<br />

in the annual incidence of invasive breast cancer, and the<br />

annual incidence of invasive plus noninvasive (DCIS) breast<br />

cancers was 0.35% on exemestane and 0.77% on placebo<br />

(HR: 0.47; 95% CI [0.27-0.79]; p�0.004). Combined LCIS<br />

and AH were reduced 64% in the exemestane group (HR:<br />

0.36; 95% CI [0.11-1.12]).<br />

ECOG study E5194 included 670 patients with DCIS<br />

treated with surgical excision without irradiation. The<br />

Oncotype DX assay was performed by quantitative reverse<br />

transcription polymerase chain reaction using<br />

formalin-fixed paraffin embedded tumor specimens<br />

from 49% of the patients. Recurrence score was calculated<br />

using the published algorithm. This multigene<br />

assay may provide individualized selection of treatment<br />

for patients with DCIS.<br />

A number of other drugs including retinoids, statins,<br />

NSAIDS, and COX-2 inhibitors are being investigated for<br />

their potential use in the reduction of breast cancer risk<br />

(including ER-negative disease), but none is yet approved<br />

for use outside of a clinical trial.<br />

Conclusion<br />

The ASCO Clinical Practice Guideline Update on the Use of<br />

Pharmacologic Interventions Including Tamoxifen, Raloxifene,<br />

and Aromatase Inhibition for <strong>Breast</strong> <strong>Cancer</strong> Risk<br />

Reduction published in 2009 recommends that tamoxifen<br />

be offered to reduce the risk of invasive ER-positive breast<br />

cancer in women who are at an increased risk for breast<br />

cancer, with benefits for at least 10 years. 16 In postmenopausal<br />

women, raloxifene may also be considered. Use of<br />

aromatase inhibitors or other SERMs to lower breast cancer<br />

risk is not recommended outside of a clinical trial. ASCO<br />

believes that discussion of risks and benefits of preventive<br />

agents by health providers is critical to patient decision<br />

making. 17-20<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

EDUCATIONAL SUMMARIES<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

3. Cuzick J, Powles T, Veronesi U, et al. Overview of the main<br />

outcomes in breast-cancer prevention trials. Lancet. 2003;361:<br />

296-300.<br />

4. Veronesi, U, Maisonneuve P, Rotmensz N, et al. Tamoxifen for<br />

the Prevention of <strong>Breast</strong> <strong>Cancer</strong>: late results of the Italian randomized<br />

tamoxifen prevention trial among women with hysterectomy.<br />

J Natl <strong>Cancer</strong> Inst. 2007;99:727-737.<br />

5. Cuzick J, Forbes JF, Sestak I, et al. Long-term results of tamoxifen<br />

prophylaxis for breast cancer: 96-month follow-up of the<br />

randomized IBIS-I trial. J Natl <strong>Cancer</strong> Inst. 2007;99:272-282.<br />

11


EDUCATIONAL SUMMARIES<br />

6. Cummings SR, Tice JA, Bauer S, et al. Prevention of breast<br />

cancer in postmenopausal women: approaches to estimating and<br />

reducing risk. J Natl <strong>Cancer</strong> Inst. 2009;101:384-398.<br />

7. Gail, MH, Costantino JP, Bryant J, et al. Weighing the risks and<br />

benefits of tamoxifen treatment for preventing breast cancer.<br />

J Natl <strong>Cancer</strong> Inst. 1999;91:1829-1846.<br />

8. Freedman AN, Costantino JP, Gail MH, et al. A benefit/risk<br />

assessment tool for breast cancer chemoprevention treatment.<br />

J Clin Oncol. 2011; 29:2327-2333.<br />

9. Goetz MP, Schaid DJ, Wickerham DL, et al. Evaluation of<br />

CYP2D6 and efficacy of tamoxifen and raloxifene in women<br />

treated for breast cancer chemoprevention: Results from the<br />

NSABP P-1 and P-2 clinical trials. Clin <strong>Cancer</strong> Res. 2011;17:<br />

6944-6951.<br />

10. Vogel VG, Costantino JP, Wickerham DL, et al. Effects of tamoxifen<br />

vs raloxifene on the risk of developing invasive breast cancer<br />

and other disease outcomes: The NSABP Study of tamoxifen and<br />

raloxifene (STAR) P-2 Trial. JAMA. 2006;295:2727-2741.<br />

11. Land SR, Wickerham DL, Costantino JP, et al. Patient-reported<br />

symptoms and quality of life during treatment with tamoxifen or<br />

raloxifene for breast cancer prevention: The NSABP study of<br />

tamoxifen and raloxifene (STAR) P-2 trial. JAMA. 2006;295:<br />

2742-2751.<br />

12. Vogel VG, Costantino JP, Wickerham DL, et al. Update of the<br />

NSABP study of tamoxifen and raloxifene (STAR) P-2 Trial:<br />

Preventing breast cancer. <strong>Cancer</strong> Prev Res. 2010;3:696-706.<br />

13. Vogel VG, Costantino JP, Wickerham DL, et al. Carcinoma in<br />

situ outcomes in National Surgical Adjuvant <strong>Breast</strong> and Bowel<br />

12<br />

Project <strong>Breast</strong> <strong>Cancer</strong> Chemoprevention Trials. J Natl <strong>Cancer</strong><br />

Inst Monographs. 2010;2010:181-186.<br />

14. LaCroix AZ, Powles T, Osborne CK, et al. <strong>Breast</strong> cancer incidence<br />

in the randomized PEARL trial of lasofoxifene in postmenopausal<br />

osteoporotic women. J Natl <strong>Cancer</strong> Inst. 2010;102:<br />

1706-1717.<br />

15. Goss PE, Ingle JN, Alés-Martínez JE, et al. Exemestane for<br />

breast-cancer prevention in postmenopausal women. N Engl<br />

J Med. 2011;364:2381-2391.<br />

16. Visvanathan K, Chlebowski RT, Hurley P, et al. <strong>American</strong> Society<br />

of Clinical Oncology practice guideline update on the use of<br />

pharmacologic intervention including tamoxifen, raloxifene, and<br />

aromatase inhibition for breast cancer risk reduction. J Clin<br />

Oncol. 2009;27:3235-3258.<br />

17. Vogel VG. Managing the risk of invasive breast cancer in women<br />

at risk for breast cancer and osteoporosis: The role of raloxifene.<br />

Clin Interv Aging. 2008;3:609-609.<br />

18. Zon RT, Goss E, Vogel VG, et al. <strong>American</strong> Society of Clinical<br />

Oncology Policy Statement: The Role of the Oncologist in <strong>Cancer</strong><br />

Prevention and Risk Assessment. J Clin Oncol. 2009;27:986-993.<br />

19. Cuzick J. DeCensi A, Arun B, et al. Preventive therapy for breast<br />

cancer: An international consensus statement. Lancet Oncol.<br />

2011;12:496-503.<br />

20. Rastogi P, Lo S, Vogel VG. Chemoprevention: clinical aspects. In:<br />

Harris J, Lippman M, Morrow M, Osborne CK (eds). Diseases of<br />

the <strong>Breast</strong>, 4th edition. Philadelphia, J.B. Lippincott, 2010;302-<br />

319.


GENERAL SESSION III:<br />

Controversies in Diagnosis of Early-Stage <strong>Breast</strong> <strong>Cancer</strong> and<br />

Management of Sentinel Nodes<br />

CO-CHAIRS<br />

Jennifer Bellon, MD<br />

Dana-Farber <strong>Cancer</strong> Institute<br />

Richard Bleicher, MD<br />

Fox Chase <strong>Cancer</strong> Center<br />

SPEAKERS<br />

Kathryn Evers, MD<br />

Fox Chase <strong>Cancer</strong> Center<br />

Constance D. Lehman, MD, PhD<br />

Seattle <strong>Cancer</strong> Care Alliance<br />

After this session, attendees should be able to<br />

● Assess the pros and cons of routine breast magnetic resonance imaging use for the diagnosis and preoperative evaluation<br />

of the non–high-risk patient with breast cancer<br />

● Compare and contrast the Z0011 surgical and radiation approaches to nodal management of sentinel node positive<br />

disease with MA.20, considering the unique patient characteristics that would impact management decisions


Magnetic Resonance Imaging Debate: Con<br />

T<br />

he role of magnetic resonance imaging (MRI) in the<br />

diagnosis and evaluation of breast carcinoma continues<br />

to evolve. Contrast-enhanced breast MRI is an extremely<br />

sensitive tool for the diagnosis of carcinoma, and<br />

the role of MRI in women at high risk for the development<br />

of breast carcinoma is well established at this time. Additionally,<br />

MRI is extremely useful in the scenario of proven<br />

axillary metastasis with no primary tumor visible on mammography<br />

or ultrasound. In this case, demonstration of<br />

the primary lesion can potentially allow the patient to have<br />

breast-conserving therapy. Similar benefits can be seen in<br />

patients with Paget’s disease of the nipple. In women with<br />

dense breasts, MRI is considerably more sensitive than<br />

mammography.<br />

For the evaluation of the patient with a new diagnosis<br />

of breast carcinoma, the role of MRI remains considerably<br />

more controversial. At this time in many practices, treatment<br />

planning for patients with newly diagnosed carcinoma<br />

is the most common reason for diagnostic breast MRI. 1 This<br />

occurs despite the lack of strong evidence for the utility of<br />

MRI in this setting.<br />

There is good evidence that MRI is more accurate than<br />

physical examination, mammography, or ultrasound for determining<br />

tumor size. Additionally, the presence or absence<br />

of invasion of the pectoralis muscle or the chest wall can be<br />

accurately assessed. Skin thickening, which may be subtle<br />

on physical examination, may be identified on breast MRI.<br />

The fact that MRI is accurate at estimating tumor size has<br />

been extended to mean that the use of MRI will allow a<br />

decrease in close or positive surgical margins. Initially,<br />

there was considerable enthusiasm for using MRI to increase<br />

the rate of negative margins at lumpectomy and to<br />

decrease the number of second procedures or conversions<br />

to mastectomy required for patients scheduled for breastconserving<br />

therapy.<br />

Significant improvement in the rate of second procedures<br />

for patients who are potential candidates for breastconserving<br />

therapy is an ambitious undertaking, since historical<br />

success rates for this procedure are 90% to 97%. 2,4<br />

Several studies have found no significant improvement in<br />

margin status in women undergoing preoperative MRI. 5,6 A<br />

recent meta-analysis 7 reports that foci of disease visible only<br />

on MRI are found in 16% of cases, with appropriate conversion<br />

to more extensive surgery in approximately 11%<br />

of cases (3% more extensive resection and 8% conversion<br />

to mastectomy). Conversion to more extensive treatment<br />

occurred in approximately 5% of patients based on MRI<br />

findings where no additional malignancy was found at pathology.<br />

The overall false–positive rate has been reported as<br />

1% to 28%. Given these statistics, it is imperative that<br />

additional findings on MRI be confirmed with core biopsy<br />

prior to surgery (Fig. 1).<br />

The largest prospective, randomized controlled trial evaluating<br />

the effectiveness of MRI is the Comparative Effec-<br />

14<br />

Kathryn Evers, MD<br />

Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA<br />

Fig. 1. Multiple areas of enhancement in a patient<br />

with known breast carcinoma. Shown is a single<br />

subtraction image of the initial postcontrast image<br />

in a patient sent for surgical treatment planning.<br />

The patient’s known primary tumor and a satellite<br />

mass (arrows) in the upper right breast are shown.<br />

A small enhancing mass in the lower right breast<br />

(arrowhead) proved to be a fibroadenoma on core<br />

biopsy.<br />

tiveness of MRI in <strong>Breast</strong> <strong>Cancer</strong> (COMICE) trial, 8 which<br />

was performed in Great Britain. In this trial, patients who<br />

were scheduled for wide excisions for breast carcinoma were<br />

randomly assigned to receive or not receive MRI preoperatively.<br />

The results showed that patients in the MRI arm had<br />

an equal number of re-excisions or conversion to mastectomy<br />

as the patients in the non-MRI arm. Moreover, in 58<br />

cases where there were abnormal results, 27.6% of patients<br />

had a mastectomy that was felt to be avoidable based on<br />

pathology results.<br />

The Dutch MR Mammography of Nonpalpable <strong>Breast</strong><br />

Tumors (MONET) trial 9 randomly assigned patients with<br />

suspicious imaging (BI-RADS 3, 4, and 5) to MRI and non-<br />

MRI arms. The endpoint of this trial was the rate of additional<br />

surgical procedures (re-excision and conversion to<br />

mastectomy). In this trial, MRI was associated with an<br />

increased re-excision rate for positive margins, although it<br />

was not statistically significant. It has been noted that there<br />

is an increased use of mastectomy, stage for stage, with the<br />

use of MRI. This has been reported as a 20% to 30% unnecessary<br />

mastectomy rate 10 or as a stage-specific likelihood of<br />

undergoing mastectomy approximately 1.8 times more than<br />

patients who did not have MRI. 11


Small foci of additional disease appear to be adequately<br />

treated in patients who have had lumpectomy and wholebreast<br />

radiation therapy because the recurrence and survival<br />

rates are equivalent to that of patients treated with<br />

mastectomy. Therefore, the utility of detecting additional<br />

small foci of disease is unclear. The same is not true for<br />

patients receiving accelerated partial-breast irradiation, as<br />

additional small foci may not be adequately treated.<br />

MRI is also capable of identifying occult carcinoma in the<br />

contralateral breast. In 2007, Lehman 12 reported a 3.1%<br />

rate of contralateral carcinoma found only by MRI. The<br />

false–positive rate remains relatively high in this regard<br />

as recommendations for biopsy in this setting range from<br />

13% to 32%, 13 although this is somewhat lower in more<br />

recent studies. Relapse in the contralateral breast is low,<br />

reported as 2% to 4% over 5 years. A study by Solin in 2008 14<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Evers, Kathryn*<br />

*No relevant relationships to disclose.<br />

Employment or<br />

Leadership<br />

Positions<br />

References<br />

Consultant or<br />

Advisory Role<br />

1. Bassett LW, Dhaliwal SG, Eradat J, et. al. National trends and<br />

practices in breast MRI. AJR Am J Roentgenol. 2008;191:332-<br />

339.<br />

2. Fisher B, Bauer M, Margolese R, et al. Five-year results of a<br />

randomized clinical trial comparing total mastectomy and segmental<br />

mastectomy with or without radiation in the treatment of<br />

breast cancer. N Engl J Med. 1985;312:665-673.<br />

3. Blichert-Toft M, Rose C, Andersen JA, et al. Danish randomized<br />

trial comparing breast conservation therapy with mastectomy:<br />

six years of life-table analysis. Danish <strong>Breast</strong> <strong>Cancer</strong> Cooperative<br />

Group. J Natl <strong>Cancer</strong> Inst Monogr. 1992:11:19-25.<br />

4. Morrow M, Strom E, Bassett LW, et al. Standard for breast<br />

conservation therapy in the management of invasive breast carcinoma.<br />

CA <strong>Cancer</strong> J Clin. 2002;52:277-300.<br />

5. Bleicher RJ, Ciocca RM, Egleston BL, et al. Association of routine<br />

pretreatment magnetic resonance imaging with time to surgery,<br />

mastectomy rate and margin status. J Am Coll Surg. 2009;<br />

209:180-187.<br />

6. Pengel KE, Loo CE, Teertstra HJ, et al. The impact of preoperative<br />

MRI on breast-conserving surgery of invasive cancer: a<br />

comparative cohort study. <strong>Breast</strong> <strong>Cancer</strong> Res Treat. 2009;116:<br />

161-169.<br />

7. Houssami N, Ciatto S, Macaskill P, et al. Accuracy and surgical<br />

impact of magnetic resonance imaging in breast cancer staging:<br />

Systematic review and meta-analysis in detection of multifocal<br />

and multicentric cancer. J Clin Oncol. 2008;26:3248-3258.<br />

8. Turnbull LW, Brown SR, Olivier C, et al. Multicentre randomised<br />

controlled trial examining the cost-effectiveness of<br />

showed no change in the contralateral breast cancer rate<br />

of patients who have or have not had breast MRI. It is<br />

unclear whether detection of these tumors will influence<br />

survival.<br />

To date, there is no evidence that the use of preoperative<br />

MRI will result in improved survival. Solin 14 reported no<br />

change in 8-year survival. A study 1 year later reported a<br />

nonstatistically significant trend towards improved survival.<br />

15 Neither of these trials demonstrate sufficient<br />

follow-up that could evaluate whether or not MRI will effect<br />

survival.<br />

In conclusion, breast MRI is a powerful tool with numerous<br />

indications. At this time, its usefulness in the setting<br />

of newly diagnosed breast cancer is unproven with no demonstrable<br />

benefit in decreasing reoperation or improving<br />

survival.<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

EDUCATIONAL SUMMARIES<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

contrast-enhanced high field magnetic resonance imaging in<br />

women with primary breast cancer scheduled for wide local<br />

excision (COMICE). Health Technol Asses. 2010;14:1-182.<br />

9. Peters NH, van Esser S, van den Bosch MA, et al. Preoperative<br />

MRI and surgical management in patients with nonpalpable<br />

breast cancer: The MONET-Randomised controlled trial. Eur J<br />

<strong>Cancer</strong>. 2011;42:879-886.<br />

10. Bedrosian I, Mick R, Orel SG, et al. Changes in the surgical<br />

management of patients with breast carcinoma based on preoperative<br />

magnetic resonance imaging. <strong>Cancer</strong>. 2003;98:468-473.<br />

11. Katipamula R ,Degnim AC, Hoskin T, et al. Trends in mastectomy<br />

rates at the Mayo Clinic Rochester: effect of surgical year<br />

and preoperative magnetic resonance imaging. J Clin Oncol.<br />

2009;27:4082-4088.<br />

12. Lehman CD, Gatsonis C, Kuhl CK, et al. MRI evaluation of the<br />

contralateral breast in women with recently diagnosed breast<br />

cancer. N Engl J Med. 2007;356:1295-1303.<br />

13. Liberman L, Morris EA, Kim CM, et al. MR imaging findings in<br />

the contralateral breast of women with recently diagnosed breast<br />

cancer. AJR Am J Roentgenol. 2003;180:333-341.<br />

14. Solin LJ, Orel SG, Hwang WT, et al. Relationship of breast<br />

magnetic resonance imaging to outcome after breastconservation<br />

treatment with radiation for women with earlystage<br />

invasive breast carcinoma or ductal carcinoma in situ.<br />

J Clin Oncol. 2008;26:386-391.<br />

15. Hwang WT, Schiller DE, Crystal P, et al. Magnetic resonance<br />

imaging in the planning of initial lumpectomy for invasive breast<br />

carcinoma: its effect on ipsilateral breast tumor recurrence after<br />

breast-conservation therapy. Ann Surg Oncol. 2009;16:3000-<br />

3009.<br />

15


GENERAL SESSION IV:<br />

Controversies in Local/Regional Treatment<br />

CO-CHAIRS<br />

Robert Kuske, MD<br />

Arizona <strong>Breast</strong> <strong>Cancer</strong> Specialists<br />

Lori J. Pierce, MD<br />

University of Michigan School of Medicine<br />

SPEAKERS<br />

Thomas B. Julian, MD<br />

National Surgical Adjuvant <strong>Breast</strong> and Bowel Project (NSABP)<br />

Abram Recht, MD<br />

Harvard Medical School and Beth Israel Deaconess Medical Center<br />

Jayant S. Vaidya, PhD<br />

University College London<br />

Julia White, MD<br />

The Ohio State University Comprehensive <strong>Cancer</strong> Center<br />

After this session, attendees should be able to<br />

● Examine the outcomes of a randomized trial using intraoperative radiation therapy and determine how the results of the<br />

trial may impact the use of this therapy in the treatment of patients with breast cancer<br />

● Review the use of intraoperative radiation therapy as a treatment option for patients with breast cancer from the<br />

perspective of potential benefits, such as preservation of breast tissue and shortened treatment periods<br />

● Investigate the use of local radiation therapy in the treatment of patients with ductal carcinoma in situ, including how<br />

this treatment option impacts disease pathology and ultimately patient outcomes


Debate on the Role of Local Radiation Therapy in Ductal<br />

Carcinoma In Situ: Pro<br />

Shivani Duggal, DO, and Thomas B. Julian, MD<br />

Allegheny <strong>Cancer</strong> Center at Allegheny General Hospital and the National Surgical<br />

Adjuvant <strong>Breast</strong> and Bowel Project (NSABP), Pittsburgh, PA<br />

T<br />

he incidence of ductal carcinoma in situ (DCIS) has<br />

seen a dramatic rise in the United States, particularly<br />

over the last 3 decades. This increase parallels improvements<br />

in imaging techniques. Since the utilization of<br />

screening mammography in the 1980s, DCIS now accounts<br />

for 14% to 30% of all diagnosed breast cancers; before such<br />

screening, DCIS was a rare entity, representing 2% of reported<br />

breast cancers. 1 Today, 1 in 1,300 mammograms will<br />

result in the detection of DCIS. 2<br />

As the incidence of DCIS rises, so does the need for<br />

improved treatment strategies. In-breast tumor recurrence<br />

(IBTR) remains the most common first failure event in the<br />

management of DCIS. While mastectomy was once the<br />

standard treatment for DCIS, with recurrence rates as low<br />

as 1.4%, 3,4 breast-conserving surgery (BCS) and the use of<br />

radiation therapy (RT) has now gained widespread acceptance<br />

as an alternative approach. While BCS and RT has<br />

been shown to be effective in local control, it is the adjuvant<br />

RT treatment following BCS for DCIS that remains controversial.<br />

Because the decision to add RT after surgical treatment<br />

of DCIS is not straightforward, there is significant<br />

heterogeneity in the management of DCIS. A populationbased<br />

analysis 5 has shown that there is regional variation<br />

in the use of adjuvant RT; approximately 50% of patients<br />

do not receive RT for DCIS after BCS. This variation may<br />

be due to differences in the perceived risk of recurrence.<br />

The phase III, randomized National Surgical Adjuvant<br />

<strong>Breast</strong> and Bowel Project (NSABP) B-17 6,7 clinical trial<br />

along with three other prospective randomized clinical<br />

trials 8-10 and their follow-up studies, 11,12 have demonstrated<br />

and continue to show the benefit of RT following<br />

segmentectomy, with a 50% reduction in local recurrence<br />

rates for both DCIS and invasive breast cancer. In a report<br />

by Wapnir and colleagues on long-term outcomes of the<br />

NSABP B-17 and B-24 trials, patients who only received<br />

lumpectomy had a local recurrence rate of 19.4% compared<br />

with 8.9% in the lumpectomy-positive RT group at 15 years<br />

of follow up, demonstrating a 52% reduction in the risk of<br />

IBTR. 13<br />

Studies have also been conducted based on the premise<br />

that we may be overtreating patients with DCIS. The Eastern<br />

Cooperative Oncology Group (ECOG) E5194 trial 14 prospectively<br />

evaluated clinical features to identify a low-risk<br />

Authors’ Disclosures of Potential Conflicts of Interest<br />

population with DCIS in whom BCS alone was adequate. To<br />

the disappointment of the study investigators, 20% of these<br />

patients had local recurrence. The negative outcome of<br />

E5194 eventually provided useful insight about the clinical<br />

application of and benefits from RT. A follow-up study used<br />

adjuvant whole-breast irradiation (WBI) in patients with<br />

DCIS who met the criteria of E5194. Results showed that<br />

WBI reduced the rate of local recurrence in this patient<br />

population by more than 70%. 15 Similarly, Wong and colleagues<br />

attempted to demonstrate that wide excision with<br />

margins larger than or equal to 1 cm alone was adequate in<br />

the treatment of small grade 1 or 2 DCIS. This prospective,<br />

single-arm clinical trial closed early to accrual because the<br />

number of local recurrences met the established stopping<br />

criteria of the study. The study concluded that despite wide<br />

margins, the local recurrence rate of DCIS after excision<br />

alone is substantial enough to warrant adjuvant therapy<br />

with radiation with or without antihormonal therapy. 16<br />

With such compelling data supporting the need for RT<br />

in the treatment of DCIS, attention may be better focused<br />

on the various modalities of RT offered today, rather than<br />

on whether to implement it or not. These include hyperand<br />

hypofractionated WBI and accelerated partial-breast<br />

irradiation (APBI), which is comprised of catheter-based<br />

brachytherapy, three-dimensional conformal externalbeam<br />

irradiation, or single-fraction intraoperative partialbreast<br />

irradiation. The majority of early data on APBI is<br />

encouraging, with local failure rates similar to those of<br />

conventional WBI, 17 along with the added benefits of<br />

shorter duration of therapy, reduced local and chronic toxicity,<br />

and improved cosmesis and quality of life (QOL). The<br />

phase III prospective randomized NSABP B-39/Radiation<br />

Therapy Oncology Group (RTOG) 0413 trial evaluating the<br />

effectiveness of APBI compared with WBI following BCS for<br />

DCIS as well as early-stage invasive breast cancer will further<br />

explore the benefits of APBI. That trial will evaluate<br />

IBTR, overall survival, disease-free survival, distant DFS,<br />

QOL issues, and the perceived convenience of care. 18<br />

Until we have identified predictive factors to tailor treatment<br />

for individual patients, all patients with DCIS are<br />

candidates for radiation therapy because we cannot yet<br />

clearly identify those in whom it is not beneficial.<br />

Employment or<br />

Leadership Consultant or Stock<br />

Research<br />

Author<br />

Positions Advisory Role Ownership Honoraria Funding<br />

Duggal, Shivani<br />

Julian, Thomas B.*<br />

*No relevant relationships to disclose.<br />

Komen Foundation<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

17


EDUCATIONAL SUMMARIES<br />

References<br />

1. Baxter NN, Virnig BA, Durham SB, et al. Trends in the treatment<br />

of ductal carcinoma in situ of the breast. J Natl <strong>Cancer</strong> Inst.<br />

2004;96:443-448.<br />

2. Ernster VL, Ballard-Barbash R, Barlow WE, et al. Detection of<br />

ductal carcinoma in situ in women undergoing screening mammography.<br />

J Natl <strong>Cancer</strong> Inst. 2002;94:1546-1554.<br />

3. Boyages J, Delaney G, Taylor R. Predictors of local recurrence<br />

after treatment of ductal carcinoma in situ: a meta-analysis.<br />

<strong>Cancer</strong>. 1999;85:616-628.<br />

4. Rashtian A, Iganej S, Liu IA, et al. Close or positive margins after<br />

mastectomy for DCIS: Pattern of relapse and potential indications<br />

for radiotherapy. Int J Radiation Oncology Biol Phys. 2008;72:<br />

1016-1020.<br />

5. Rakovitch E, Pignol JP, Chartier C, et al. The management of<br />

ductal carcinoma in situ of the breast: A screened populationbased<br />

analysis. <strong>Breast</strong> <strong>Cancer</strong> Res Treat. 2007;101:335-347.<br />

6. Fisher B, Costantino J, Redmond C, et al. Lumpectomy compared<br />

with lumpectomy and radiation therapy for the treatment of<br />

intraductal breast cancer. N Engl J Med. 1993;328:1581-1586.<br />

7. Fisher B, Dignam J, Wolmark N, et al. Lumpectomy and radiation<br />

therapy for the treatment of intraductal breast cancer: findings<br />

from National Surgical Adjuvant <strong>Breast</strong> and Bowel Project B-17.<br />

J Clin Oncol. 1998;16:441-452.<br />

8. EORTC <strong>Breast</strong> <strong>Cancer</strong> Cooperative Group and EORTC Radiotherapy<br />

Group; Bijker N, Meijnen P, et al. <strong>Breast</strong>-conserving<br />

treatment with or without radiotherapy in ductal carcinoma-insitu:<br />

ten-year results of European Organisation for Research and<br />

Treatment of <strong>Cancer</strong> randomized phase III trial 10853—a study<br />

by the EORTC <strong>Breast</strong> <strong>Cancer</strong> Cooperative Group and EORTC<br />

Radiotherapy Group. J Clin Oncol. 2006;24:3381-3387.<br />

9. Houghton J, George WD, Cuzick J, et al. UK Coordinating Committee<br />

on <strong>Cancer</strong> Research; Ductal Carcinoma in situ Working<br />

Party; DCIS trialists in the UK, Australia, and New Zealand.<br />

Radiotherapy and tamoxifen in women with completely excised<br />

ductal carcinoma in situ of the breast in the UK, Australia, and<br />

18<br />

New Zealand: Randomised controlled trial. Lancet. 2003;362:95-<br />

102.<br />

10. Holmberg L, Garmo H, Granstrand B, et al. Absolute risk reductions<br />

for local recurrence after postoperative radiotherapy after<br />

sector resection for ductal carcinoma in situ of the breast. J Clin<br />

Oncol. 2008;26:1247-1252.<br />

11. Solin, LJ. The impact of adding radiation treatment after breast<br />

conservation surgery for ductal carcinoma in situ of the breast.<br />

J Natl <strong>Cancer</strong> Inst Monogr. 2010;41:187-192.<br />

12. Bijker N, van Tienhoven G. Local and systemic outcomes in<br />

DCIS based on tumor and patient characteristics: The radiation<br />

oncologist’s perspective. J Natl <strong>Cancer</strong> Inst Monogr. 2010;2010:<br />

178-180.<br />

13. Wapnir IL, Dignam JJ, Fisher B, et al. Long-term outcomes of<br />

invasive ipsilateral breast tumor recurrences after lumpectomy<br />

in NSABP B-17 and B-24 randomized clinical trials for DCIS.<br />

J Natl <strong>Cancer</strong> Inst. 2011;103:478-488.<br />

14. Hughes LL, Wang M, Page DL, et al. Local excision alone without<br />

irradiation for ductal carcinoma in situ of the breast: A trial of<br />

the Eastern Cooperative Oncology Group. J Clin Oncol. 2009;27:<br />

5319-5324.<br />

15. Motwani SB, Goyal S, Moran MS, et al. Ductal carcinoma in situ<br />

treated with breast-conserving surgery and radiotherapy: a comparison<br />

with ECOG Study 5194. <strong>Cancer</strong>. 2011;117:1156-1162.<br />

16. Wong JS, Kaelin CM, Troyan SL, et al. Prospective study of wide<br />

excision alone for ductal carcinoma in situ of the breast. J Clin<br />

Oncol. 2006;24:1031-1036.<br />

17. Vicini F, Beitsch P, Quiet C, et al. Five-year analysis of treatment<br />

efficacy and cosmesis by the <strong>American</strong> Society of <strong>Breast</strong> Surgeons<br />

MammoSite <strong>Breast</strong> Brachytherapy Registry Trial in patients<br />

treated with accelerated partial breast irradiation. Int J<br />

Radiat Oncol Biol Phys. 2011;79:808-817.<br />

18. Arthur DW, Kuerer HM, Haffty B, et al. A phase II study of<br />

repeat breast preserving surgery and 3D-conformal partial<br />

breast re-irradiation (PBRI) for local recurrence of breast carcinoma.<br />

Radiation Therapy Oncology Group (RTOG) 1014. http://<br />

rpc.mdanderson.org/rpc/credentialing/files/1014.pdf. Accessed<br />

May 24, <strong>2012</strong>.


Targeted Intraoperative Radiotherapy: Analysis of a<br />

Randomized Trial—The TARGIT-A Trial<br />

Jayant S. Vaidya, MBBS, MS, DNB, PhD<br />

University College London, London, United Kingdom<br />

On behalf of the TARGIT Trialists Group<br />

We have previously published preclinical, 1,2 clinical, 3-5<br />

radiobiological, and translational 6-8 studies that support<br />

the use of targeted intraoperative radiotherapy<br />

(TARGIT). In June 2010, we published the results of the<br />

TARGIT-A multicenter, randomized clinical trial 9 that<br />

compared “one-size-fits-all” radiotherapy (whole breast<br />

radiotherapy-external beam radiotherapy [EBRT] group)<br />

with risk-adjusted radiotherapy (single-dose TARGIT with<br />

additional whole breast radiotherapy only if adverse prognostic<br />

factors were found).<br />

Trial eligibility criteria are described previously; 9 briefly,<br />

patient were eligible if they were older than 45 and had<br />

invasive duct carcinoma less than approximately 3.5 cm<br />

that was suitable for breast-conserving surgery. In the trial,<br />

82% of patients were less than age 70, and 64% of tumors<br />

were more than 1 cm; 85% of tumors were less than 2 cm<br />

and grades 1 or 2; 83% were node negative; and 90% were<br />

ER positive. Within the TARGIT group, whole breast radiotherapy<br />

was recommended if margins were positive (� 1<br />

mm) at therapeutic excision before giving intraoperative<br />

radiotherapy, extensive in situ component, or unexpected<br />

invasive lobular carcinoma, or other adverse prognostic factors<br />

(such as multiple positive nodes, lymphovascular invasion,<br />

etc.) as predefined in each center’s treatment policy<br />

document. In such cases, TARGIT served as a tumor-bed<br />

boost. We concluded that the risk of local recurrence in the<br />

TARGIT group was noninferior to that of the EBRT group<br />

(the difference between the two arms was 0.25% at 4 years).<br />

When the TARGIT-A trial was launched internationally,<br />

10 we expected the background recurrence rate to be<br />

6%. We chose to use an absolute difference in local recurrence,<br />

2.5%, as a noninferiority margin. The sample size<br />

needed was 2,232 patients. However, over the last decade,<br />

the local recurrence rates of breast cancer have plummeted;<br />

the Standardisation of <strong>Breast</strong> Radiotherapy (START) trial<br />

reported a local recurrence rate of 2.3%, 11 and when we<br />

analyzed the data for the TARGIT-A trial, we found that the<br />

Kaplan-Meier estimate of local recurrence at 5 years was<br />

1.5%. In the main paper 9 and in subsequent correspondence,<br />

12 we have discussed the rationale for retaining the<br />

absolute difference of 2.5% as an acceptable noninferiority<br />

margin.<br />

In addition, we discussed the issue of absolute versus<br />

relative risks. It was suggested that risk ratios should be<br />

used for our power calculations. However, when the recurrence<br />

rates are so low, risk ratio is less informative at best<br />

and misleading at worst. In any case, power calculations<br />

fade into insignificance when faced with the reality of our<br />

observations. We found that the actual difference between<br />

the local recurrence rate of the two groups was 0.25%<br />

(1.2% vs. 0.95%, p � 0.41), which is 10 times lower than our<br />

original margin of noninferiority.<br />

Furthermore, one can estimate the effect of a 2.5% absolute<br />

difference in local recurrence at 5 years, on breast<br />

cancer mortality at 15 years, using the 1:4 ratio derived<br />

from the Oxford overview. Thus, a 2.5% difference at 5 years<br />

can be expected to make a difference of 0.625% in mortality<br />

at 15 years, arguably a value that is clinically acceptable. In<br />

fact, we estimated 9 that the number of patients needed to<br />

detect a 2.5% difference with a background of 1.5% was<br />

much smaller than we had recruited for. We would have<br />

only needed 585 patients to have adequate power to detect<br />

the difference between the two arms of the trial. When this<br />

number (585) of patients reach a minimum 5 years of followup,<br />

and if the difference between the two treatment arms is<br />

less than 2.5%, we could be confident of the robustness of a<br />

noninferior result.<br />

What is the absolute difference that is acceptable? Hitherto<br />

in the TARGIT-A trial, this decision was based on an<br />

“intelligent and experienced” guess by the team of experienced<br />

clinicians/researchers assisted by two patients with<br />

breast cancer who sit on the steering/data monitoring committee.<br />

The acceptable difference now has more empirical<br />

evidence to support it. Sophisticated work using individual<br />

patient interviews, conducted in Australia by Tammy Corica<br />

and colleagues 13 and in the United States by Michael<br />

Alvarado and colleagues, 14 suggests that 2.5% may be the<br />

value that patients are ready to accept as an increased risk<br />

of local recurrence as a trade-off for the convenience of a<br />

single-dose intraoperative radiotherapy as opposed to several<br />

weeks of daily radiotherapy. They would need to factor<br />

in not only the inconvenience of several weeks of radiotherapy,<br />

but also the fact that the quality of life of women<br />

Fig. 1. Targeted intraoperative radiotherapy (TARGIT).<br />

Meticulous surgical placement that opposes the<br />

tumour bed to the spherical applicator, while protecting<br />

the skin and deeper structures.<br />

19


EDUCATIONAL SUMMARIES<br />

Fig. 2. K-M estimate of local recurrence in TARGIT-A trial. Total blinded local recurrence rate in November<br />

<strong>2012</strong> compared with May 2010.<br />

receiving intraoperative radiotherapy has been found to be<br />

significantly better. 15<br />

We had previously responded 12 to the criticisms about the<br />

relatively short follow-up. As oft repeated, unplanned analysis<br />

comparing two groups in a randomized trial can carry<br />

the risk of a false–positive result. For this reason, we remained<br />

blinded to the further recurrences in the trial according<br />

to allocated treatment and proposed and performed<br />

a blinded analysis of the local recurrence rate for the whole<br />

cohort in November 2011, 18 months after the Lancet publication.<br />

We plotted the Kaplan-Meier plots and estimated<br />

the 4-year recurrence rate.<br />

Among the first 2,232 patients who were randomly assigned,<br />

there were 13 recurrences at the time of the Lancet<br />

publication and by November 2011 we had confirmed 10<br />

additional recurrences. Those who have completed at least 4<br />

years of follow-up increased from 420 to 692. As seen in Fig.<br />

2, we found that the 4-year Kaplan Meier estimate of total<br />

local recurrence was 1.08% (95% CI 0.59-1.96) at the time of<br />

the Lancet publication and in November 2011 it was 1.28%<br />

(95% CI 0.77-2.13). Thus, with a longer follow-up, the overall<br />

4-year rates of the TARGIT-A trial have remained low<br />

and stable.<br />

At the 12 th St. Gallen International <strong>Breast</strong> <strong>Cancer</strong> Conference<br />

in March 2011, a consensus panel of 52 experts in<br />

breast cancer voted in favor of intraoperative radiotherapy<br />

as part of the acceptable treatment program for suitable<br />

patients. When asked whether they would use partial breast<br />

irradiation (intraoperative) as the only radiotherapy for<br />

unselected patients, 48.9% said “yes,” 35.6% said “no,” and<br />

15.6% said “don’t know.” However, when applied to se-<br />

20<br />

lected patients (e.g., older than age 70) only 6.7% said “no”<br />

(86.7% said “yes” and 6.7% said “don’t know”). The panel<br />

was also in favor of using intraoperative radiotherapy as a<br />

tumor-bed boost instead of an EBRT boost (60.9% said<br />

“yes,” 17.4% said “no,” and 21.7% said “don’t know”).<br />

When patients wish to receive TARGIT as their definitive<br />

radiation treatment, we usually follow these steps: make<br />

the appropriate patient selection, per the characteristics of<br />

trial patients (for example, older than age 50, invasive ductal,<br />

T1, grade 1 or 2, ER-positive, etc.). Every patient is given<br />

the literature: the Lancet paper 9 and the subsequent correspondence<br />

in the Lancet, lay summary of the trial results, a<br />

patient information leaflet approved by our institutions,<br />

and website with previous literature (www.targit.org.uk).<br />

We stress that this is only for selected patients and that<br />

about 20% of patients will require whole breast radiotherapy.<br />

We emphasize that the noninferiority results are at 4<br />

years, and recurrence may be higher with longer follow-up.<br />

The patient is given consultations with the surgeon and<br />

radiation oncologist as well as several days to make their<br />

decision. The case is discussed in a multidisciplinary team<br />

meeting. We have developed a free iPhone app called<br />

TARGIT, which may be used by clinicians while making a<br />

decision about using TARGIT in different clinical situations,<br />

with the usual disclaimers.<br />

We are planning to close recruitment in the TARGIT-A<br />

trial after completing over 3,400 patients in June <strong>2012</strong>. We<br />

plan to analyze and publish an unblinded analysis in autumn<br />

of <strong>2012</strong>, including an updated one of the first cohort of<br />

2,232 patients.


Author’s Disclosures of Potential Conflicts of Interest<br />

Employment or<br />

Leadership Consultant or Stock<br />

Author<br />

Positions Advisory Role Ownership Honoraria<br />

Vaidya, Jayant S. Carl Zeiss<br />

References<br />

1. Vaidya JS, Vyas JJ, Chinoy RF, et al. Multicentricity of breast<br />

cancer: whole-organ analysis and clinical implications. Br J <strong>Cancer</strong>.<br />

1996;74:820-824.<br />

2. Baum M, Vaidya JS, Mittra I. Multicentricity and recurrence of<br />

breast cancer. Lancet. 1997;349:208.<br />

3. Vaidya JS, Baum M, Tobias JS, et al. Targeted intra-operative<br />

radiotherapy (Targit): an innovative method of treatment for<br />

early breast cancer. Ann Oncol. 2001;12:1075-1080.<br />

4. Vaidya JS, Baum M, Tobias JS, et al. Long-term results of<br />

targeted intraoperative radiotherapy (Targit) boost during<br />

breast-conserving surgery. Int J Radiat Oncol Biol Phys. 2011;<br />

81:1091-1097.<br />

5. Kraus-Tiefenbacher U, Bauer L, Scheda A, et al. Long-term<br />

toxicity of an intraoperative radiotherapy boost using low energy<br />

X-rays during breast-conserving surgery. Int J Radiat Oncol Biol<br />

Phys. 2006;66:377-381.<br />

6. Belletti B, Vaidya JS, D’Andrea S, et al.. Targeted intraoperative<br />

radiotherapy impairs the stimulation of breast cancer cell proliferation<br />

and invasion caused by surgical wounding. Clin <strong>Cancer</strong><br />

Res. 2008;14:1325-1332.<br />

7. Herskind C, Griebel J, Kraus-Tiefenbacher U, et al. Sphere of<br />

equivalence—a novel target volume concept for intraoperative<br />

radiotherapy using low-energy X rays. Int J Radiat Oncol Biol<br />

Phys. 2008;72:1575-1581.<br />

8. Herskind C, Wenz F. Radiobiological comparison of hypo-<br />

Research<br />

Funding<br />

EDUCATIONAL SUMMARIES<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

fractionated accelerated partial-breast irradiation (APBI) and<br />

single-dose intraoperative radiotherapy (IORT) with 50-kV<br />

X-rays. Strahlenther Onkol. 2010;186:444-451.<br />

9. Vaidya JS, Joseph DJ, Tobias JS, et al. Targeted intraoperative<br />

radiotherapy versus whole breast radiotherapy for breast cancer<br />

(TARGIT-A trial): an international, prospective, randomised,<br />

noninferiority phase 3 trial. Lancet. 2010;376:91-102.<br />

10. Vaidya JS. A novel approach for local treatment of early breast<br />

cancer. http://www.ucl.ac.uk/�rmhkjsv/papers/thesis.htm. Accessed<br />

June 10, <strong>2012</strong>.<br />

11. Bentzen SM, Agrawal RK, Aird EG, et al. The UK Standardisation<br />

of <strong>Breast</strong> Radiotherapy (START) Trial B of radiotherapy<br />

hypofractionation for treatment of early breast cancer: a randomised<br />

trial. Lancet. 2008;371:1098-1107.<br />

12. Vaidya JS. Intraoperative radiotherapy for early breast cancer—<br />

authors’ reply. Lancet. 2010;376:1143-1144.<br />

13. Corica T, Nowak A, Saunders C, et al. Patient preferences<br />

for adjuvant radiotherapy in early breast cancer—an Australian<br />

substudy of the International TARGIT Trial [Abstract<br />

482]. Eur J <strong>Cancer</strong>. <strong>2012</strong>;48(suppl):S187.<br />

14. Alvarado M, Connolly J, Oboite M, et al. Patient preference for<br />

choosing intraoperative or external-beam radiotherapy following<br />

breast conservation. Eur J <strong>Cancer</strong> Suppl. 2010;8:126-127.<br />

15. Welzel G, Hofmann F, Blank E, et al. Health-related quality of<br />

life after breast-conserving surgery and intraoperative radiotherapy<br />

for breast cancer using low-kilovoltage X-rays. Ann Surg<br />

Oncol. 2010;3:359-367.<br />

21


Debate on the Role of Radiation Therapy in Ductal<br />

Carcinoma In Situ: Con<br />

Abram Recht, MD<br />

Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA<br />

O<br />

ne can never be absolutely certain that there is no<br />

residual ductal carcinoma in situ (DCIS) left after<br />

breast-conserving surgery (BCS). Radiotherapy (RT),<br />

therefore, should always reduce the risk of local failure,<br />

as demonstrated in randomized trials (Table 1). 1-4 However,<br />

in these trials recurrence rates in patients treated with BCS<br />

alone were high. This was likely because these studies included<br />

relatively unselected patients who were eligible if<br />

there was no DCIS at the inked margins. A number of<br />

prospective and retrospective studies showed ipsilateral local<br />

failure rates of 5% to15% in certain patient subgroups<br />

treated with BCS without RT (Table 2). 5-11 The two most<br />

important factors determining the local failure rate following<br />

BCS without RT appear to be nuclear grade and margin<br />

width. (The effects of other factors, such as lesion size,<br />

patient age, 12 and necrosis, 13 are more controversial.)<br />

Patients with high-grade DCIS (whose main characteristic<br />

is high nuclear grade, although exact definitions differ<br />

from one group to the next) appear to have a substantial risk<br />

of local recurrence by 5 years following BCS, with little<br />

increase after that. 14 Patients with low- or intermediategrade<br />

DCIS have much lower rates of recurrence by 5 years.<br />

Their risk increases appreciably between years 5 and 10,<br />

although not to the same ultimate level as patients with<br />

high-grade DCIS.<br />

The minimum tumor-free margin width needed to<br />

achieve excellent results is not clear. Margins of less than 1<br />

mm are clearly inadequate, 14 whereas patients with margins<br />

wider than 10 mm, or no tumor on re-excision, appear<br />

to have low failure rates, except possibly for patients whose<br />

DCIS contains a high-grade component. 8 Patients with<br />

margin widths of 3 to 10 mm had very similar results to<br />

those with margin widths of 10 mm or more in one prospective<br />

trial. 9 A smaller retrospective study found a 6% failure<br />

rate in patients with margins wider than 5 mm. 11<br />

What are other potential consequences of not giving RT<br />

initially? Approximately one-half to two-thirds of patients<br />

having local failure after lumpectomy alone are treated with<br />

salvage breast-conserving therapy. 15 This relatively low<br />

rate of breast preservation seems due mainly to the psychological<br />

trauma caused by a recurrence for both patients and<br />

physicians, rather than the size of the recurrence or other<br />

technical factors that would prevent BCS. However, this<br />

reality means that giving RT initially to all patients might<br />

slightly decrease the ultimate number undergoing mastectomy.<br />

About one-half of local recurrences after lumpectomy<br />

alone are invasive and could potentially metastasize. Fortunately,<br />

this risk is small. Cure rates following salvage surgery<br />

for patients with invasive local recurrences are about<br />

80% to 90%. 1,15 The high rate of success for such treatment<br />

is likely why RT did not reduce the risk of distant metastases<br />

or breast cancer deaths in the randomized trials, despite<br />

the substantial local failure rates in their unirradiated patients<br />

(Table 1). The trials were too small to exclude a very<br />

small difference in such outcome (e.g., 0.5 to 1% at 10 years<br />

or more), but how meaningful would such differences be to<br />

patients even if they exist?<br />

The acute toxicities of RT are fortunately quite modest. 16<br />

Radiation therapy can impair the cosmetic results of treatment<br />

and increase rates of breast pain, but improved technology<br />

can minimize though not eliminate such problems. 17<br />

More ominously, however, radiation-related heart disease,<br />

sarcomas, lung cancers, and leukemias could increase death<br />

rates 10 to 20 years or more after treatment. However, this<br />

risk will likely be smaller than 1% among patients irradiated<br />

using current techniques.<br />

In summary, careful selection and treatment results<br />

in a modest risk of local failure following BCS without<br />

RT. Radiotherapy will likely reduce the recurrence rate by<br />

only a few percent in such individuals, with no impact on<br />

breast cancer–specific mortality. I therefore prefer to<br />

irradiate initially only women at high risk of local recurrence<br />

(particularly if the recurrence may be a high-<br />

Table 1. Randomized Trials Comparing <strong>Breast</strong>-Conserving Surgery with and without Radiotherapy<br />

Trial No. of<br />

Patients<br />

Median<br />

Follow-up<br />

(Months)<br />

Local Failure Distant Failure or<br />

<strong>Breast</strong> <strong>Cancer</strong> Death<br />

BCS BCS�RT BCS BCS�RT<br />

20% 3% c<br />

5%<br />

9% 3% a<br />

2%<br />

15% 4% b<br />

4%<br />

12% 2% a<br />

3%<br />

NSABP 1<br />

813 207 35% a<br />

UK/ANZ 2<br />

1,694 152 23% a<br />

EORTC 3<br />

1,010 126 26% b<br />

Sweden 4<br />

1,046 101 27% a<br />

Abbreviations: BCS, breast-conserving surgery; EORTC, European Organisation for Research and Treatment of <strong>Cancer</strong>; NSABP, National Surgical Adjuvant <strong>Breast</strong> and<br />

Bowel Program; RT, radiotherapy; UK/ANZ, United Kingdom, Australia, and New Zealand.<br />

a Crude rates.<br />

b 10-year rates.<br />

c 15-year rates.<br />

22


Table 2. Local Failure Rates in a Series of Highly Selected Patients Treated with <strong>Breast</strong>-Conserving Surgery without<br />

Radiotherapy<br />

Study Selection Criteria No. of<br />

Patients<br />

USC-Van Nuys 5<br />

Bologna 6<br />

Bordeaux 7<br />

Harvard 8<br />

ECOG 9<br />

Nottingham 10<br />

Linköping 11<br />

grade invasive cancer) and treat other patients only<br />

after relapse. I routinely recommend BCS without RT<br />

to women older than age 40 with lesions 20 mm or<br />

smaller by both radiologic and pathologic measures<br />

with highest nuclear grade 1 or 2 and margins 5 mm or<br />

wider or no DCIS on re-excision. Nonetheless, I will<br />

irradiate patients with low-risk DCIS if they prefer, after I<br />

have told them my views. The fear of recurrence can<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Employment or<br />

Leadership Consultant or<br />

Author<br />

Positions Advisory Role<br />

Recht, Abram CareCore<br />

References<br />

1. Wapnir IL, Dignam JJ, Fisher B, et al. Long-term outcomes of<br />

invasive ipsilateral breast tumor recurrences after lumpectomy<br />

in NSABP B-17 and B-24 randomized clinical trials for DCIS.<br />

J Natl <strong>Cancer</strong> Inst. 2011;103:478-488.<br />

2. Cuzick J, Sestak I, Pinder SE, et al. Effect of tamoxifen and<br />

radiotherapy in women with locally excised ductal carcinoma in<br />

situ: long-term results from the UK/ANZ DCIS trial. Lancet<br />

Oncol. 2011;12:21-29.<br />

3. Bijker N, Meijnen P, Peterse JL, et al. <strong>Breast</strong>-conserving<br />

treatment with or without radiotherapy in ductal carcinomain-situ:<br />

ten-year results of European Organisation for Research<br />

and Treatment of <strong>Cancer</strong> randomized phase III trial<br />

10853—a study by the EORTC <strong>Breast</strong> <strong>Cancer</strong> Cooperative<br />

Group and EORTC Radiotherapy Group. J Clin Oncol. 2006;<br />

24:3381-3387.<br />

4. Holmberg L, Garmo H, Granstrand B, et al. Absolute risk reductions<br />

for local recurrence after postoperative radiotherapy after<br />

sector resection for ductal carcinoma in situ of the breast. J Clin<br />

Oncol. 2008;26:1247-1252.<br />

5. Silverstein MJ, Lagios MD. Choosing treatment for patients with<br />

ductal carcinoma in situ: fine tuning the University of Southern<br />

California/Van Nuys Prognostic Index. J Natl <strong>Cancer</strong> Inst<br />

Monogr. 2010;2010:193-196.<br />

6. Di Saverio S, Catena F, Santini D, et al. 259 Patients with DCIS<br />

cause enormous anxiety. For some, this anxiety is worse<br />

that the possibility of short- and long-term toxicities<br />

from RT, even if such treatment will not change their<br />

ultimate chance of developing metastases or dying of<br />

breast cancer. In the end, only the patient can balance<br />

the advantages and disadvantages of different treatment<br />

options and make the decision that best meets her<br />

needs and values.<br />

Stock<br />

Ownership Honoraria<br />

Median<br />

Follow-up<br />

(Months)<br />

Research<br />

Funding<br />

Measure Local<br />

Failure<br />

USC-VN score 4-6 Unknown 75 12-year 6%<br />

USC-VN score 4-6 144 130 10-year 5%<br />

VN score 3-4 134 71 Crude 13%<br />

NG 1-2, no NG 3;<br />

margins 10 mm<br />

146 40 Crude 6%<br />

Grade 1 to 2,<br />

margin 3 mm<br />

565 74 5/7-year 6%/11%<br />

Margin 10 mm 256 86 Crude 11%<br />

Margin 5 mm 33 60 Crude 6%<br />

Abbreviations: ECOG, Eastern Cooperative Oncology Group; RT, radiotherapy; USC, University of Southern California; VN, Van Nuys.<br />

EDUCATIONAL SUMMARIES<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

of the breast applying USC/Van Nuys prognostic index: a retrospective<br />

review with long term follow up. <strong>Breast</strong> <strong>Cancer</strong> Res<br />

Treat. 2008;109:405-416.<br />

7. de Mascarel I, Bonichon F, MacGrogan G, et al. Application of the<br />

Van Nuys prognostic index in a retrospective series of 367 ductal<br />

carcinomas in situ of the breast examined by serial macroscopic<br />

sectioning: practical considerations. <strong>Breast</strong> <strong>Cancer</strong> Res Treat.<br />

2000;61:151-159.<br />

8. Wong JS, Kaelin CM, Troyan SL, et al. Prospective study of wide<br />

excision alone for ductal carcinoma in situ of the breast. J Clin<br />

Oncol. 2006;24:1031-1036.<br />

9. Hughes LL, Wang M, Page DL, et al. Local excision alone without<br />

irradiation for ductal carcinoma in situ of the breast: a trial of the<br />

Eastern Cooperative Oncology Group. J Clin Oncol. 2009;27:<br />

5319-5324.<br />

10. Rampaul RS, Valasiadou P, Pinder SE, et al. Wide local excision<br />

with 10 mm clearance without radiotherapy for DCIS (abstr.).<br />

Eur J Surg Oncol. 2001;27:788.<br />

11. Arnesson LG, Smeds S, Fagerberg G, et al. Follow-up of two<br />

treatment modalities for ductal carcinoma in situ of the breast.<br />

Br J Surg. 1989;76:672-675.<br />

12. Vicini FA, Kestin LL, Goldstein NS, et al. Impact of young age<br />

on outcome in patients with ductal carcinoma-in-situ treated<br />

with breast-conserving therapy. J Clin Oncol. 2000;18:296-<br />

306.<br />

23


EDUCATIONAL SUMMARIES<br />

13. Fisher ER, Dignam J, Tan-Chiu E, et al. Pathologic findings<br />

from the National Surgical Adjuvant <strong>Breast</strong> Project (NSABP)<br />

eight-year update of Protocol B-17. <strong>Cancer</strong>. 1999;86:429-438.<br />

14. Silverstein M, Recht A, Lagios M. Margin width as the sole<br />

predictor of local recurrence in patients with DCIS of the breast.<br />

In Silverstein M, Recht A, Lagios M (eds). Ductal Carcinoma in<br />

Situ of the <strong>Breast</strong> (2nd ed). Baltimore, MD: Lippincott Williams<br />

& Wilkins, 2002;643-650.<br />

15. Recht A. Salvage of local-regional failure. In Silverstein M, Recht<br />

24<br />

A, Lagios M (eds). Ductal Carcinoma in Situ of the <strong>Breast</strong> (2nd<br />

ed). Baltimore, MD: Lippincott Williams & Wilkins, 2002;539-544.<br />

16. Recht A. <strong>Breast</strong> <strong>Cancer</strong>: Stages I and II. In Gunderson LL,<br />

Tepper JE (eds). In Clinical Radiation Oncology (3rd ed). New<br />

York, NY: Elsevier, 2011;1321-1338.<br />

17. Donovan E, Bleakley N, Denholm E, et al. Randomised trial of<br />

standard 2D radiotherapy (RT) versus intensity modulated radiotherapy<br />

(IMRT) in patients prescribed breast radiotherapy.<br />

Radiother Oncol. 2007;82:254-264.


Intraoperative Radiation Therapy for <strong>Breast</strong> <strong>Cancer</strong><br />

Julia White, MD<br />

The Ohio State University Comprehensive <strong>Cancer</strong> Center<br />

I<br />

ntraoperative radiation therapy (IORT) refers to the<br />

delivery of a single high dose of irradiation directly to<br />

the postexcision tumor bed during surgery. IORT has been<br />

used as a boost method before standard whole breast irradiation<br />

(WBI) or as an intraoperative partial breast irradiation<br />

(PBI) as the sole treatment method postlumpectomy.<br />

The advantage of IORT is the accuracy of delivery of radiation<br />

to the postexcision tumor bed and convenience to patients<br />

who can avoid daily trips for treatment. The primary<br />

disadvantage is that the final pathology is unknown at time<br />

of delivery, and a proportion of patients will have adverse<br />

pathology features rendering them unsuitable for breast<br />

conservation despite having received IORT.<br />

The initial experiences using IORT for breast cancer<br />

delivered a boost to the lumpectomy cavity immediately<br />

postresection. Typically, standard WBI is initiated postsurgery<br />

with IORT at either 3 to 5 weeks, or later at 18 to 24<br />

weeks postchemotherapy when indicated. The largest published<br />

experience is from a pooled retrospective analysis of<br />

six institutions from Europe that are part of the International<br />

Society of Intraoperative Radiotherapy (ISIORT). 3<br />

This analysis included 1,131 women who received linacbased<br />

IORT with 6 to 8 MeV electrons that delivered a mean<br />

dose of 9.7 Gy to the lumpectomy cavity. In this population,<br />

37 % had positive axillary nodes and greater than 95% had<br />

some type of systemic therapy. At a median follow-up of 52.3<br />

months, there were five in-breast cancer recurrences yielding<br />

a local tumor control rate of 99.4%. In this experience,<br />

43 patients who had positive margins at final pathology<br />

postresection with IORT were converted to mastectomy.<br />

The Milan group has reported the preliminary results<br />

of their electrons intraoperative therapy (ELIOT) method<br />

used as a boost in a single-arm, prospective trial that enrolled<br />

211 premenopausal women. 4 The ELIOT approach<br />

uses 9 MeV electrons from a mobile linear accelerator to<br />

deliver a single dose of 12 Gy to the tumor bed immediately<br />

after excision, while shielding the chest wall with a lead<br />

plate. The WBI began 4 weeks afterward with a hypofrationated<br />

regimen of 37 Gy in 13 fractions of 2.85 Gy. With<br />

very short follow-up (11 months) reported so far, low rates<br />

(2.7%) of grade 3 or higher acute toxicity were seen, and no<br />

in-breast recurrences had occurred.<br />

The intrabeam method of IORT reported their results<br />

delivering a boost in 321 women enrolled on a pilot trial. 5<br />

Intrabeam is a miniature electron generator and accelerator.<br />

Electrons are accelerated along a tube and strike the tip<br />

to generate soft X-rays that are modulated by a spherical<br />

applicator to give a uniform field of radiotherapy. The<br />

spherical applicator is placed into the cavity following resection,<br />

and radiation can be delivered in routine operating<br />

rooms without specialized shielding. The IORT dose delivered<br />

in this pilot study was 18 to 20 Gy at the applicator<br />

surface and 5 to 7 Gy at 1 cm from the applicator. Standard<br />

WBI was delivered afterwards typically with 2 Gy fractions<br />

from 45 to 50 Gy. With a median follow-up of 60.5 months,<br />

eight patients have had in-breast cancer recurrences for a<br />

very low 5-year rate of 1.73%.<br />

There is mounting experience supporting the use of IORT<br />

as PBI to deliver the sole radiotherapy treatment postlumpectomy<br />

in select cases. The TARGIT phase III trial<br />

randomly assigned 2,232 women undergoing breastconserving<br />

treatment to TARGIT (1,113 patients) and/or<br />

standard external beam WBI (1,119 patients) postlumpectomy.<br />

6 In the TARGIT arm, 86% received just the IORT,<br />

and 14% received IORT with conventional WBI because of<br />

the presence of adverse pathology features. The dosing was<br />

similar to the boost pilot; patients received 20 Gy to the<br />

surface of the applicator and 5 to 7 Gy at 1 cm depth. The<br />

median age was 63 in this population of mostly stage I<br />

hormone-sensitive breast cancers. With a median follow-up<br />

of approximately 24 months, there were six in-breast recurrences<br />

in the TARGIT arm and five in the WBI arm for a<br />

4-year rate of 1.2% and 0.95%, respectively (p � 0.41).<br />

The outcome from 842 women accrued to a phase III trial<br />

by the Milan group randomizing to ELIOT 21 Gy versus<br />

standard WBI 50 Gy in 25 fractions plus boost postlumpectomy<br />

is awaited. However, the outcome of 1,822 patients<br />

with breast cancer undergoing quadrantectomy who were<br />

treated with ELIOT off of the clinical protocol has been<br />

reported. 7 In this population the median age was 58, median<br />

tumor size was 1.3 cm, 89% of patients were receptor positive,<br />

and 71% of patients had node-negative disease. With a<br />

mean follow-up of 40 months, there have been 66 ipsilateral<br />

breast cancer events (3.6%), 42 (2.3%) were characterized<br />

as a true local recurrence when it occurred in the treated<br />

quadrant and 24 (1.3%) were characterized as ipsilateral<br />

new breast cancer when it occurred in a new quadrant. A<br />

subsequent study classified all 1,822 cases by the <strong>American</strong><br />

Society for Radiation Oncology Consensus Guidelines of<br />

suitable, cautionary, and unsuitable for receipt of postlumpectomy<br />

accelerated partial breast irradiation when not<br />

enrolled in a clinical trial. 8 The 5-year rate of in-breast<br />

recurrence for the suitable, cautionary, and unsuitable patients<br />

were 1.5%, 4.4%, and 8.8%, respectively (p � 0.0003).<br />

The authors concluded that the ASTRO recommendation<br />

offered useful guidance to judge the appropriateness of PBI<br />

using ELIOT.<br />

Given the long documented history of success with standard<br />

WBI following lumpectomy, it is important that alternatives<br />

to this standard for breast radiotherapy such as<br />

IORT demonstrate comparable therapeutic effectiveness.<br />

So far, IORT appears effective as a boost method. Initial<br />

experience with IORT for PBI appears promising; however,<br />

longer follow up is necessary to clearly validate its efficacy<br />

prior to more general adoption outside of a clinical trial.<br />

25


EDUCATIONAL SUMMARIES<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

White, Julia*<br />

*No relevant relationships to disclose.<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

References<br />

1. Seldmayer F, Fastner G, Merz F, et al. IORT with electrons as<br />

boost strategy during breast conserving therapy in limited stage<br />

breast cancer: Results of an ISIORT pooled analysis. Strahlenther<br />

Onkol. 2007;183:32-34.<br />

2. Ivaldi GB, Leonardi MC. Orecchia R, et al. Preliminary results of<br />

electron intraoperative therapy boost and hypofractionated external<br />

beam radiotherapy after breast-conserving surgery in premenopausal<br />

women. Int J Radiation Oncology Biol Phys. 2008;72:<br />

485-493.<br />

3. Vaidya JS, Baum M, Tobias, JS, et al. Long term results of<br />

targeted intraoperative radiotherapy (TARGIT) boost during<br />

breast conserving surgery. Int J Radiation Oncology Biol Phys.<br />

2011;81:1091-1097.<br />

26<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

4. Vaidya JS, Joseph DJ, Tobias JS, et al. Targeted intraoperative<br />

radiotherapy versus whole breast radiotherapy for breast cancer<br />

(TARGIT – A trial): an international, prospective, randomized,<br />

non-inferiority phase 3 trial. Lancet. 2010;376:91-102.<br />

5. Veronesi U, Orecchia R, Luini A, et al. Intraoperative radiotherapy<br />

during breast conserving surgery: study of 1,822 cases treated<br />

with electrons. <strong>Breast</strong> <strong>Cancer</strong> Res Treat. 2010;124:141-151.<br />

6. Leonardi MC, Maisonneuve P, Mastropasqua MG, et al. How do<br />

the ASSTRO consensus statement guidelines for application of<br />

accelerated partial breast irradiation fit intraoperative radiotherapy?<br />

A retrospective analysis of patients treated at the European<br />

Institute of Oncology. Int J Radiation Oncology Biol Phys. <strong>2012</strong>;<br />

83:806-8013.


GENERAL SESSION V:<br />

Genomics for the Clinician<br />

CHAIR<br />

Mark D. Pegram, MD<br />

Stanford University<br />

SPEAKERS<br />

Matthew J. Ellis, MD<br />

Washington University<br />

Charles E. Geyer Jr., MD<br />

Clinical Trials Network of Texas<br />

Ana M. Gonzalez-Angulo, MD, MSc<br />

University of Texas M. D. Anderson <strong>Cancer</strong> Center<br />

After this session, attendees should be able to<br />

● Assess the future of breast cancer genomics and determine how new discoveries can be applied to the prevention,<br />

diagnosis, and treatment of patients with breast cancer<br />

● Examine the current use of genomics to inform the treatment of patients with metastatic disease and determine future<br />

directions to more effectively prevent disease and treat patients by targeting known mutations in cancer susceptibility<br />

genes<br />

● Analyze the potential applications for the use of genomic testing in node-positive patients, including a determination of<br />

appropriate use during diagnosis, treatment, and post-treatment


PI3K in <strong>Breast</strong> <strong>Cancer</strong><br />

Toby M. Ward, PhD, Rebecca Olson, MS, Mark D. Pegram, MD,<br />

and Charles E. Geyer Jr., MD<br />

Stanford University, Stanford, CA, and<br />

Statewide Clinical Trials Network of Texas, Addison, TX<br />

T<br />

he product of the phosphoinositide 3-kinase (PI3KCA)<br />

proto-oncogene is involved in transmitting growth<br />

and survival signals from receptor tyrosine kinases and/or<br />

RAS proteins to downstream effectors, including AKT and<br />

the mTOR complexes. A role for PI3K in cancer was first<br />

elucidated by the identification of a viral oncogene homolog<br />

in avian sarcoma virus 16. 1 Subsequent studies of human<br />

malignancy have revealed that the PI3K signaling pathway<br />

is deregulated frequently in a wide variety of human cancers,<br />

including breast, ovarian, lung, and endometrial cancers,<br />

and several mechanisms of deregulation have been<br />

described. 2-5 Thus, this pathway has received intense scrutiny<br />

as a target for therapeutic intervention in the treatment<br />

of breast cancer, as well as other malignancies.<br />

Structure and Function of PI3KCA<br />

Gene Product<br />

Type I PI3K kinases, of which PI3KCA is a member, function<br />

as heterodimers composed of regulatory and catalytic<br />

subunits, such as p85 and p110 (Fig. 1). 6 Activation of<br />

cell-surface receptors, such as the insulin receptor and the<br />

epidermal growth factor receptor family of proteins, recruits<br />

PI3K, providing a docking site for the p85 regulatory<br />

subunit (either by direct interaction with the receptor or via<br />

adaptor proteins). Upon activation, PI3K catalyzes the addition<br />

of a phosphate group to the membrane lipid phosphatidylinositol<br />

(4,5)-bisphosphate (PIP2), yielding the active<br />

signaling lipid phosphatidylinositol (3,4,5)-trisphosphate<br />

(PIP3). This activity is opposed by the tumor-suppressor<br />

phosphatase and tensin homolog deleted on chromosome<br />

10 (PTEN). PIP3 is able to recruit and interact with signaling<br />

proteins that contain pleckstrin-homology (PH) or<br />

other lipid-binding domains, transmitting signals that eventually<br />

influence cell growth and survival pathways (among<br />

others). 2-5<br />

Fig. 1. Canonical PI3K signaling.<br />

28<br />

PI3KCA Alterations in <strong>Breast</strong> <strong>Cancer</strong><br />

Human breast cancer clinical specimens exhibit a high rate<br />

of PI3KCA gene mutation (approximately 30% of unselected<br />

primary tumors), which occur across the various<br />

intrinsic subtypes, based upon gene expression array phenotypes.<br />

7,8 According to a recent report from a The <strong>Cancer</strong><br />

Genome Atlas (TCGA) study, PI3KCA is the most commonly<br />

mutated gene in luminal A and luminal B intrinsic<br />

subtypes (49.7% and 30.1% of tumors harbored single nucleotide<br />

variants [SNVs], respectively), and is second only<br />

to p53 in ERBB2-amplified and basal breast cancer (where<br />

40.4% and 9.7% of tumors harbored PI3KCA SNVs, respectively).<br />

7,8 PI3KCA was one of only four genes with a mutation<br />

frequency of more than 10% in human breast cancers,<br />

underscoring its importance in the oncogenic process.<br />

The majority of these mutations within PI3KCA are<br />

in the helical domain (E542K, E545K) and kinase domain<br />

(H1047R) of the protein, and both types of mutation increase<br />

PI3K catalytic activity. 9 Moreover, amplification<br />

of the PI3KCB gene has also been described in ERBB2positive<br />

breast cancers, 10 and somatic mutation of other<br />

components of the PI3K signaling axis are also detected in<br />

breast cancer specimens, including mutations in the IRS<br />

and PI3KR1 genes. 11<br />

PTEN<br />

Another mechanism of PI3K signaling pathway activation<br />

is deletion or mutation of the PTEN tumor suppressor, a<br />

phosphatase which catalyzes removal of a phosphate from<br />

PIP3 to generate PIP2, thus opposing the activity of PI3K.<br />

In breast cancer, PTEN loss of heterozygosity can occur by<br />

deletion, mutation, or epigenetic silencing of the gene and<br />

has been extensively reviewed. 12<br />

PI3K Pathway Inhibitors<br />

Due to its activation and involvement in breast and other<br />

cancers, inhibition of the PI3K signaling pathway is currently<br />

the subject of intense clinical research. Indeed, in a<br />

recent review of clinical trials aimed at testing PI3K pathway<br />

inhibitors, approximately 20 different drugs were reported<br />

as being in preclinical or clinical development at the<br />

present time. 13 These drugs target a suite of signaling intermediates<br />

in the PI3K pathway, including (but not limited<br />

to) PI3KCA, AKT, and the mTOR complexes (Table 1).<br />

PI3K Mutation versus Pathway Activation:<br />

Effect on <strong>Breast</strong> <strong>Cancer</strong> Therapy<br />

Vis-à-vis therapeutic rationale, knowledge of PI3KCA somatic<br />

mutational status may not be as informative as as-


Table 1. Current Clinical Trials of Agents Targeting PI3K Pathway<br />

Clinical Trials Agent Company NCT<br />

Phase I BYL719 Novartis NCT01219699<br />

Phase I PX-866 Oncothyreon Inc NCT00726583<br />

Phase I BKM-120 Novartis NCT01068483<br />

Phase I SF1126 Semafore Pharmaceuticals NCT00907205<br />

Phase I BEZ-235 Novartis NCT00620594<br />

Phase I GSK1059615 GlaxoSmithKline NCT00695448<br />

Phase I GSK690693 GlaxoSmithKline NCT00493818<br />

Phase I AZD-8055 AstraZeneca NCT00973076<br />

Phase I OSI-027 OSI Pharmaceuticals NCT00698243<br />

Phase I INK-128 Intellikine NCT01058707<br />

Phase I/II XL-147 Exelixis NCT01042925<br />

Phase II CAL-101 Calistoga Pharmaceuticals, Inc. NCT00710528<br />

Phase II GDC-0941 Genentech Inc NCT00960960<br />

Phase II XL-765 Exelixis NCT01082068<br />

Phase II Perifosine Keryx/AOI Pharmaceuticals NCT00847366<br />

Phase II MK-2206 Merck NCT00963547<br />

Phase II Sirolimus Wyeth/Pfizer NCT00411788<br />

Phase II Temsirolimus Wyeth/Pfizer NCT01111825<br />

Phase II Ridaforolimus ARIAD/Merck NCT01234857<br />

Phase II/III Everolimus Novartis NCT01061788<br />

Modified from Bartholomeusz C and Gonzalez-Angulo AM. 13<br />

sessment of activation of the signaling pathway as a whole.<br />

Indeed, while mutation of PI3KCA occurs most frequently<br />

in the luminal A breast cancer subtype (approximately 45%<br />

of tumors in TCGA study), mRNA and proteomic “signatures”<br />

corresponding to PI3K pathway activation are arguably<br />

more pronounced in the luminal B, ERBB2-positive,<br />

and basal groups. 8 Paradoxically, PI3KCA mutation may<br />

actually be associated with improved clinical outcome in<br />

Authors’ Disclosures of Potential Conflicts of Interest<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Author<br />

Ward, Toby M.*<br />

Olson, Rebecca*<br />

Pegram, Mark D. Dava Oncology (I) Bristol-Myers<br />

Squibb;<br />

Genentech;<br />

GlaxoSmithKline;<br />

Novartis; Pfizer/<br />

Wyeth (U);<br />

Sanofi<br />

Geyer, Charles E., Jr.*<br />

*No relevant relationships to disclose.<br />

ER-positive breast cancers treated with tamoxifen monotherapy.<br />

14 Hence, selection of therapy to include PI3K inhibitors<br />

arguably should not be based solely on somatic<br />

mutation of PI3KCA in patient tumors alone. Rather, biomarker<br />

analysis of key constituents of the PI3K signaling<br />

pathway may prove to be more informative predictive markers<br />

of potential clinical benefit for therapies targeting the<br />

PI3K pathway.<br />

Stock<br />

Ownership Honoraria<br />

Genentech;<br />

GlaxoSmithKline;<br />

Sanofi<br />

Research<br />

Funding<br />

Expert<br />

Testimony<br />

Sanofi Novartis<br />

EDUCATIONAL SUMMARIES<br />

Other<br />

Remuneration<br />

29


EDUCATIONAL SUMMARIES<br />

References<br />

1. Chang HW, Aoki M, Fruman D, et al. Transformation of chicken<br />

cells by the gene encoding the catalytic subunit of PI 3-kinase.<br />

Science. 1997;276:1848-1850.<br />

2. Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT<br />

pathway in human cancer. Nat Rev <strong>Cancer</strong>. 2002;2:489-501.<br />

3. McAuliffe PF, Meric-Bernstam F, Mills GB, et al. Deciphering<br />

the role of PI3K/Akt/mTOR pathway in breast cancer biology and<br />

pathogenesis. Clin <strong>Breast</strong> <strong>Cancer</strong>. 2010;10(suppl):S59-65.<br />

4. Tokunaga E, Oki E, Egashira A, et al. Deregulation of the Akt<br />

pathway in human cancer. Curr <strong>Cancer</strong> Drug Targets. 2008;8:27-<br />

36.<br />

5. Bader AG, Kang S, Zhao L, et al. Oncogenic PI3K deregulates<br />

transcription and translation. Nat Rev <strong>Cancer</strong>. 2005;5:921-929.<br />

6. Vadas O, Burke JE, Zhang X, et al. Structural basis for activation<br />

and inhibition of class I phosphoinositide 3-kinases. Sci Signal.<br />

2011;4:re2.<br />

7. Mardis, E. The genomic mutation landscape of breast cancer: a<br />

TCGA report. <strong>Cancer</strong> Research. 2011;71:92s.<br />

8. Creighton C. Integrative genomic analyses of breast cancer from<br />

30<br />

The <strong>Cancer</strong> Genome Atlas (TCGA). <strong>Cancer</strong> Research. 2011;71:<br />

93s.<br />

9. Zhao L, Vogt PK. Class I PI3K in oncogenic cellular transformation.<br />

Oncogene. 2008;27:5486-5496.<br />

10. Crowder RJ, Phommaly C, Tao Y, et al. PIK3CA and PIK3CB<br />

inhibition produce synthetic lethality when combined with estrogen<br />

deprivation in estrogen receptor-positive breast cancer. <strong>Cancer</strong><br />

Res. 2009;69:3955-3962.<br />

11. Jaiswal BS, Janakiraman V, Kljavin NM, et al. Somatic mutations<br />

in p85alpha promote tumorigenesis through class IA PI3K<br />

activation. <strong>Cancer</strong> Cell. 2009;16:463-474.<br />

12. Hollander MC, Blumenthal GM, Dennis PA. PTEN loss in the<br />

continuum of common cancers, rare syndromes and mouse models.<br />

Nat Rev <strong>Cancer</strong>. 2011;11:289-301.<br />

13. Bartholomeusz C, Gonzalez-Angulo AM. Targeting the PI3K<br />

signaling pathway in cancer therapy. Expert Opin Ther Targets.<br />

<strong>2012</strong>;16:121-130.<br />

14. Loi S, Haibe-Kains B, Majjaj S, et al. PIK3CA mutations associated<br />

with gene signature of low mTORC1 signaling and better<br />

outcomes in estrogen receptor-positive breast cancer. Proc Natl<br />

Acad Sci USA.2010;107:10208-10213.


Role of Genomic Testing in Patients with<br />

Node-Positive <strong>Breast</strong> <strong>Cancer</strong><br />

Shaheenah Dawood, MRCP, MPH, and Ana M. Gonzalez-Angulo, MD, MSc<br />

Dubai Hospital, Dubai, United Arab Emirates and<br />

University of Texas M. D. Anderson <strong>Cancer</strong> Center, Houston, TX<br />

O<br />

ver the last decade a number of important advances<br />

have been made in the realm of breast cancer. First<br />

has been the documented decline in mortality associated<br />

with breast cancer, attributed to a combination of screening<br />

programs and incorporation of optimal polychemotherapy<br />

and endocrine regimens that have reduced the annual odds<br />

of breast cancer recurrence by approximately 50% to 60%. 1<br />

Second has been the realization that breast cancer is not a<br />

homogenous disease but rather a conglomeration of a<br />

number of subtypes including luminal A, luminal B, HER2enriched,<br />

and basal-like subtypes; each is associated with<br />

a different prognostic outcome. 2 Third has been the development—through<br />

high-throughput techniques—of gene<br />

expression–profiling platforms that are able to not only<br />

predict prognostic outcome but also, as the data indicate,<br />

predict for responsiveness to chemotherapy and thus be<br />

able to identify groups who may not require chemotherapy,<br />

thereby avoiding both short- and long-term toxicities typically<br />

associated with chemotherapy. 3 The two prototypes<br />

that have the most clinical data reported and are being<br />

evaluated in prospective clinical trials are MammaPrint<br />

(Agendia, Inc., Amsterdam, the Netherlands) and Oncotype<br />

Dx (Genomic Health, Inc., Santa Clara, CA). Analysis of the<br />

NSABP-20 study, 3 a trial that randomly assigned women<br />

with node-negative, estrogen receptor–positive breast<br />

cancer to either tamoxifen alone or tamoxifen and chemotherapy,<br />

revealed that women with a high recurrence<br />

score (RS) (greater than or equal to 31) had a large benefit<br />

from the addition of chemotherapy while those with low<br />

RS (less than 18) derived minimal if any benefit from the<br />

addition of chemotherapy. Such data led to the endorsement<br />

for the use of Oncotype DX RS by the <strong>American</strong><br />

Society of Clinical Oncology and the National Comprehensive<br />

<strong>Cancer</strong> Network as both a prediction and prognostic<br />

tool among women with estrogen receptor–positive, nodenegative<br />

breast cancer. However, the question remains as<br />

to whether such tools would be useful among women with<br />

node-positive disease.<br />

Do All Women with Node-Positive <strong>Breast</strong><br />

<strong>Cancer</strong> Require Chemotherapy?<br />

The current standard of care among women with nodepositive<br />

disease is to receive adjuvant chemotherapy; a contention<br />

derived from results of prospective clinical trials<br />

that have shown a survival benefit among women with<br />

estrogen receptor–positive and –negative disease. 1,4 However,<br />

the question remains, do all women within this cohort<br />

benefit from chemotherapy? In a retrospective subset analysis<br />

of three prospective clinical trials looking at women<br />

with node-positive disease, Berry and colleagues showed<br />

that the addition of chemotherapy resulted in an absolute<br />

improvement in 5-year disease-free survival of 22.8% and<br />

7% among women with estrogen receptor–negative and estrogen<br />

receptor–positive disease, respectively. 5 The results<br />

clearly indicate that the largest benefit attained from the<br />

addition of chemotherapy was achieved among women with<br />

estrogen receptor–negative disease. The question then<br />

arises as to whether all women with estrogen receptor–<br />

positive, node-positive disease benefit equally from the addition<br />

of chemotherapy to endocrine therapy?<br />

Can Genomic Profiling Help Refine<br />

Chemotherapy Prediction among<br />

Node-Positive Disease?<br />

Using an unselected cohort of women with early-stage<br />

breast cancer, of whom 51% had N1 disease, Knauer and<br />

colleagues 6 used the 70-gene MammaPrint signature to<br />

determine the benefit of adding chemotherapy to endocrine<br />

therapy. The authors showed that the 5-year breast cancer–<br />

specific survival was similar within the 70-gene low-risk<br />

group who did and did not receive chemotherapy (99% vs.<br />

97%); among the 70-gene high-risk group, a significant benefit<br />

was demonstrated with the addition of chemotherapy to<br />

endocrine therapy (94% vs. 81%; p � 0.01). Looking specifically<br />

at a cohort of women with one to three positive nodes<br />

who received endocrine therapy the Trans-BIG collaborators<br />

demonstrated that the prognostic outcome among<br />

women categorized within the 70-gene low risk group was<br />

similar among those who did and did not receive chemotherapy.<br />

7 In a recent exploratory study of women enrolled in the<br />

SWOG 8814 trial with node-positive, hormone receptor–<br />

positive disease (where approximately 60% of patients had<br />

one to three positive nodes), women were randomly assigned<br />

to endocrine therapy alone or to endocrine therapy<br />

and chemotherapy and Albain and colleagues 4 used the<br />

Oncotype DX RS to determine the benefit of chemotherapy.<br />

The authors demonstrated no benefit with the addition of<br />

chemotherapy among women with a RS less than 18 while a<br />

significant benefit was demonstrated among those with a<br />

RS greater than or equal to 31. Taken collectively, these<br />

data indicate that multigene assays may be able to define a<br />

group within the node-positive, hormone receptor–positive<br />

population that may not require chemotherapy. Furthermore<br />

the data described above looked at a cohort of women<br />

treated with tamoxifen. It is likely that among postmenopausal<br />

women, the use of aromatase inhibitors will show<br />

additional benefit in terms of prognostic outcome.<br />

How Will RxPONDER Answer the Question?<br />

Clearly the previously described data indicate that<br />

women with node-positive, endocrine- responsive disease<br />

31


EDUCATIONAL SUMMARIES<br />

defined as high risk using multigene assays benefit the most<br />

from the addition of chemotherapy to endocrine therapy.<br />

The low- and intermediate-risk groups appear not to derive<br />

as much benefit, and larger prospective clinical trials are<br />

needed to confirm this. RxPONDER (treatment for nodepositive,<br />

endocrine-responsive breast cancer) is a phase III<br />

clinical trial whose primary objective is to define the benefit<br />

of chemotherapy among women with hormone receptor–<br />

positive breast cancer with one to three positive nodes receiving<br />

state-of-the-art endocrine therapy. The modified<br />

schema of the protocol is illustrated in Fig. 1. The trial will<br />

use Oncotype DX to enroll women with a RS that is less than<br />

or equal to 25 who will then be randomly assigned to endocrine<br />

therapy alone or endocrine therapy plus chemotherapy.<br />

The RS of 25 was chosen to incorporate all low-risk<br />

groups as well as the most intermediate-risk group of<br />

women with the hypothesis that along the RS range of 0 to<br />

25 a point of equivalence will be identified that will function<br />

as a cutoff point identifying patients who would and would<br />

not benefit from the addition of chemotherapy. The investigators<br />

of the trial hypothesize that the point of equivalence<br />

may emerge at a RS range of 19 to 20; where patients<br />

scoring higher would benefit from chemotherapy. Should<br />

Fig. 1. Schema of the RxPonder trial (S1007).<br />

32<br />

this hypothesis be proven, it is estimated that approximately<br />

67% of women with one to three positive nodes and<br />

endocrine-responsive disease would avoid chemotherapy<br />

and all the associated morbidity and cost.<br />

Conclusion<br />

We certainly have come a long way from the one-size-fitsall<br />

model of breast cancer treatment to a more personalized<br />

approach. The use of genomic profiling has certainly influenced<br />

the practice of oncology and studies indicate that<br />

for women with node-negative disease, RS change recommendations<br />

for adjuvant therapy occur approximately 17%<br />

to 26% of the time in clinical practice. 8 Women with nodepositive<br />

disease have traditionally been considered to be<br />

at highest risk of recurrence. However, multigene assays<br />

may be able to identify a group of women with endocrineresponsive,<br />

node-positive disease that may not require<br />

chemotherapy, thus avoiding the associated toxicities. However,<br />

prospective trials will be essential to accurately identify<br />

the groups that will and will not benefit from<br />

chemotherapy. Until results of such studies are available,<br />

the current guidelines still endorse the use of adjuvant<br />

chemotherapy among all women with node-positive disease.


Authors’ Disclosures of Potential Conflicts of Interest<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Author<br />

Dawood, Shaheenah*<br />

Gonzalez-Angulo, Ana M. Bayer; Bristol-<br />

Myers Squibb;<br />

Celgene;<br />

Genentech;<br />

Merck; Novartis<br />

*No relevant relationships to disclose.<br />

References<br />

1. Early <strong>Breast</strong> <strong>Cancer</strong> Trialists’ Collaborative Group (EBCTCG).<br />

Effects of chemotherapy and hormonal therapy for early breast<br />

cancer on recurrence and 15-year survival: an overview of the<br />

randomised trials. Lancet. 2005;365:1687-1717.<br />

2. Sorlie T. Molecular portraits of breast cancer. Tumour subtypes<br />

as distinct disease entities. Eur J <strong>Cancer</strong>. 2004;40:2667-2675.<br />

3. Paik S, Tang G, Shak S et al. Gene expression and benefit of<br />

chemotherapy in women with node negative, estrogen receptorpositive<br />

breast cancer. J Clin Oncol. 2006;24:3726-3734.<br />

4. Albain KS, Barlow WE, Ravdin PM, et al. Adjuvant chemotherapy<br />

and timing of tamoxifen in postmenopausal patients with<br />

endocrine-responsive, node-positive breast cancer: a phase 3, openlabel,<br />

randomised controlled trial. Lancet. 2009;374:2055-2063.<br />

EDUCATIONAL SUMMARIES<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

5. Berry DA, Cirrincione C, Henderson IC, et al. Estrogen-receptor<br />

status and outcomes of modern chemotherapy for patients with<br />

node-positive breast cancer. JAMA. 2006;295:1658-1667.<br />

6. Knauer M, Mook S, Rutgers EJ, et al. The predictive value of the<br />

70-gene signature for adjuvant chemotherapy in early breast cancer.<br />

<strong>Breast</strong> <strong>Cancer</strong> Res Treat. 2010;120:655-661.<br />

7. Mook S, Schmidt MK, Viale G, et al. The 70-gene prognosissignature<br />

predicts disease outcome in breast cancer patients with<br />

1-3 positive lymph nodes in an independent validation study.<br />

<strong>Breast</strong> <strong>Cancer</strong> Res Treat. 2009;116:295-302.<br />

8. Oratz R, Paul D, Cohn AL, et al. Impact of a commercial reference<br />

laboratory test recurrence score on decision making in early-stage<br />

breast cancer. J Oncol Pract. 2007;3:182-186.<br />

33


<strong>Breast</strong> <strong>Cancer</strong> Whole Genome Sequencing<br />

Matthew J. Ellis, MB BChir, PhD<br />

The <strong>Breast</strong> <strong>Cancer</strong> Program, Siteman <strong>Cancer</strong> Center,<br />

Washington University, St. Louis, MO<br />

T<br />

he application of whole cancer genome sequencing<br />

approaches to breast cancer has changed our thinking<br />

about the disease. Since this technique is unbiased, all<br />

classes of somatic mutations are revealed, and because it is<br />

digital in nature, information on gene copy number and<br />

minor allele frequencies reveal a degree of complexity hitherto<br />

unsuspected. We have focused this technology on two<br />

areas. First is an analysis of samples from clinical trials, only<br />

in this setting can adequate prospective consent for genetic<br />

research and data sharing, high quality samples, and detailed<br />

clinical outcomes can be obtained. Second is a comparison<br />

of the whole genome sequences of human breast<br />

cancers with their counterpart xenograft sample after passage<br />

in immunodeficient mice. The objective of these mouse<br />

modelling studies is to document the genome-wide similarities<br />

and differences as a prelude to the use of xenograft<br />

panels for therapeutic modelling based on pharmacological<br />

hypotheses generated by analysis of genomic data.<br />

To correlate the variable clinical features of estrogen<br />

receptor–positive breast cancer with somatic alterations, we<br />

studied pretreatment tumor biopsies accrued from patients<br />

in two studies of neoadjuvant aromatase inhibitor therapy.<br />

Eighteen significantly mutated genes were identified, including<br />

five genes (RUNX1, CBFB, MYH9, MLL3, and<br />

SF3B1) previously linked to hematopoietic disorders. These<br />

findings suggest that breast cancer, like leukemia, can be<br />

viewed as a stem-cell disorder that produces indolent or<br />

aggressive tumors that display varying phenotypes depending<br />

on differentiation blocks generated by different mutation<br />

repertoires. Mutant MAP3K1 was associated with<br />

luminal A status, low grade histology, and low proliferation<br />

rates, whereas mutant TP53 was associated with the<br />

opposite pattern. Moreover, mutant GATA3 correlated with<br />

suppression of proliferation upon aromatase inhibitor treatment.<br />

Pathway analysis demonstrated that mutations in<br />

MAP2K4, a MAP3K1 substrate, produced similar perturbations<br />

as MAP3K1 loss. These analyses also identified transcriptional<br />

associations to Ki67 response residing in a<br />

connected network under the control of several key “hub”<br />

genes including MYC, FYN, and MAP kinases, among oth-<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Ellis, Matthew J. AstraZeneca;<br />

GlaxoSmithKline;<br />

Novartis; Pfizer<br />

References<br />

ers. Targeting these hubs in resistant tumors could produce<br />

therapeutic advances. Aside from the common PIK3CA mutations,<br />

druggable mutations were relatively uncommon<br />

but were observed in HER2, KIT, PDGFA, DDR1/2, MET,<br />

JAK1, CSFR1, LTK, BRAF, and AKT1/2. If these mutations<br />

are activating and drug sensitive, these are findings<br />

are significant given that breast cancer is so common that<br />

drug-sensitizing mutations with a 1% prevalence represent<br />

a population with a similar magnitude to chronic myeloid<br />

leukemia and promyelocytic leukemia, diseases in which a<br />

randomized trials are feasible. Distinct phenotypes in estrogen<br />

receptor–positive breast cancer are associated with specific<br />

patterns of somatic mutations that map into cellular<br />

pathways linked to tumor biology, but most recurrent mutations<br />

are relatively infrequent. Prospective clinical trials<br />

based on these findings will require comprehensive genome<br />

sequencing and large mutation-screening programs. 1<br />

Regarding paired whole genome analysis of human breast<br />

cancer samples and their counterpart xenografts, our Human<br />

and Mouse Linked Evaluation of Tumors (HAMLET)<br />

program has successfully obtained over 30 breast tumor<br />

lines from poor prognosis subtypes (luminal B, basal-like,<br />

claudin-low, and HER2-enriched). As others have found,<br />

luminal A breast cancers are absent from these panels,<br />

possibly because their growth fraction is too low. Our initial<br />

report documented that the genome sequences of a breast<br />

primary tumor, a brain metastasis, and a xenograft derived<br />

from the primary tumor exhibit great similarities in their<br />

mutational repertoires; however, mutation frequencies<br />

were remodeled during the xenograft formation and metastatic<br />

process when compared with the primary tumor. 2<br />

Furthermore, new mutations were detected. Whole genome<br />

analysis of multiple, additional xenograft pairs reveal a<br />

similar pattern but considerable variation in the number of<br />

genome-wide, newly detected mutations. In several cases,<br />

the xenograft-specific mutations are likely to be functionally<br />

significant. These data suggest that serial monitoring of the<br />

xenograft genome will be necessary to ensure that new<br />

mutations have not arisen, producing inconsistencies in the<br />

therapeutic modelling data.<br />

Stock<br />

Ownership Honoraria<br />

Bioclassifier;<br />

University<br />

Genomics<br />

1. Ellis MJ, Ding L, Shen D, et al. Whole-genome analysis informs<br />

breast cancer response to aromatase inhibition. Nature. <strong>2012</strong>;486:<br />

353-360.<br />

34<br />

AstraZeneca;<br />

GlaxoSmithKline;<br />

Novartis<br />

Research<br />

Funding<br />

AstraZeneca;<br />

Novartis<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

2. Ding L, Ellis MJ, Shunqiang L et al. Genome remodelling in a<br />

basal-like breast cancer metastasis and xenograft. Nature. 2010;<br />

464:999-1005.


GENERAL SESSION VI:<br />

Health Care Delivery and Prevention<br />

CHAIR<br />

Bruce Haffty, MD<br />

The <strong>Cancer</strong> Institute of New Jersey<br />

SPEAKERS<br />

Benjamin O. Anderson, MD<br />

Fred Hutchinson <strong>Cancer</strong> Research Center, University of Washington<br />

Rowan T. Chlebowski, MD, PhD<br />

Los Angeles Biomedical Research Institute at Harbor-UCLA<br />

Los Angeles Medical Center<br />

After this session, attendees should be able to<br />

● More effectively integrate currently available evidence-based strategies to reduce breast cancer risk and mortality<br />

● Partner with advocacy and other support resources to address health care disparities in patient care


<strong>Breast</strong> Health Care Delivery in Context: The World Health<br />

Organization’s Initiative for Noncommunicable Diseases<br />

D<br />

espite the common misconception that breast cancer<br />

is primarily a problem of high-income countries,<br />

breast cancer affects countries at all economic levels. <strong>Breast</strong><br />

cancer is world’s most common cancer among women and<br />

the most likely reason that a woman will die of cancer<br />

around the globe, with over 50% of the 425,000 breast<br />

cancer deaths in 2010 occurred in developing, rather than<br />

developed, countries. The number of young lives lost to<br />

breast cancer is even more disproportionately distributed.<br />

In 2010, breast cancer killed 68,000 women age 15 to 49 in<br />

developing countries compared with 26,000 in developed<br />

countries. If cancer will seriously be addressed as a global<br />

health issue, then breast cancer in low- and middle-income<br />

countries (LMCs) cannot be ignored. 1<br />

The World Health Organization (WHO) reported that in<br />

2008, an estimated 36 million of the 57 million global deaths<br />

were due to noncommunicable diseases (NCDs), principally<br />

cardiovascular diseases, cancers, chronic respiratory diseases,<br />

and diabetes, including 9 million deaths before the<br />

age of 60, nearly 80% of which occurred in developing countries.<br />

In response, the United Nations (UN) held an historic<br />

high-level meeting from September 19 to 20, 2011, to consider<br />

the prevention and control of NCDs. According to<br />

WHO, the aim was to adopt “a concise, action-oriented<br />

outcome document that will shape the global agendas for<br />

generations to come.” 2 While the NCD political declaration<br />

mentions cancer generally, it provides no specific reference<br />

to breast cancer as a leading problem or to cancer treatment<br />

priorities in practice. The document promotes “increased<br />

access to cost-effective cancer screening programs, as determined<br />

by national situations” but otherwise gives no guidance<br />

about how diagnosed cancers should be managed. 3<br />

This UN “action-oriented outcome document” on NCDs<br />

is surprisingly silent on breast cancer. The UN and WHO<br />

have embraced tobacco control, arguably the lowest hanging<br />

fruit of cancer prevention, and discussed the prevention<br />

of infection-associated cancers like cervical and liver cancer<br />

through increased access to cost-effective hepatitis B (HBV)<br />

and human papillomavirus (HPV) vaccinations; however,<br />

they limited the discussion of common cancers that lack an<br />

infectious origin, like breast and colon cancers, to the modification<br />

of risk factors (unhealthy diet, obesity, and physical<br />

inactivity), interventions which are unlikely to make major<br />

shifts in breast cancer outcome. Prior to the UN summit,<br />

WHO released an executive summary, “Global Status Report<br />

on Noncommunicable Diseases 2010,” which outlines<br />

the core obstacles in addressing NCDs in LMCs. 4 This analysis<br />

notes that biennial mammographic screening (age 50 to<br />

70) to downstage disease combined with breast cancer treatment<br />

at all stages are among “the best buys” for health care<br />

interventions to tackle NCDs. The report asserts that these<br />

breast cancer interventions could avert 19% of the cancer<br />

36<br />

Benjamin O. Anderson, MD<br />

Fred Hutchinson <strong>Cancer</strong> Research Center,<br />

University of Washington, Seattle, WA<br />

burden, and, as such, is cost-effective. However, the document’s<br />

editor and principal author, Ala Alwan, MD, who<br />

then served as WHO Assistant Director-General of NCDs<br />

and Mental Health, concluded that breast cancer interventions<br />

are nonetheless impractical for poorer countries, both<br />

because of implementation costs and limited feasibility of<br />

treatment in the primary care setting in LMCs. Is this<br />

pessimistic perspective the end of the road for breast cancer<br />

in developing countries, or are there sequential steps that<br />

can be taken to address the most common cancer among<br />

women?<br />

<strong>Breast</strong> <strong>Cancer</strong> Early Detection Strategies:<br />

Screening versus Clinical Downstaging<br />

<strong>Breast</strong> cancer screening has been a source of heated debate<br />

among health care policy makers. The discussion about<br />

mammographic screening’s strengths, limitations, frequency,<br />

and target age group, which has relevance to high<br />

income countries with established health care infrastructures,<br />

has overshadowed more practical questions for addressing<br />

breast cancer outcomes in LMCs. Throughout the<br />

world, women in developing countries are most often first<br />

diagnosed with locally advanced (stage III) or metastatic<br />

(stage IV) cancer. These large, palpable, visible, or ulcerated<br />

cancers are easily detected on inspection and clinical examination,<br />

obviating the value of screen detection. Advanced<br />

cancers are more expensive to treat, less likely to respond<br />

to therapy, and most often prove incurable in any health<br />

care setting. The critical questions, then, must not revolve<br />

around mammographic availability in LMCs, but instead<br />

should focus on interventions whereby cancers can be diagnosed<br />

at less advanced stages, so-called “clinical downstaging,”<br />

followed by adequate locoregional and systemic<br />

treatments. 5<br />

It is critically important that WHO provide direct technical<br />

guidance to member states about breast cancer as a<br />

relevant NCD in LMCs. Education about early detection,<br />

breast lumps, and self-examination is not a costly intervention<br />

and may help a woman seek curative treatment while<br />

it is still realistic, rather than waiting until the tumor is<br />

advanced and likely untreatable except through palliative<br />

care. For countries to make informed decisions, they need<br />

meaningful data, including median tumor size at diagnosis,<br />

as a key, first step in assessing the real burden and<br />

options for solutions in addressing the most common lifethreatening<br />

female cancer on earth.<br />

As simple as it might sound, knowledge of median invasive<br />

tumor size provides a powerful indicator regarding the<br />

state of breast cancer detection and sheds light for early<br />

detection strategies in LMCs. Regions where median tumor<br />

size exceeds 3 to 4 cm are dominated by clinically detectible<br />

disease and therefore need to improve public knowledge


about breast cancer and access to health care facilities<br />

where cancer diagnoses can be made in an accurate and<br />

timely fashion. Practical interventions include outreach<br />

and public education about the signs and symptoms of<br />

breast cancer, the importance of early detection when it is<br />

still a small palpable mass or thickening, the promotion of<br />

breast self-examination (BSE), and the establishment of<br />

diagnostic tissue sampling and analysis to permit the accu-<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Anderson, Benjamin O. GE Healthcare;<br />

Navidea<br />

Biopharmaceuticals<br />

References<br />

1. Forouzanfar MH, Foreman KJ, Delossantos AM, et al. <strong>Breast</strong> and<br />

cervical cancer in 187 countries between 1980 and 2010: a systematic<br />

analysis. Lancet. 2011;378:1461-1484.<br />

2. United Nations high-level meeting on noncommunicable disease<br />

prevention and control. http://www.who.int/nmh/events/un_ncd_<br />

summit2011/en/. Accessed June 26, <strong>2012</strong>.<br />

3. Political Declaration of the High-level Meeting of the General<br />

rate and timely diagnosis of clinically detectible disease.<br />

Image detection of clinically occult cancers only becomes<br />

relevant when the median tumor size is less than 2 to 3 cm,<br />

and even then, the best methods for detection remain an<br />

important area of investigation. The decision of how to<br />

intervene in breast cancer will be made at the individual<br />

country level and must be adapted to existing resources.<br />

One size does not fit all.<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

GE Healthcare;<br />

Sanofi-Aventis<br />

EDUCATIONAL SUMMARIES<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

Assembly on the Prevention and Control of Noncommunicable<br />

Diseases (Resolution A/RES/66/2). http://www.un.org/en/ga/66/<br />

resolutions.shtml. Accessed June 26, <strong>2012</strong>.<br />

4. Alwan A, et al. Global status report on noncommunicable diseases<br />

2010. Geneva: WHO Press; <strong>2012</strong>.<br />

5. Anderson BO, Yip CH, Smith RA, et al. Guideline implementation<br />

for breast healthcare in low-income and middle-income countries:<br />

overview of the <strong>Breast</strong> Health Global Initiative Global Summit<br />

2007. <strong>Cancer</strong>. 2008;113:2221-2243.<br />

37


Reducing <strong>Breast</strong> <strong>Cancer</strong> Risk: Progress toward Resolution<br />

Rowan T. Chlebowski, MD, PhD<br />

Los Angeles Biomedical Research Institute at Harbor-UCLA<br />

Los Angeles Medical Center, Torrance, CA<br />

A<br />

series of randomized, full-scale clinical trials have recently<br />

addressed lifestyle changes in adjuvant breast<br />

cancer, changed concepts regarding menopausal hormone<br />

therapy and breast cancer, and identified new approaches to<br />

breast cancer risk reduction.<br />

The Women’s Intervention Nutrition Study (WINS) is a<br />

multicenter clinical trial for postmenopausal patients with<br />

early-stage resected breast cancer. Two thousand four hundred<br />

and thirty seven women were randomly assigned to<br />

standard therapy with or without a lifestyle intervention<br />

targeting fat intake. Intervention participants reduced dietary<br />

fat intake, lost weight, and experienced a significant<br />

increase in disease-free survival (hazard ratio [HR]: 0.76;<br />

95% CI [0.60-0.98]; p � 0.03). 1 Long-term follow-up is<br />

underway and other ongoing studies are evaluating the<br />

emerging hypothesis that weight loss along with increased<br />

physical activity will improve breast cancer outcome.<br />

The Women’s Health Initiative (WHI) hormone therapy<br />

program included two separate randomized, placebocontrolled<br />

trials. In one, 16,608 postmenopausal women<br />

with no prior hysterectomy were randomly assigned to estrogen<br />

plus progestin or placebo. In the second, 10,739<br />

postmenopausal women with prior hysterectomy were randomly<br />

assigned to estrogen alone or placebo. The use of<br />

estrogen plus progestin significantly increased breast cancer<br />

incidence, delayed breast cancer diagnosis, and increased<br />

breast cancer mortality (HR: 1.96; 95% CI [1.00-<br />

4.05]; p � 0.048). 2, 3 Surprisingly, the use of estrogen alone<br />

had an opposite effect, significantly reducing breast cancer<br />

incidence and reducing breast cancer mortality (HR: 0.37;<br />

95% CI [0.13-0.91]; p � 0.03). 4 The initial report of the<br />

adverse effects of estrogen plus progestin seen in the WHI<br />

trial led to a worldwide reduction in hormone-therapy use<br />

with substantial reduction in breast cancer incidence seen<br />

in many countries. The hypothesis was raised that the<br />

decrease in breast cancer was related to the decrease in<br />

hormone therapy. Analyses in the WHI of breast cancer<br />

incidence and mammography use after the estrogen plus<br />

progestin trial were stopped, identifying a rapid reduction<br />

in breast cancer incidence while mammography frequency<br />

was comparable between study groups and in the interval<br />

before and after the study intervention ended. 5 Preclinical<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

38<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Chlebowski, Rowan T. AstraZeneca;<br />

Novartis; Pfizer<br />

studies have suggested a hypothesis to explain the influence<br />

of estrogen alone in reducing breast cancer risk. Typically,<br />

estrogen stimulates breast cellular proliferation and inhibits<br />

apoptosis. However, after a period of estrogen deprivation,<br />

mammary tumor gene expression profile will change<br />

and estrogen then induces apoptosis. This finding suggests<br />

that many breast cancers in postmenopausal women can<br />

survive only a limited range of estrogen exposure. 4 While<br />

tamoxifen and raloxifene are both approved for breast cancer<br />

risk reduction, their use is uncommon due, in part, to<br />

toxicity concerns. As aromatase inhibitors have been found<br />

to have more breast cancer activity with more favorable<br />

toxicity profiles than tamoxifen, the MAP.3 trial evaluated<br />

exemestane versus placebo influence on breast cancer incidence<br />

in a primary prevention setting. After about 3 years<br />

of follow-up, breast cancer incidence was significantly lower<br />

in the exemestane versus placebo group (HR: 0.35; 95% CI<br />

[0.18-0.70]; p � 0.002). 6 The toxicity profile was quite favorable,<br />

and no life threatening side effect differences emerged<br />

when comparing study groups. Finally, several cohort analyses<br />

in the large WHI population have provided support for<br />

additional agents with a potential for breast cancer chemoprevention.<br />

In cohort analyses in 154,768 WHI study participants,<br />

breast cancer incidence was significantly lower in<br />

bisphosphonate users compared with nonusers in analyses<br />

adjusted for potential bone mineral density differences (HR:<br />

0.68; 95% CI [0.52-0.88]; p � 0.01). Most recently, in cohort<br />

analyses of 68,119 WHI clinical trial participants, women<br />

with diabetes using metformin had lower breast cancer<br />

incidence compared with women with diabetes treated with<br />

other agents (HR: 0.75; 95% CI [0.57-0.99]; p � 0.04).<br />

In a series of randomized clinical trials and emerging<br />

cohort studies, our understanding of estrogen plus progestin<br />

as compared with the influence of estrogen alone on<br />

breast cancer incidence in postmenopausal women has been<br />

altered. In addition, these studies have identified a range of<br />

interventions including aromatase inhibitor use, lifestyle<br />

change, and bisphosphonate and metformin—which can be<br />

used alone or in combination—to address breast cancer risk<br />

reduction. Their favorable side effect profiles suggest that<br />

even women at modest breast cancer risk could be considered<br />

for future intervention trials.<br />

Stock<br />

Ownership Honoraria<br />

AstraZeneca;<br />

Novartis<br />

Research<br />

Funding<br />

Amgen<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration


References<br />

1. Chlebowski RT, Blackburn G, Thomson CA, et al. Dietary fat<br />

reduction and breast cancer outcome: interim efficacy results<br />

from the Women’s Intervention Nutrition Study (WINS). J Natl<br />

<strong>Cancer</strong> Inst. 2006;98:1767-1776.<br />

2. Chlebowski RT, Hendrix SL, Langer RD, et al. Estrogen plus<br />

progestin influence on breast cancer and mammography in<br />

healthy postmenopausal women: the Women’s Health Initiative<br />

randomized trial. JAMA. 2003;289:3243-3253.<br />

3. Chlebowski RT, Anderson GL, Gass M, et al. Influence of estrogen<br />

plus progestin on breast cancer incidence and mortality. JAMA.<br />

2010;304:1684-1692.<br />

EDUCATIONAL SUMMARIES<br />

4. Anderson GL, Chlebowski RT, Aragaki AK, et al. Conjugated<br />

equine oestrogen and breast cancer incidence and mortality in<br />

postmenopausal women with hysterectomy: extended follow-up<br />

of the Women’s Health Initiative randomised placebo-controlled<br />

trial. Lancet Oncol. <strong>2012</strong>;13:476-486.<br />

5. Chlebowski RT, Kuller L, Prentice RL, et al. <strong>Breast</strong> cancer after<br />

estrogen plus progestin use in postmenopausal women. N Engl<br />

J Med. 2009;360:573-587.<br />

6. Goss PE, Ingle JN, Ales-Martinez J, et al. Exemestane for breastcancer<br />

prevention in postmenopausal women. N Engl J Med.<br />

2011;364:2381-2391.<br />

39


GENERAL SESSION VII:<br />

Survivorship<br />

CO-CHAIRS<br />

Kathy S. Albain, MD<br />

Loyola University Medical Center<br />

Sandra M. Swain, MD<br />

Washington <strong>Cancer</strong> Institute, Washington Hospital Center<br />

SPEAKERS<br />

Patrick G. Morris, MD, MSc<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center<br />

Patrick Neven, MD<br />

University Hospitals Leuven<br />

Denise J. O’Neill, BS<br />

Survivors Offering Support<br />

Ann H. Partridge, MD, MPH<br />

Dana-Farber <strong>Cancer</strong> Institute<br />

After this session, attendees should be able to<br />

● Outline a plan of care to advise patients how to transition into normal life after active treatment, including the<br />

importance of maintaining a healthy lifestyle and engaging in survivorship programs that connect similar individuals<br />

and provide ongoing support<br />

● Describe why survivors struggling with obesity have a greater risk for recurrence and recommend strategies to lengthen<br />

survival and improve quality of life, such as regular physical activity and healthy eating habits<br />

● Review recent advances in hormonal menopausal management and assist patients in selecting appropriate therapies<br />

that address menopausal symptoms while preserving the efficacy of cancer treatments already in place<br />

● Use quality measures to ensure that current clinical practices model best practices to support survivorship needs, such<br />

as coordination of care, physician-provider communication, support programs and services, as well as management of the<br />

long-term effects of treatment


Inflammation in the Pathogenesis and<br />

Progression of <strong>Breast</strong> <strong>Cancer</strong><br />

Patrick G. Morris, MD, MSc, Kotha Subbaramaiah, PhD,<br />

Andrew J. Dannenberg, MD, and Clifford A. Hudis, MD<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center and<br />

Weill Cornell Medical College, New York, NY<br />

O<br />

besity, with increasing worldwide incidence, is a risk<br />

factor for the development of several epithelial malignancies,<br />

including estrogen receptor (ER)– and progesterone<br />

receptor (PR)–positive breast cancer in postmenopausal<br />

women. 1,2 Following menopause, the conversion of androgen<br />

precursors in peripheral adipose tissue is the main<br />

source of estrogen synthesis. The rate-limiting step in this<br />

process is catalyzed by the enzyme aromatase, which is<br />

encoded by the CYP19 gene. 3 Circulating estrogens, such<br />

as estradiol and its derivatives, are known to have stimulatory,<br />

proliferative, and (possibly) genotoxic effects on breast<br />

epithelial cells. Therefore, the increased risk of ER/PRpositive<br />

breast cancer in obese, postmenopausal women<br />

has been partially attributed to higher circulating levels<br />

of estradiol, reflecting both increased adipose tissue and<br />

elevated aromatase expression within those tissues. 1,4,5<br />

However, the mechanisms underlying this obesity–breast<br />

cancer link have been incompletely understood.<br />

Obesity causes subclinical inflammation in adipose<br />

tissue. 2,6-8 In both mouse models of obesity and in obese<br />

humans, macrophages infiltrate visceral and subcutaneous<br />

adipose tissue around necrotic adipocytes to form characteristic<br />

crown-like structures (CLS). 7-10 These macrophages<br />

produce a variety of proinflammatory mediators, which are<br />

commonly found in the circulation of obese women and have<br />

been linked to breast cancer progression and mortality. 11-13<br />

Recently, our group demonstrated that in dietary and genetic<br />

mouse models of obesity, CLS occur in both the adipose<br />

tissue of the mouse mammary gland and the visceral<br />

fat. 14 Importantly, in these mouse models, the number of<br />

CLS lesions increases as a function of body weight, resulting<br />

in parallel increases in several proinflammatory mediators,<br />

which, in turn leads to increased aromatase expression.<br />

Collectively, this results in a complex system of paracrine<br />

interactions between macrophages, adipocytes, and other<br />

cells. In this proposed model (Fig. 1), obesity leads to increased<br />

lipolysis with resultant release of saturated fatty<br />

acids, which stimulates the transcription factor, nuclear<br />

factor kappa-B (NF-�B), to induce a series of proinflammatory<br />

mediators in macrophages, including tumor necrosis<br />

factor alpha (TNF-alpha), interleukin-1-beta (IL-1-beta),<br />

and cyclooxygenase-2 (COX-2). Critically, in the mammary<br />

gland and visceral fat of obese, oophorectomized mice, inflammatory<br />

mediators including COX-2-derived prostaglandin<br />

E2 (PGE2) activate the transcription of the CYP19<br />

gene thereby encoding aromatase in neighboring cells, leading<br />

to elevated expression and activity of aromatase. Consequently,<br />

estrogen biosynthesis should be enhanced, leading<br />

to upregulation of ER-target genes, including PR.<br />

In translational clinical studies, we and others recently<br />

described these same crown-like structures in the human<br />

breast (CLS-B). 15,16 Although these lesions can be seen with<br />

standard hematoxylin and eosin (H�E) staining in the<br />

white adipose tissue of the breast, the use of immunohistochemistry<br />

for CD68, a macrophage marker, increases detection<br />

(Fig. 2). 15 This observation may explain why the<br />

presence and biological significance of CLS-B has only recently<br />

been appreciated. Importantly, the presence and<br />

extent of breast inflammation, as evidenced by CLS-B, correlates<br />

with increasing body mass index (BMI). In a pilot<br />

study of 30 patients undergoing mastectomy, CLS-B were<br />

seen in one out of 12 (approximately 8%) women with<br />

normal BMI, seven out of 10 (70%) overweight women (BMI<br />

25-29.9), and six out of eight (75%) obese women (BMI �<br />

30). 15 Mirroring preclinical models, the presence of CLS-B<br />

was associated with higher NF-kappa-B-binding activity<br />

and increased levels of proinflammatory mediators (TNFalpha,<br />

IL-1-beta, COX-2, and PGE 2), CYP19 gene expression,<br />

and aromatase activity. 15,17 Although aromatase<br />

activity correlates with both BMI and the extent of CLS-B,<br />

the correlation appears to be stronger with extent of CLS-B,<br />

suggesting that obesity-associated inflammation rather<br />

than obesity alone is the critical driver of aromatase activation<br />

in breast tissue. Overall, these results establish the<br />

existence of an obesity-inflammation-aromatase axis occur-<br />

Fig. 1. Paracrine interactions between macrophages<br />

and other cell types in breast tissue of obese<br />

women and mice. 14 Reprinted with permission from<br />

the <strong>American</strong> Association for <strong>Cancer</strong> Research.<br />

41


EDUCATIONAL SUMMARIES<br />

Fig. 2. Immunohistochemical stain with CD68, a<br />

macrophage marker, showing a prototypic crownlike<br />

structure of the breast (CLS-B), consisting of a<br />

necrotic adipocyte surrounded by macrophages<br />

(200x). 15 Reprinted with permission from the <strong>American</strong><br />

Association for <strong>Cancer</strong> Research.<br />

ring locally in breast tissue that is likely to contribute to the<br />

increased risk of hormone receptor–positive breast cancer<br />

and its progression in postmenopausal women. 14,15,17<br />

Given the link between chronic inflammation and carci-<br />

Authors’ Disclosures of Potential Conflicts of Interest<br />

Author<br />

Morris, Patrick G.*<br />

Subbaramaiah, Kotha*<br />

Dannenberg, Andrew J.*<br />

Hudis, Clifford A.*<br />

*No relevant relationships to disclose.<br />

Employment or<br />

Leadership<br />

Positions<br />

References<br />

Consultant or<br />

Advisory Role<br />

1. Cleary MP, Grossmann ME. Minireview: obesity and breast<br />

cancer: the estrogen connection. Endocrinology. 2009;150:2537-<br />

2542.<br />

2. van Kruijsdijk RC, van der Wall E, Visseren FL. Obesity and<br />

cancer: the role of dysfunctional adipose tissue. <strong>Cancer</strong> Epidemiol<br />

Biomarkers Prev. 2009;18:2569-2578.<br />

3. Lorincz AM, Sukumar S. Molecular links between obesity and<br />

breast cancer. Endocr Relat <strong>Cancer</strong>. 2006;13:279-292.<br />

4. Key TJ, Appleby PN, Reeves GK, et al. Body mass index, serum<br />

sex hormones, and breast cancer risk in postmenopausal women.<br />

J Natl <strong>Cancer</strong> Inst. 2003;95:1218-1226.<br />

5. Wake DJ, Strand M, Rask E, et al. Intra-adipose sex steroid<br />

metabolism and body fat distribution in idiopathic human obesity.<br />

Clin Endocrinol (Oxf). 2007;66:440-446.<br />

6. Cancello R, Henegar C, Viguerie N, et al. Reduction of macrophage<br />

infiltration and chemoattractant gene expression changes<br />

in white adipose tissue of morbidly obese subjects after surgeryinduced<br />

weight loss. Diabetes. 2005;54:2277-2286.<br />

7. Cinti S, Mitchell G, Barbatelli G, et al. Adipocyte death defines<br />

42<br />

nogenesis in other tissues, 18 it is possible that obesityrelated<br />

breast inflammation, as indicated by the extent of<br />

CLS-B, contributes to the risk for other subtypes of breast<br />

cancer. For example, there are some data linking obesity<br />

with an increased risk of triple-negative breast cancer. 19 It<br />

is possible that chronic breast inflammation (CLS-B) contributes<br />

to this process by mechanisms other than induction<br />

of aromatase or that estrogen production facilitates transformation<br />

and invasion by nonhormonally dependent tumors.<br />

Possibly, obesity-induced inflammation manifested<br />

as CLS may also contribute to other types of malignancy.<br />

The discovery of the obesity-inflammation-aromatase<br />

connection provides a basis for the development of risk<br />

reduction and novel therapeutic strategies. For example,<br />

the effects of dietary modification and lifestyle interventions,<br />

such as exercise, on this inflammatory process are<br />

of considerable interest. Furthermore, the observation that<br />

breast inflammation leading to induction of COX-2 and<br />

PGE 2 occurs in most, but not all, overweight/obese women<br />

might explain inconsistent results seen in epidemiological<br />

studies in terms of the benefit of aspirin as a chemopreventive<br />

agent. It is possible that aspirin and other nonsteroidal<br />

anti-inflammatory drugs orprototypic inhibitors of COX<br />

enzymes, might only be of benefit to women with increased<br />

CLS-B–associated COX-2/PGE 2 signaling. Hence, these<br />

and other agents with anti-inflammatory properties might<br />

be of interest for future intervention studies that more<br />

specifically target the at-risk subset of subjects or patients<br />

with CLS-B.<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

macrophage localization and function in adipose tissue of obese<br />

mice and humans. J Lipid Res. 2005;46:2347-2355.<br />

8. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin<br />

resistance. Annu Rev Physiol. 2010;72:219-246.<br />

9. Murano I, Barbatelli G, Parisani V, et al. Dead adipocytes, detected<br />

as crown-like structures, are prevalent in visceral fat<br />

depots of genetically obese mice. J Lipid Res. 2008;49:1562-1568.<br />

10. Weisberg SP, McCann D, Desai M, et al. Obesity is associated<br />

with macrophage accumulation in adipose tissue. J Clin Invest.<br />

2003;112:1796-1808.<br />

11. Bachelot T, Ray-Coquard I, Menetrier-Caux C, et al. Prognostic<br />

value of serum levels of interleukin 6 and of serum and plasma<br />

levels of vascular endothelial growth factor in hormonerefractory<br />

metastatic breast cancer patients. Br J <strong>Cancer</strong>. 2003;<br />

88:1721-1726.<br />

12. Dandona P, Weinstock R, Thusu K, et al. Tumor necrosis factoralpha<br />

in sera of obese patients: fall with weight loss. J Clin<br />

Endocrinol Metab. 1998;83:2907-2910.<br />

13. Vozarova B, Weyer C, Hanson K, et al. Circulating interleukin-6<br />

in relation to adiposity, insulin action, and insulin secretion.<br />

Obes Res. 2001;9:414-417.


14. Subbaramaiah K, Howe LR, Bhardwaj P, et al. Obesity is associated<br />

with inflammation and elevated aromatase expression in<br />

the mouse mammary gland. <strong>Cancer</strong> Prev Res (Phila). 2011;4:329-<br />

346.<br />

15. Morris PG, Hudis CA, Giri D, et al. Inflammation and increased<br />

aromatase expression occur in the breast tissue of obese women<br />

with breast cancer. <strong>Cancer</strong> Prev Res (Phila). 2011;4:1021-1029.<br />

16. Sun X, Casbas-Hernandez P, Bigelow C, et al. Normal breast<br />

tissue of obese women is enriched for macrophage markers and<br />

macrophage-associated gene expression. <strong>Breast</strong> <strong>Cancer</strong> Res<br />

Treat. <strong>2012</strong>;131:1003-1012.<br />

EDUCATIONAL SUMMARIES<br />

17. Subbaramaiah K, Morris PG, Zhou XK, et al. Increased levels of<br />

COX-2 and prostaglandin E2 contribute to elevated aromatase<br />

expression in inflamed breast tissue of obese women. <strong>Cancer</strong><br />

Discov. <strong>2012</strong>;2:356-565.<br />

18. Karin M, Greten FR. NF-kappaB: linking inflammation and<br />

immunity to cancer development and progression. Nat Rev Immunol.<br />

2005;5:749-759.<br />

19. Phipps AI, Chlebowski RT, Prentice R, et al. Body size, physical<br />

activity, and risk of triple-negative and estrogen receptorpositive<br />

breast cancer. <strong>Cancer</strong> Epidemiol Biomarkers Prev. 2011;<br />

20:454-463.<br />

43


Managing Menopausal Symptoms in Patients<br />

with <strong>Breast</strong> <strong>Cancer</strong><br />

Patrick Neven, MD, and Anneleen Lintermans, MD<br />

University Hospitals Leuven, Leuven, Belgium<br />

I<br />

n the Western world, improved adjuvant treatments<br />

and earlier diagnosis increase the number of breast<br />

cancer survivors. Although many patients report positive<br />

aspects of the survivorship experiences, most want better<br />

symptom control of menopausal complaints. 1,2 In premenopausal<br />

women, certain breast cancer treatments (e.g.,<br />

chemotherapy, ovarian suppression) result in early and<br />

sudden onset of premature menopause. For postmenopausal<br />

women on hormone replacement therapy (HRT), a<br />

breast cancer diagnosis requires one to end treatment and<br />

thus symptoms return. Endocrine agents like tamoxifen<br />

and aromatase inhibitors (AIs) lower relapse from estrogen<br />

receptor–positive breast cancer but worsen acute menopausal<br />

symptoms.<br />

Menopausal symptoms during cancer treatment vary, but<br />

hot flashes, night sweats, arthralgia, and sexual dysfunction<br />

are most frequently reported after chemotherapy, ovarian<br />

suppression, tamoxifen, and oral AIs. These menopausal<br />

symptoms are also more severe in patients with breast<br />

cancer than they would be in healthy women experiencing<br />

natural menopause and often affect professional activities<br />

and quality of life to the point that patients will stop or<br />

consider stopping treatment if they do not receive help in<br />

relieving these symptoms. 3,4 The most effective treatment<br />

for menopausal symptoms is HRT, but cannot be used<br />

for patients with breast cancer. Most patients do not want<br />

to use drugs to treat side effects induced by drugs and,<br />

although nonhormonal medical treatments can reduce<br />

symptoms, side effects have to be anticipated. Nonpharmacological<br />

interventions have variable results but are often<br />

not better than placebo; however, there is some progress<br />

according to results from recent research. 5<br />

Recent literature from the last year shows that the medical<br />

community has invested a lot in the pathophysiology<br />

and management of menopausal symptoms to improve<br />

quality of life, which will hopefully improve treatment compliance.<br />

In focusing on that literature, we are able to discuss<br />

the management of acute menopausal symptoms.<br />

Vasomotor Symptoms<br />

Most patients with breast cancer experience hot flashes,<br />

night sweats, or both and report their symptoms as ranging<br />

from slightly disturbing to unbearable. Endocrine agents<br />

exacerbate symptoms, which compromise treatment adherence<br />

and impact treatment efficacy. 3,4 Management options<br />

remain symptomatic and large, randomized placebocontrolled<br />

trials to evaluate the numerous suggested<br />

remedies are scarce. Significant reductions in hot flash frequency<br />

and intensity are seen with the use of placebo. Even<br />

more reductions in hot flash management have been reported<br />

with selective serotonin reuptake inhibitors (SSRIs),<br />

serotonin-norepinephrine reuptake inhibitors (SNRIs), clonidine,<br />

and gabapentine when compared with placebo; how-<br />

44<br />

ever, side effects cause many patients outside of clinical<br />

trials to discontinue this supportive therapy prior to the<br />

next appointment. 6 Nonmedical treatments like adapting<br />

life style, exercise, dressing in layers, avoiding triggers<br />

(stress situations), carrying cold drinks, and moderating<br />

spicy food, tobacco, and caffeine may also offer some relief.<br />

Placebo-controlled clinical trials have failed to demonstrate<br />

a significant benefit when comparing complementary<br />

and alternative medicine with placebo. 7,8 For example, acupuncture<br />

and yoga has been suggested as a remedy for<br />

menopausal symptoms. 9 When vasomotor symptoms persist,<br />

promising results for short-term stellate ganglion block<br />

have been reported, although further evaluation in a randomized,<br />

placebo-controlled trial is necessary. 10 A recently<br />

published randomized study concluded that group cognitive<br />

behavior therapy (e.g., relaxation, paced respiration, and<br />

sleep advice) is a safe and effective treatment option for<br />

women to better tolerate their hot flashes and night sweats<br />

and provides additional benefits to mood, sleep, and quality<br />

of life. 11 Hormonal therapy, typically contraindicated in<br />

patients with breast cancer, may be used when other treatment<br />

modalities fail but only after careful consideration<br />

and informed consent, as this treatment might affect relapse<br />

rates.<br />

Vaginal Dryness and Sexual Dysfunction<br />

Vaginal dryness is mainly seen with AI use but tamoxifen<br />

can also induce vulvovaginal irritation due to excessive<br />

discharge. This may lead to dyspareunia and can enhance<br />

loss of libido, already affected by a recent breast cancer<br />

diagnosis and breast surgery. In the management of dyspareunia,<br />

nonhormonal therapy (e.g., lubricants, vaginal<br />

moisturizers) can be used to symptomatically treat vaginal<br />

dryness and prevent postcoital irritation. 6 Cold cream, an<br />

emulsion of water and certain fats, helps soften the vulval<br />

skin. Topical vaginal hormones are helpful but available<br />

data in AI users show increased circulating estradiol levels<br />

after vaginal application of estradiol. 12 In our department,<br />

our group uses off-label, low-dose vaginal estriol pessaries<br />

twice weekly for 4 weeks followed by weekly applications in<br />

patients who have failed nonhormonal therapy. 6 We participate<br />

in an international prospective clinical study of pharmacokinetics<br />

that looks at the efficacy and safety of vaginal<br />

application of lyophilised lactobacilli and 0.03 mg estriol<br />

(Gynoflor) on atrophic vaginitis in postmenopausal patients<br />

with breast cancer treated with nonsteroidal AIs. 13 Other<br />

modalities that require clinical trials in AI-treated patients<br />

with breast cancer include dehydroepiandrosterone vaginal<br />

ovules, intravaginal tamoxifen, ospemifene tablets, and intravaginal<br />

testosterone gel. Sexual dysfunction, in particular,<br />

decreased libido, as well as the inability to become<br />

aroused and achieve orgasm is a field that needs further


investigation. After 2 to 3 years of follow-up, sexual dysfunction<br />

is seen more in patients switching to an AI compared<br />

with patients who remain on tamoxifen; some consider this<br />

problem as more important than arthralgia. 14 Interventions<br />

at the psychosocial level, like education, lifestyle<br />

changes, and couples counseling can be helpful but efficacy<br />

data are lacking. 15,16<br />

Arthralgia and Bone Health<br />

Musculoskeletal symptoms, the most frequently reported<br />

complaint by AI users, now referred to as the AI-induced<br />

musculoskeletal syndrome (AIMSS), affects daily functioning<br />

and treatment adherence. 4,6,17 Pre-existing musculoskeletal<br />

symptoms should be assessed and promptly treated<br />

as they are likely to increase. There is fluid accumulation in<br />

peripheral joints and tendon sheaths thicken; patients report<br />

early morning stiffness and start pain, and hand-grip<br />

strength weakens. They suddenly feel like they are 100<br />

years old. 18 Patients and family should be educated concerning<br />

possible AI-induced joint problems. 19 Patients should<br />

be encouraged to adapt their lifestyle (weight control, exercise,<br />

physiotherapy, massage, etc.) when necessary. Randomized,<br />

controlled trials of exercise suggest beneficial<br />

changes in inflammation; however, the evidence is still<br />

preliminary. 20 Nonsteroidal anti-inflammatory drugs<br />

(NSAID), acetaminophen, and cyclooxygenase-2 inhibitors<br />

can be used if these adaptations fail to relieve the symptoms;<br />

however, adverse effects should be closely monitored. When<br />

Authors’ Disclosures of Potential Conflicts of Interest<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Neven, Patrick AstraZeneca;<br />

Lilly; Novartis;<br />

Pfizer; Roche<br />

Lintermans, Anneleen*<br />

*No relevant relationships to disclose.<br />

References<br />

1. Documet PI, Trauth JM, Key M, et al. <strong>Breast</strong> cancer survivors’<br />

perception of survivorship. Oncol Nurs Forum. <strong>2012</strong>;39:309-315.<br />

2. DL Stan, S Pruthi, Jenkins S, et al. Needs and preferences of<br />

breast cancer survivors: a cross-sectional survey. ASCO <strong>Breast</strong><br />

<strong>Cancer</strong> <strong>Symposium</strong> 2011.<br />

3. Banning M. Adherence to adjuvant therapy in post-menopausal<br />

breast cancer patients: a review. Eur J <strong>Cancer</strong> Care (Engl). <strong>2012</strong>;<br />

21:10-19.<br />

4. Henry NL, Azzouz F, Desta Z, et al. Predictors of aromatase<br />

inhibitor discontinuation as a result of treatment-emergent<br />

symptoms in early-stage breast cancer. J Clin Oncol. <strong>2012</strong>;30:<br />

936-942.<br />

5. Mann E, Smith MJ, Hellier J, et al. Cognitive behavioral treatment<br />

for women who have menopausal symptoms after breast<br />

cancer treatment (MENOS 1): a randomized controlled trial.<br />

Lancet Oncol. <strong>2012</strong>;13:309-318.<br />

6. Loibl S, Lintermans A, Dieudonné AS, et al. Management of<br />

menopausal symptoms in breast cancer patients. Maturitas.<br />

2011;68:148-154.<br />

7. Pockaj BA, Gallagher JG, Loprinzi CL, et al. Phase III doubleblind<br />

randomized, placebo-controlled crossover trial of black<br />

not contraindicated, maximum start dosage of NSAID with<br />

stomach protection should be prescribed, and efficacy of<br />

reduced doses can be evaluated.<br />

A drug holiday or switching to another AI may offer pain<br />

relief in some patients. 21,22 When symptoms persist, switching<br />

to tamoxifen can be another option. Patients suffering<br />

from carpal tunnel syndrome (nine times more likely in AI<br />

than tamoxifen users) or those with arthritis should be<br />

referred to a specialist for proper diagnosis and therapy. 23<br />

New therapeutic strategies under investigation include<br />

glucosamine, diuretics, duloxetine, probiotics, vitamin D,<br />

capsaicin, acupuncture, and yoga. However, randomized,<br />

placebo-controlled studies are warranted to establish the<br />

safety and efficacy of these interventions. Notably, recent<br />

data from the VITAL trial in 147 letrozole users shows<br />

promising results for adding high doses of vitamin D. 24<br />

In summary, a subset of patients with breast cancer<br />

experience severe, acute menopausal symptoms mainly related<br />

to endocrine agents that need to be taken for at least 5<br />

years. Patients, already bothered by the diagnosis and previous<br />

treatments, are confronted with the emergence of<br />

such symptoms and the severity of these symptoms can<br />

threaten treatment compliance. 25 Further research in the<br />

pathophysiology of symptoms like AIMSS is required 26 and,<br />

in the mean time, management strategies to tackle menopausal<br />

symptoms are important to get a maximal benefit<br />

from adjuvant therapy.<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

EDUCATIONAL SUMMARIES<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

cohosh in the management of hot flashes: NCCTG Trial N01CC1.<br />

J Clin Oncol. 2006;24:2836-2841.<br />

8. Penotti M, Fabio E, Modena AB, et al. Effect of soy-derived<br />

isoflavones on hot flushes, endometrial thickening, and the pulsatility<br />

index of the uterine and cerebral arteries. Fertil Steril.<br />

2003;79:1112-1117.<br />

9. Hervik J, Mjaland O. Quality of life of breast cancer patients<br />

mediated with anti-estrogens, 2 years after acupuncture treatment:<br />

a qualitative study. Eur J <strong>Cancer</strong>. <strong>2012</strong>;48:5148.<br />

10. Haest K, Kumar A, Van Calster B, et al. Stellate ganglion block<br />

for the management of hot flashes and sleep disturbances in<br />

breast cancer survivors: an uncontrolled experimental study<br />

with 24 weeks of follow-up. Ann Oncol. <strong>2012</strong>;23:1449-1454.<br />

11. Mann E, Smith MJ, Hellier J, et al. Cognitive behavioural treatment<br />

for women who have menopausal symptoms after breast<br />

cancer treatment (MENOS 1): a randomised controlled trial.<br />

Lancet Oncol. <strong>2012</strong>;13:309-318.<br />

12. Moegele M, Buchholz S, Seitz S, et al. Vaginal estrogen therapy<br />

in postmenopausal breast cancer patients treated with aromatase<br />

inhibitors. Arch Gynecol Obstet. <strong>2012</strong>;285:1397-1402.<br />

13. A clinical study of pharmacokinetics, efficacy and safety of vaginal<br />

application of lyophilised lactobacilli and 0.03 mg estriol<br />

45


EDUCATIONAL SUMMARIES<br />

(Gynoflor®) on atrophic vaginitis in postmenopausal breast cancer<br />

patients treated with aromatase inhibitors. 2011. NCT01370551.<br />

14. Van Calster B, Van Ginderachter J, Vlasselaer J, et al. Uterine<br />

and quality of life changes in postmenopausal women with an<br />

asymptomatic tamoxifen-thickened endometrium randomized<br />

to continuation of tamoxifen or switching to anastrozole. Menopause.<br />

2011;18:224-229.<br />

15. Krychman ML, Katz A. <strong>Breast</strong> cancer and sexuality: multi-modal<br />

treatment options. J Sex Med. <strong>2012</strong>;9:5-15.<br />

16. Bulletins-Gynecology CoP. ACOG Practice Bulletin No. 126:<br />

Management of gynecologic issues in women with breast cancer.<br />

Obstet Gynecol. <strong>2012</strong>;119:666-682.<br />

17. Mieog JS, Morden JP, Bliss JM, et al. Carpal tunnel syndrome<br />

and musculoskeletal symptoms in postmenopausal women with<br />

early breast cancer treated with exemestane or tamoxifen after<br />

2-3 years of tamoxifen: a retrospective analysis of the Intergroup<br />

Exemestane Study. Lancet Oncol. <strong>2012</strong>;13:420-432.<br />

18. Winters L, Habin K, Flanagan J, et al. “I feel like I am 100 years<br />

old!” managing arthralgias from aromatase inhibitors. Clin J<br />

Oncol Nurs. 2010;14:379-382.<br />

19. Tanna N, Buijs H, Pitkin J. Exploring the breast cancer patient<br />

journey: do breast cancer survivors need menopause management<br />

support? Menopause Int. 2011;17:126-131.<br />

20. Ballard-Barbash R, Friedenreich CM, Courneya KS, et al. Phys-<br />

46<br />

ical activity, biomarkers, and disease outcomes in cancer survivors:<br />

a systematic review. J Natl <strong>Cancer</strong> Inst. <strong>2012</strong>.<br />

21. Briot K, Tubiana-Hulin M, Bastit L, et al. Effect of a switch of<br />

aromatase inhibitors on musculoskeletal symptoms in postmenopausal<br />

women with hormone-receptor-positive breast cancer:<br />

the ATOLL (articular tolerance of letrozole) study. <strong>Breast</strong><br />

<strong>Cancer</strong> Res Treat. 2010;120:127-134.<br />

22. Henry NL, Azzouz F, Desta Z, et al. Predictors of aromatase inhibitor<br />

discontinuation as a result of treatment-emergent symptoms in<br />

early-stage breast cancer. J Clin Oncol. <strong>2012</strong>;30:936-942.<br />

23. Din OS, Dodwell D, Winter MC, et al. Current opinion of aromatase<br />

inhibitor-induced arthralgia in breast cancer in the UK. Clin<br />

Oncol (R Coll Radiol.) 2011;23:674-680.<br />

24. Khan Q, Kimler B, Reddy P. Randomized trial of vitamin D3 to<br />

prevent worsening of musculoskeletal symptoms and fatigue<br />

in women with breast cancer starting adjuvant letrozole: the<br />

VITAL trial. J Clin Oncol. <strong>2012</strong>;30(suppl; abstr 9000).<br />

25. Wagner LI, Zhao F, Chapman JW, et al. Patient-reported predictors<br />

of early treatment discontinuation: NCIC JMA.27/E1Z03<br />

quality of life study of postmenopausal women with primary<br />

breast cancer randomized to exemestane or anastrozole. San<br />

Antonio <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong>. <strong>Cancer</strong> Res. 2011;71(24<br />

suppl; abstrS6–2).<br />

26. Lintermans A, Neven P. Pharmacology of arthralgia with estrogen<br />

deprivation. Steroids. 2011;76:781-785.


The Transition from Active Treatment to<br />

Normal Life as a Survivor<br />

Survivors Offering Support (SOS) functions within 12<br />

hospitals in Maryland and Washington, DC, and is<br />

funded by each respective hospital and a grant from the<br />

Susan G. Komen for the Cure Maryland affiliate. SOS contracts<br />

with hospitals to establish peer-to-peer breast cancer<br />

support services that are integrated into the standard of<br />

care within each hospital. Since 2004, SOS has assisted over<br />

3,800 women diagnosed with breast cancer. While peer<br />

mentoring serves as the cornerstone of the SOS program,<br />

the quest for each patient to process her own personal<br />

cancer experience and develop a strategy to successfully<br />

transition into life after treatment has led to the development<br />

of a workshop called Transition to Wellness (Transitions).<br />

This “jump start into survivorship” workshop has<br />

bridged the gap for patients moving from active treatment<br />

to life as a breast cancer survivor. With 2.6 million breast<br />

cancer survivors living in the United States today, 1 survivorship<br />

care programs that provide information, practical<br />

knowledge, and role modeling to assist patients in developing<br />

healthy, new-normal skills for life after cancer are imperative.<br />

Transitions is targeted at patients completing treatment<br />

and moving into the extended survivorship phase. 2 Attendance<br />

is highest at hospitals where medical staff integrate<br />

the workshop with survivorship care plans and strongly<br />

encourage patients to attend in order to prepare for life as a<br />

cancer survivor. <strong>Breast</strong> cancer survivors share similar concerns<br />

with survivors of other cancers. SOS surveys mirror<br />

findings of the 2010 LIVESTRONG Report 3 with: fear of<br />

recurrence; sadness and depression; and lowered energy<br />

and concentration as concerns shared by over 50% of survivors.<br />

SOS surveys of 210 patients over the past 2 years<br />

indicate a need for information in critical areas like nutrition,<br />

self-advocacy, long-term follow-up care, exercise, and<br />

sexual intimacy. Transitions participants indicate a desire<br />

to make positive changes in their lives and to improve their<br />

future health and well-being by using the information presented<br />

at workshops by peer mentors. Over a third of participants<br />

indicate a desire to improve both diet and exercise.<br />

Reducing anxiety and stress levels were noted as being<br />

important to 25% of survey respondents.<br />

Survivorship Care Models<br />

As survivorship care models evolve, programs may vary<br />

from higher-cost, vertical hospital-based programs typically<br />

run in large university hospital settings to lower-cost, horizontal<br />

programs where managed survivorship-care plans<br />

are augmented by partnerships with independent not-forprofit<br />

organizations. Partner organizations like SOS offer<br />

specialized programs for survivors in the areas of peer support<br />

and role modeling, exercise, nutrition, integrative medicine,<br />

and psychosocial counseling. Economic constraints<br />

Denise J. O’Neill, BS<br />

Survivors Offering Support<br />

enhance the viability of the horizontal-partnering model of<br />

survivorship care and leverage nonmedical-based program<br />

cost efficiencies.<br />

SOS Transitions Overview<br />

The SOS Transitions workshops utilize trained peer mentors<br />

to share information vetted by medical staff and act as<br />

behavioral models for patients. The 3-hour workshop is<br />

designed as a starting point for transitional survivors and<br />

offers insights and practical knowledge for the challenges<br />

that patients may face as they enter life after cancer treatment.<br />

The workshop has three sections. First, a survivor<br />

panel with similar surgery and treatment experiences as the<br />

survivor audience shares their own fears and difficulties of<br />

early survivorship while imparting coping strategies and<br />

success stories. Topics like the fear of recurrence, low energy,<br />

sadness, and life-style changes are discussed with a<br />

trained moderator highlighting key information for participants.<br />

The panel is followed by the moderator sharing 10<br />

healthy lifestyle ideas for breast cancer survivors. The<br />

Healthy Lifestyle Checklist includes topics like limiting alcohol,<br />

smoking avoidance, and the importance of exercise,<br />

nutrition, and sleep. The lifestyle overview provides information<br />

that encourages survivors to begin to take control of<br />

their lives after treatment.<br />

The trained peer moderator then shares information<br />

about the survivor’s role as an advocate of their own care<br />

plan. Key topic areas include maintaining a medical appointment<br />

calendar and the importance of adhering to<br />

follow-up care appointments and tests. Discussion about<br />

selecting a lead physician, communicating side effects with<br />

medical staff, and complying with daily hormonal therapy<br />

medications are also included.<br />

The workshop ends with a brief presentation and demonstration<br />

from a wellness practitioner who shares how their<br />

particular practice can improve a patient’s overall health<br />

and well-being. A one-page survey is completed by the par-<br />

Sidebar. Survey Comments from Survivors<br />

Offering Support<br />

“Transitions was educational and made me think differently<br />

about moving forward.”<br />

“It consolidated so many issues, strategies, and challenges<br />

that I need to consider...very hands on.”<br />

“It helped me organize from treatment plan to life<br />

beyond.”<br />

“Transitions left me less anxious with a more positive<br />

and hopeful outlook.”<br />

47


EDUCATIONAL SUMMARIES<br />

ticipants listing what has been learned, and what changes, if<br />

any, may be incorporated into a personal survivorship action<br />

plan. The surveys are mailed back to participants 6<br />

months later with a letter that encourages each survivor to<br />

review their progress on their action plans.<br />

Surveys indicate that over 90% of attendees benefited<br />

from the peer-led workshop and that the timing of the<br />

workshop was critical. Peer survivors are uniquely qualified<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

O’Neill, Denise J.*<br />

*No relevant relationships to disclose.<br />

Employment or<br />

Leadership<br />

Positions<br />

References<br />

Consultant or<br />

Advisory Role<br />

1. National <strong>Cancer</strong> Institute. U.S. cancer survivors grows to nearly<br />

12 million. http://www.cancer.gov/newscenter/pressreleases/2011/<br />

survivorshipMMWR2011. Accessed May18, <strong>2012</strong>.<br />

2. Dana-Farber <strong>Cancer</strong> Institute. 2010 Spring/Summer Paths of<br />

48<br />

to share wisdom and information that can decrease patient<br />

anxiety, provide direction, and reinforce compliance of medically<br />

supervised care plans.<br />

In summary, trained peer survivors can successfully assist<br />

patients to navigate from active treatment into life as a<br />

breast cancer survivor. Peer survivor-led workshops may be<br />

a cost-effective way for hospitals to enhance future survivorship<br />

care.<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

Progress. Kenneth Miller, MD. http://www.dana-farber.org/News<br />

room/Publications/<strong>Cancer</strong>-s-Seasons-of-Survivorship-.aspx. Accessed<br />

May 18, <strong>2012</strong>.<br />

3. LIVESTRONG. How <strong>Cancer</strong> Has Affected Post-Treatment Survivors:<br />

A LIVESTRONG Report. June, 2010. http://www.livestrong.<br />

org/pdfs/3-0/LSSurvivorSurveyReport. Accessed April 30, <strong>2012</strong>.


GENERAL SESSION VIII:<br />

The Future of Health Policy<br />

CHAIR<br />

Richard L. White Jr., MD<br />

Carolinas Medical Center<br />

SPEAKERS<br />

Claudia R. Baquet, MD, MPH<br />

University of Maryland School of Medicine<br />

Gary H. Lyman, MD, MPH<br />

Duke University and the Duke <strong>Cancer</strong> Institute<br />

Sandra M. Swain, MD<br />

Washington <strong>Cancer</strong> Institute, Washington Hospital Center<br />

After this session, attendees should be able to<br />

● Evaluate current practice and suggest strategies that could be implemented to reduce the potential for health care<br />

disparities for patients at risk<br />

● Compare and contrast the processes used in changing practice standards to include a new drug versus a new technology<br />

● Describe how Rapid Learning Systems can be used to improve the speed of clinical research and adoption of evidencebased<br />

medicine in the oncology field and evaluate the potential of these systems to impact patient care and patient<br />

outcomes


What Are the Differences between Approving and<br />

Using New Drugs and New Devices<br />

in the United States?<br />

Gary H. Lyman, MD, MPH<br />

Duke University and the Duke <strong>Cancer</strong> Institute, Durham, NC<br />

W<br />

hy does new drug development in oncology take so<br />

long and cost so much whereas a new technology,<br />

such as a device, test, diagnostic, or prognostic tool, undergoes<br />

much less scrutiny and may get approved with no<br />

demonstrated clinical validity or utility with an investment<br />

of fewer resources? To better understand these differences,<br />

it is important to review the process by which the United<br />

States makes decisions on therapeutic/diagnostic modalities<br />

and how these differences influence the utilization of these<br />

interventions. Regulation of drugs and devices at the federal<br />

level in the United States is handled by different agencies.<br />

Drugs and biologics are governed by the Centers for Drug<br />

Evaluation and Research (CDER) and Center for Biologics<br />

Evaluation and Research (CBER), respectively, and devices<br />

are handled by the Center for Devices and Radiological<br />

Health (CDRH). A medical device is defined by the U.S.<br />

Food and Drug Administration (FDA) as any health care<br />

product that does not work through chemical action or by<br />

metabolism and can include diagnostic aids such as in vitro<br />

tests.<br />

History of Drug and Device Approval in the<br />

United States<br />

Prior to 1900, there were essentially no laws or regulations<br />

governing the sale or use of drugs or devices in the United<br />

States. In 1906, the Federal Food and Drug Act was passed<br />

preventing interstate transport of drugs that did not meet<br />

certain standards. 1 However, it was not until the Federal<br />

Food, Drug, and Cosmetic Act of 1938 that a formal process<br />

for obtaining approval by the FDA was established based on<br />

the demonstration of safety. 2 Following the thalidomide<br />

disaster in 1962, applicants for new drug approvals were<br />

required to demonstrate efficacy as well as some information<br />

on safety. 2,3 Finally, it was not until 1976 that sponsors<br />

were required to register new devices with the FDA. Although<br />

subsequent legislation has further strengthened<br />

and modernized these regulations, the oversight of medical<br />

devices has always lagged behind drug regulation.<br />

Current Regulation of Drugs and Devices in<br />

the United States<br />

Today, to conduct research on new drugs or biologics in<br />

humans, sponsors must submit an investigational new drug<br />

(IND) application. Clinical studies can then be initiated<br />

unless the FDA places a hold on the application within<br />

30 days of submission. For regulatory approval, sponsors<br />

of new drugs must submit a new drug application (NDA).<br />

An abbreviated new drug application (ANDA) can be submitted<br />

for generic drugs not requiring clinical trials but<br />

only demonstration of bioequivalence to the innovator<br />

agent. Following often extensive preclinical work, a new<br />

50<br />

drug must demonstrate substantial evidence of effectiveness<br />

and safety in at least one controlled clinical trial. Since<br />

1992, drugs or biologics for treating serious diseases can be<br />

granted accelerated approval based on surrogate endpoints<br />

that are reasonably likely to predict clinical benefit. Such<br />

approval is conditional on subsequent clinical studies confirming<br />

the magnitude of benefit and safety seen with<br />

the early studies. Accelerated approval has been granted to<br />

35 oncology products over the past 20 years. The entire<br />

drug development and approval process takes an average<br />

of 8 to 10 years and sometimes longer. 2,4 Still to be determined<br />

are the detailed processes for seeking approval of a<br />

biosimilar or bioequivalent to approved innovator biologic<br />

agents. 5,6<br />

Following registry with the FDA, the risk of medical<br />

devices is then classified as Class I (lowest risk) through<br />

Class III (highest risk). 3 The risk determined by the CDRH<br />

will determine the appropriate pathway the device sponsor<br />

must follow in seeking approval. By law, most Class I and<br />

some Class II devices are considered exempt from formal<br />

review. The majority of Class II devices go through a standard<br />

Premarket Notification as a 501[k] submission, which<br />

requires demonstration of equivalence to a previous device<br />

performing in a similar fashion with equal or better efficacy<br />

and safety. Because demonstration of equivalence rarely<br />

involves an actual clinical trial, once complete, the device is<br />

said to be FDA-cleared but not approved. A new device not<br />

demonstrated to be substantially equivalent to a prior device<br />

will be placed in Class III. Most Class III devices go<br />

through a more thorough Premarket Application (PMA),<br />

which may require preclinical and interventional clinical<br />

trials demonstrating reasonable assurance of effectiveness<br />

and safety. This is often based on a single study with valid<br />

scientific evidence demonstrating that the likely benefits<br />

outweigh risks.<br />

Similarities and Differences between Drug<br />

and Devices Regulation and Marketing<br />

Review and approval of drugs and devices are similar in a<br />

number of ways. All medical devices must be registered with<br />

the FDA and are reviewed for adulteration or misbranding. 3<br />

There is a process for both drugs and devices to be studied in<br />

humans either through an IND or an investigational device<br />

exemption (IDE). Review of both drugs and devices by the<br />

FDA is required. Cleared or approved drugs and devices<br />

must adhere to approved regulations for production, labeling,<br />

advertising, and postapproval surveillance, and they<br />

can be marketed only for their intended purpose. 3 Whereas<br />

all drugs and many devices must demonstrate both safety<br />

and efficacy, neither is currently required to demonstrate


enefit over existing approaches, and neither needs to show<br />

cost-effectiveness for their intended use.<br />

However, FDA review and approval of drugs and devices<br />

differ in a number of other ways. This explains, in part, the<br />

considerable differences in the time and resources required<br />

to bring these products to market. Although all medical<br />

devices must register with the FDA and adhere to good<br />

manufacturing practices and proper labeling, most are marketed<br />

in the United States through a much less demanding<br />

regulatory process than that imposed on drugs. Intervention<br />

trials are generally not required for diagnostic devices.<br />

Therefore, FDA allowance of low- or intermediate-risk medical<br />

devices does not imply that clinical trials have been<br />

conducted or that efficacy and safety have been established<br />

based on a clinical study prior to market approval similar to<br />

that required of drugs. Only 2% of medical devices are<br />

approved through the PMA process, and only rarely have<br />

randomized controlled trials been required prior to approval<br />

of high risk devices under a PMA. 3 Finally, it should<br />

also be noted that manufacturers of drugs are required to go<br />

through FDA inspections while the same is not true for<br />

manufacturers of medical devices.<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Conclusions<br />

Employment or<br />

Leadership Consultant or Stock<br />

Research<br />

Author<br />

Positions Advisory Role Ownership Honoraria Funding<br />

Lyman, Gary H. Amgen<br />

References<br />

1. Hyman PM, Carvajal R. Drugs and other product choices. Dermatol<br />

Ther. 2009;22:216-224.<br />

2. Moore SW. An overview of drug development in the United States<br />

and current challenges. South Med J. 2003;96:1244-1255.<br />

3. Sweet BV, Schwemm AK, Parsons DM. Review of the processes for<br />

FDA oversight of drugs, medical devices, and combination products.<br />

J Manag Care Pharm. 2011;17:40-50.<br />

4. Lipsky MS, Sharp LK. From idea to market: the drug approval<br />

process. J Am Board Fam Pract. 2001;14:362-367.<br />

5. Hirsch BR, Lyman GH. Biosimilars: are they ready for prime-<br />

EDUCATIONAL SUMMARIES<br />

The reasons for the major differences in the review and<br />

monitoring of drugs and devices in the United States are<br />

multiple and include legislative restrictions, study design<br />

challenges, and ethical considerations. Likewise, the great<br />

diversity of devices developed suggests that not all require<br />

the same level of scrutiny. Finally, review and monitoring of<br />

the enormous volume of device submissions challenge current<br />

FDA resources. 7,8 Although the market for drugs in the<br />

United States far exceeds that for devices, nearly five times<br />

as many new device submissions are received each year as<br />

new drug applications. At the same time, nearly six-fold<br />

more reports of adverse events from drugs are reported to<br />

the FDA annually than from devices. Clearly, the review<br />

and approval processes and appropriate postmarketing<br />

monitoring of drugs and biologics as well as medical devices<br />

in the United States needs to be reevaluated and appropriately<br />

resourced. Doing so will provide efficient and safe<br />

oversight of an ever-expanding portfolio of diagnostic and<br />

therapeutic interventions available for the management of<br />

patients with cancer.<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

time in the United States? J Natl Compr Canc Netw. 2011;9:934-<br />

942.<br />

6. Zelenetz AD, Ahmed I, Braud EL, et al. NCCN Biosimilars White<br />

Paper: regulatory, scientific, and patient safety perspectives.<br />

J Natl Compr Canc Netw. 2011;9 (Suppl 4):S1–22.<br />

7. Feldman MD, Petersen AJ, Karliner LS, et al. Who is responsible<br />

for evaluating the safety and effectiveness of medical devices?<br />

The role of independent technology assessment. J Gen Intern<br />

Med. 2008;23 (Suppl 1):57-63.<br />

8. Ramsey SD, Luce BR, Deyo R, et al. The limited state of technology<br />

assessment for medical devices: facing the issues. Am J Manag<br />

Care. 1998;4:SP188–199.<br />

51


GENERAL SESSION IX:<br />

New Directions in Systemic Therapy for Advanced Disease<br />

CHAIR<br />

Heather McArthur, MD<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center<br />

SPEAKERS<br />

Lisa A. Carey, MD<br />

Lineberger Comprehensive <strong>Cancer</strong> Center<br />

Francisco J. Esteva, MD, PhD<br />

University of Texas M. D. Anderson <strong>Cancer</strong> Center<br />

Mark D. Pegram, MD<br />

Stanford University<br />

After this session, attendees should be able to<br />

● Assess current advances in HER2-positive breast cancer research, focusing on the implications that new data may have<br />

on the standard of care for patients presenting with this genetic variant<br />

● Review the prevailing systemic therapies available for the treatment of hormone-responsive breast cancer and recommend<br />

appropriate treatment options to patients based on currently available evidence<br />

● Describe the unique considerations for patients presenting with triple-negative breast cancer and recommend the most<br />

appropriate treatment strategies based on current evidence


Hormone-Responsive Disease<br />

Francisco J. Esteva, MD, PhD<br />

University of Texas M. D. Anderson <strong>Cancer</strong> Center, Houston, TX<br />

E<br />

ndocrine therapy is the safest and most effective therapeutic<br />

option for patients with estrogen receptor<br />

(ER)- and/or progesterone receptor (PR)-positive metastatic<br />

breast cancer. Traditionally, for a breast cancer to be considered<br />

ER-positive or PR-positive, these receptors had to be<br />

expressed in at least 10% of invasive cancer cells. In 2010,<br />

the <strong>American</strong> Society of Clinical Oncology recommended<br />

that tumors should be considered ER- and/or PR-positive<br />

if at least 1% of breast cancer cells stained positive using<br />

immunohistochemistry. 1 This guideline was based on limited<br />

retrospective data 2 and it remains controversial. 3 In<br />

premenopausal women, tamoxifen with or without luteinizing<br />

hormone-releasing hormone agonists remains the<br />

standard of care. In postmenopausal women, aromatase<br />

inhibitors are the most effective front-line therapy. For<br />

patients treated with anastrozole or letrozole in the adjuvant<br />

setting, exemestane or fulvestrant exhibit similar efficacy<br />

in the front-line metastatic setting. 4<br />

Until recently, combination endocrine therapy was not<br />

superior to single agents, and the latter was the preferred<br />

approach. Several trials reported in the last 12 months have<br />

challenged this concept, including the FACT, SWOG-S0226,<br />

TAMRAD, and BOLERO-2 trials.<br />

Bergh and collaborators compared the efficacy of anastrozole<br />

versus a combination of fulvestrant and anastrozole in<br />

women in the first relapse of endocrine-responsive breast<br />

cancer (FACT trial). 5 In this study, postmenopausal<br />

women, or premenopausal women receiving a gonadotropinreleasing<br />

hormone agonist, with ER- and/or PR-positive<br />

breast cancer at first relapse after primary treatment of<br />

localized disease were randomly assigned treatment with<br />

fulvestrant (loading dose followed by 250 mg monthly)<br />

plus 1 mg of anastrozole daily compared with only 1 mg<br />

of anastrozole daily. Overall survival was similar in both<br />

groups. It should be noted, however, that a higher dose of<br />

fulvestrant (500 mg monthly) is more effective than the<br />

dose used in this trial. 6<br />

Mehta and colleagues reported preliminary results of the<br />

SWOG-S0226 trial at the 2011 <strong>Cancer</strong> Therapy and Research<br />

Center (CTRC)-<strong>American</strong> Association of <strong>Cancer</strong> Research<br />

(AACR) San Antonio <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong>. 7 In<br />

this study, postmenopausal women with ER- and/or PRpositive<br />

metastatic breast cancer were randomly assigned to<br />

receive single agent anastrozole or anastrozole plus fulvestrant.<br />

The progression-free and overall survival among the<br />

694 patients studied were 1.5 and 6.4 months longer, respectively,<br />

for those randomly assigned to anastrozole plus<br />

fulvestrant compared with those given anastrozole alone.<br />

Unplanned subgroup analyses suggested that these benefits<br />

were largely restricted to women who had not received<br />

adjuvant tamoxifen.<br />

The study populations on the FACT and SWOG-S0226<br />

trials were quite different. Most patients who participated<br />

in SWOG-S0226 had not received prior endocrine therapy<br />

and were therefore more likely to respond to the combination<br />

therapy than patients who had previously received<br />

hormone therapy (e.g., FACT). Furthermore, crossover occurred<br />

more frequently on the FACT trial because the<br />

SWOG-S0226 protocol did not allow crossing over to combination<br />

therapy. Taken together, these data support the<br />

testing of anastrozole plus fulvestrant in the adjuvant setting<br />

in patients with ER- and PR-positive breast cancer.<br />

The mTOR pathway plays an important role in the development<br />

of resistance to endocrine therapy. 8 Everolimus is<br />

an oral mTOR inhibitor that has been shown to be effective<br />

in patients with renal cell carcinoma, human epidermal<br />

growth factor receptor 2 (HER2)–overexpressing breast<br />

cancer, and neuroendocrine tumors. 9-11 In the TAMRAD<br />

trial, 12 postmenopausal women with hormone receptor–<br />

positive, HER2-negative, aromatase inhibitor–resistant<br />

metastatic breast cancer were randomized to receive tamoxifen<br />

with everolimus or tamoxifen alone. In this randomized<br />

phase II trial, the combination of tamoxifen plus everolimus<br />

improved the 6-month clinical benefit rate (61% vs. 42%)<br />

and the time to progression (4.5 months vs. 8.6 months).<br />

The main toxicities associated with tamoxifen with everolimus<br />

compared with tamoxifen alone were fatigue (72%<br />

vs. 53%, respectively), stomatitis (56% vs. 7%, respectively),<br />

rash (44% vs. 7%, respectively), anorexia (43% vs. 18%,<br />

respectively), and diarrhea (39% vs. 11%, respectively).<br />

BOLERO-2 was a phase III randomized trial of exemestane<br />

plus everolimus versus exemestane plus placebo in<br />

women with ER- and/or PR-positive metastatic breast<br />

cancer who had progressed on prior nonsteroidal aromatase<br />

inhibitor therapy. 13 This study showed a median progression-free<br />

survival of 6.9 months with everolimus plus exemestane<br />

and 2.8 months with placebo plus exemestane<br />

(p � 0.001).<br />

Taken together, data from the TAMRAD and BOLERO-2<br />

trials provide strong evidence for the ability of everolimus to<br />

overcome resistance to endocrine therapy. Combination of<br />

everolimus and endocrine therapy is currently under investigation<br />

in the adjuvant setting. Our group is also exploring<br />

a combination of exemestane, everolimus, and metformin in<br />

obese postmenopausal women with ER- and/or PR-positive<br />

metastatic breast cancer.<br />

In summary, patients with hormone-responsive breast<br />

cancer have several treatment options. Combination of<br />

anastrozole and fulvestrant is an attractive option for patients<br />

with stage IV ER- and/or PR-positive breast cancer.<br />

For patients who develop metastatic disease while receiving<br />

adjuvant endocrine therapy or whose metastatic breast<br />

cancer progresses after single agent, front-line endocrine<br />

therapy, the combination of exemestane and everolimus is<br />

an attractive option. Novel combinations will build on this<br />

approach in the front-line metastatic setting and ultimately<br />

in the adjuvant setting.<br />

53


EDUCATIONAL SUMMARIES<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Esteva, Francisco J. Genentech;<br />

Novartis<br />

References<br />

1. Hammond ME, Hayes DF, Dowsett M, et al. <strong>American</strong> Society of<br />

Clinical Oncology/College Of <strong>American</strong> Pathologists guideline<br />

recommendations for immunohistochemical testing of estrogen<br />

and progesterone receptors in breast cancer. J Clin Oncol. 2010;<br />

28:2784-2795.<br />

2. Harvey JM, Clark GM, Osborne CK, et al. Estrogen receptor<br />

status by immunohistochemistry is superior to the ligandbinding<br />

assay for predicting response to adjuvant endocrine<br />

therapy in breast cancer. J Clin Oncol. 1999;17:1474-1481.<br />

3. Iwamoto T, Booser D, Valero V, et al. Estrogen receptor (ER)<br />

mRNA and ER-related gene expression in breast cancers that are<br />

1% to 10% ER-positive by immunohistochemistry. J Clin Oncol.<br />

<strong>2012</strong>;30:729-734.<br />

4. Chia S, Gradishar W, Mauriac L, et al. Double-blind, randomized<br />

placebo controlled trial of fulvestrant compared with exemestane<br />

after prior nonsteroidal aromatase inhibitor therapy in postmenopausal<br />

women with hormone receptor-positive, advanced<br />

breast cancer: results from EFECT. J Clin Oncol. 2008;26:1664-<br />

1670.<br />

5. Bergh J, Jonsson PE, Lidbrink EK, et al. FACT: An Open-Label<br />

Randomized Phase III Study of Fulvestrant and Anastrozole in<br />

Combination Compared With Anastrozole Alone as First-Line<br />

Therapy for Patients With Receptor-Positive Postmenopausal<br />

<strong>Breast</strong> <strong>Cancer</strong>. J Clin Oncol. <strong>2012</strong>;30:1919-1925.<br />

6. Di Leo A, Jerusalem G, Petruzelka L, et al. Results of the<br />

CONFIRM phase III trial comparing fulvestrant 250 mg with<br />

fulvestrant 500 mg in postmenopausal women with estrogen<br />

54<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

GlaxoSmithKline<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

receptor-positive advanced breast cancer. J Clin Oncol. 2010;28:<br />

4594-4600.<br />

7. Mehta RS, Barlow WE, Albain KS, et al. A phase III randomized<br />

trial of anastrozole versus anastrozole and fulvestrant as firstline<br />

therapy for postmenopausal women with metastatic breast<br />

cancer: SWOG S0226. Abstract S1–1. Presented at: 2011 CTRC-<br />

AACR San Antonio <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong>; December 2011;<br />

San Antonio, TX.<br />

8. deGraffenried LA, Friedrichs WE, Russell DH, et al. Inhibition of<br />

mTOR activity restores tamoxifen response in breast cancer cells<br />

with aberrant Akt Activity. Clin <strong>Cancer</strong> Res. 2004;10:8059-8067.<br />

9. Morrow PK, Wulf GM, Ensor J, et al. Phase I/II study of trastuzumab<br />

in combination with everolimus (RAD001) in patients<br />

with HER2-overexpressing metastatic breast cancer who progressed<br />

on trastuzumab-based therapy. J Clin Oncol. 2011;29:<br />

3126-3132.<br />

10. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic<br />

neuroendocrine tumors. N Engl J Med. 2011;364:514-523.<br />

11. Escudier B, Szczylik C, Porta C, et al. Treatment selection in<br />

metastatic renal cell carcinoma: expert consensus. Nat Rev Clin<br />

Oncol. <strong>2012</strong>;9:327-337.<br />

12. Bachelot T, Bourgier C, Cropet C, et al. Randomized Phase II<br />

Trial of Everolimus in Combination With Tamoxifen in Patients<br />

With Hormone Receptor-Positive, Human Epidermal Growth<br />

Factor Receptor 2-Negative Metastatic <strong>Breast</strong> <strong>Cancer</strong> With Prior<br />

Exposure to Aromatase Inhibitors: A GINECO Study. J Clin<br />

Oncol. <strong>2012</strong>. Epub <strong>2012</strong> May 7.<br />

13. Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal<br />

hormone-receptor-positive advanced breast cancer.<br />

N Engl J Med. <strong>2012</strong>;366:520-529.


Therapeutic Strategies Targeting ERBB2:<br />

An Update on ERBB2-Positive <strong>Breast</strong> <strong>Cancer</strong><br />

T<br />

he discovery of ERBB2 gene amplification in approximately<br />

20% of human breast cancers (BC) and its<br />

association with an adverse clinical prognosis, indicating<br />

that it may be playing a critical role in disease pathogenesis,<br />

has revolutionized the diagnosis and treatment of BC. 1-4<br />

Trastuzumab, when added to standard adjuvant chemotherapy,<br />

reduces relapse risk by approximately half and<br />

reduces mortality by about one third in ERBB2-altered<br />

early-stage BC, and to date, has withstood the test of time. 5-7<br />

Trastuzumab, and more recently lapatinib, have established<br />

benchmarks for treatment of ERBB2-positive disease.<br />

8,9 These agents have set a high efficacy bar to<br />

overcome for newer emerging ERBB2-targeted therapeutic<br />

approaches. However, within the past year, we have witnessed<br />

whole new paradigms that will challenge the chemotherapy<br />

plus trastuzumab, and lapatinib plus capecitabine,<br />

the current gold standards, as treatment for ERBB2positive<br />

disease. These new approaches include the addition<br />

of a second generation anti-ERBB2 antibody (pertuzumab)<br />

in combination with trastuzumab plus chemotherapy, and<br />

the development of an antibody-drug conjugate (ADC)<br />

based upon trastuzumab. 10-13 Both of these new approaches<br />

could prove to be practice changing, once again raising the<br />

bar for clinical efficacy (and therapeutic index) in ERBB2positive<br />

BC. Given this rapid pace of discovery research and<br />

clinical validation, the year <strong>2012</strong> will likely be remembered<br />

as a golden age of ERBB2-targeted therapy clinical research<br />

for BC.<br />

Second Generation Anti-ERBB2 Antibody<br />

Pertuzumab with Binding Epitope Distinct<br />

from Trastuzumab<br />

According to Dennis J. Slamon, MD, PhD (oral communication,<br />

September 1991), in an early screen of a panel of<br />

murine monoclonal anti-ERBB2 antibodies, two antibodies<br />

stood apart from several others in terms of demonstration of<br />

preclinical efficacy in cell proliferation assays in vitro, and in<br />

primary human tumor xenotransplants grown in the subrenal<br />

capsule of athymic mice in vivo (personal communication).<br />

Two promising murine antibodies to emerge from<br />

these screens were 4D5, which when humanized became<br />

trastuzumab, and 2C4, which following humanization is<br />

now called pertuzumab. Experimentally, it became evident<br />

that these two anti-ERBB2 antibodies were distinct, both<br />

functionally as well as structurally. 14,15 In contrast to trastuzumab,<br />

which binds to a juxtamembrane epitope in domain<br />

IV of the ERBB2 extracellular domain (ECD),<br />

pertuzumab binds to the dimerization interface contained<br />

in domain II of the ERBB2 ECD (Fig. 1). This distinction is<br />

biologically relevant in that pertuzumab disrupts the ability<br />

of ERBB2 to dimerize with any other ERBB receptor family<br />

member, thus attenuating signaling events triggered by<br />

Mark D. Pegram, MD<br />

Stanford University, Stanford, CA<br />

ERBB receptor family ligands. 16 For a time it was hoped<br />

that this could set the stage for pertuzumab treatment of<br />

ERBB2 nonamplified tumor types, if their pathogenesis<br />

proved to be driven by ligand-activated heterodimeric complexes<br />

containing ERBB2 as a coreceptor. 17 Unfortunately,<br />

with the possible exception of platinum-refractory ovarian<br />

cancer, 18 this hypothesis was not supported by early clinical<br />

trial data of single agent pertuzumab in ERBB2-negative<br />

malignancies, thus focus returned to development of pertuzumab<br />

in ERBB2-positive disease states, particularly<br />

BC. 19-21 In preclinical models, trastuzumab plus pertuzumab<br />

combination synergistically inhibited the survival of<br />

ERBB2-amplified breast cancer cells. 22 Combination drug<br />

treatment reduced levels of total and phosphorylated<br />

ERBB2 protein and blocked receptor signaling through<br />

Akt, resulting in induction of apoptosis, but did not affect<br />

mitogen-activated protein kinase. 22 These results suggested<br />

the possibility that combining ERBB2-targeting treatment<br />

agents may be a more effective therapeutic strategy than<br />

with a single ERBB2-directed antibody. Consequently, following<br />

phase II proof of concept clinical studies, pivotal<br />

randomized trials designed to explore the paradigm of dual<br />

antibody therapy for ERBB2-positive breast cancer ensued.<br />

19,23 In a clinical evaluation of pertuzumab and trastuzumab<br />

(CLEOPATRA) combination in first-line metastatic<br />

disease, 808 ERBB2-positive patients were randomly assigned<br />

to receive docetaxel plus trastuzumab and placebo<br />

versus docetaxel and trastuzumab plus pertuzumab. In an<br />

independently-assessed analysis of clinical efficacy, the median<br />

progression-free survival (PFS) in the control arm was<br />

12.4 months, compared with 18.5 months in the pertuzumab<br />

arm (hazard ratio [HR] � 0.62; p � 0.001). 24 An<br />

interim analysis of overall survival (OS) also showed a<br />

strong trend in favor of pertuzumab plus trastuzumab plus<br />

docetaxel. However, although the p value was 0.005, it did<br />

not cross the prespecified O’Brien-Fleming value of p �<br />

0.001, thus the OS result must be considered preliminary<br />

pending the protocol-planned final analysis when sufficient<br />

numbers of events have ultimately been recorded. Notably,<br />

the safety profile was generally similar in the two treatment<br />

groups, and there was no increase in left ventricular systolic<br />

dysfunction; although the rates of grade 3 or higher febrile<br />

neutropenia and diarrhea were larger in the pertuzumabtreated<br />

group. 24 These data suggested that further exploration<br />

of this approach in the early disease setting was<br />

warranted. Accordingly, a randomized phase II trial of<br />

pertuzumab/trastuzumab combination in the neoadjuvant<br />

setting has been conducted (NeoSphere). 10 In this multicenter,<br />

open-label, phase II study, treatment-naive women<br />

with ERBB2-positive BC were randomly assigned to receive<br />

four neoadjuvant cycles of either trastuzumab plus docetaxel;<br />

pertuzumab and trastuzumab plus docetaxel; pertuzumab<br />

and trastuzumab; or pertuzumab plus docetaxel.<br />

55


EDUCATIONAL SUMMARIES<br />

Fig. 1. Pertuzumab and trastuzumab bind to distinct epitopes on the ERBB2 extracellular domain. In contrast<br />

to trastuzumab, which binds to a juxtamembrane epitope in domain IV of the ERBB2 extracellular domain<br />

(ECD), pertuzumab binds to the dimerization interface contained in domain II of the ERBB2 ECD. Pertuzumab<br />

disrupts the ability of ERBB2 to dimerize with any other ERBB receptor family member, thus attenuating<br />

signaling events triggered by ERBB receptor family ligands. Reprinted from <strong>Cancer</strong> Cell with permission from<br />

Elsevier. 32<br />

Patients given pertuzumab and trastuzumab plus docetaxel<br />

had a significantly improved pathological complete response<br />

(pCR) rate (45.8%) as compared with those given<br />

trastuzumab plus docetaxel (29%; p � 0.0141), without<br />

substantial differences in tolerability. 10 It should be noted<br />

however, that the time to relapse event data have not yet<br />

been reported in this study, therefore it is not yet known<br />

whether improved pCR rates for the combination arm<br />

will translate into improved relapse-free or overall survival.<br />

Therefore, in contrast to the CLEOPATRA study in<br />

advanced disease, the NeoSphere data cannot yet be<br />

considered to be practice changing. Taken together, the<br />

findings from CLEOPATRA and NeoSphere suggest that<br />

targeting ERBB2-positive tumors with two anti-ERBB2<br />

monoclonal antibodies that have complementary mechanisms<br />

of action results in a more comprehensive blockade of<br />

ERBB2 and highlights the clinical importance of preventing<br />

the ligand-dependent formation of ERBB2-containing<br />

dimers (particularly ligand-activated heterodimers) in order<br />

to best silence ERBB2-driven downstream signaling<br />

events. 25 A study of pertuzumab plus trastuzumab adjuvant<br />

therapy in patients with newly diagnosed ERBB2-positive<br />

early breast cancer has been initiated, and is currently<br />

underway (APHINITY; NCT01358877). Pertuzumab was<br />

recently FDA-approved (June <strong>2012</strong>) for combination therapy<br />

with trastuzumab and docetaxel in the metastatic<br />

setting.<br />

56<br />

Antibody–Drug Conjugates (ADCs) based on<br />

Trastuzumab<br />

ADCs can be constructed by direct covalent linkage of a<br />

therapeutic drug (often a cytotoxic/chemotherapeutic species)<br />

directly to a monoclonal antibody backbone. By design,<br />

the antibody component of the ADC is merely being used as<br />

a delivery vehicle in order to transport a cytotoxic payload<br />

selectively to tumor cells. In ADC constructs the linker<br />

chemistry is critical to the success of the approach, since if<br />

the chemistry is too labile, the molecule will be unstable in<br />

the circulation and could lead to excessive toxicity if the<br />

cytotoxic species is liberated in normal tissue compartments.<br />

On the other hand, at least in theory, if the linker<br />

chemistry is “too stable” then the cytotoxic species may not<br />

be appropriately liberated in situ in the tumor microenvironment,<br />

thus limiting potential for antitumor efficacy. Accordingly,<br />

fine-tuning of linker chemistry is required for<br />

optimization of ADC efficacy and may be unique to each<br />

target, and to each cytotoxic moiety. We previously reported<br />

preclinical efficacy of an ADC based upon trastuzumab. 26<br />

For these experiments, a bifunctional linker was used to<br />

create a targeted prodrug that links trastuzumab to the<br />

chemotherapeutic agent paclitaxel, via an energy-reversible<br />

ester bond. This study demonstrated in vivo efficacy of a<br />

single-treatment of trastuzumab-paclitaxel ADC in decreasing<br />

tumor volume and tumor cell density of human ERBB2-


Fig. 2. Anti-ERBB2 antibody drug-conjugate trastuzumab-DM1<br />

(T-DM1). T-DM1 retains ERBB2targeting<br />

properties of trastuzumab, along with<br />

targeted delivery of the potent anti-microtubule<br />

derivative, DM1. Following binding to ERBB2 at the<br />

cell surface, T-DM1 undergoes receptor-mediated<br />

endocytosis, ultimately resulting in intracellular release<br />

of DM1 species (see text), and selective cytotoxicity<br />

to ERBB2-overexpressing tumor cells.<br />

positive BT-474 mammary tumor cells implanted in SCID<br />

mice. Moreover, the ADC was more effective in killing tumor<br />

cells than equivalent concentrations of coadministered<br />

free trastuzumab and free paclitaxel. 26 Thus the therapeutic<br />

index (efficacy divided by toxicity) could be increased<br />

markedly by an ADC approach.<br />

Another more potent trastuzumab-based ADC is<br />

trastuzumab-DM1 (T-DM1; trastuzumab emtansine),<br />

which is composed of a maytansinoid derivative, linked with<br />

a nonreducible thioether linker termed MCC (Fig. 2). Derivatization<br />

of trastuzumab in this manner occurs predominantly<br />

on the epsilon amino groups of lysines, which are<br />

abundant (n � 91) and distributed throughout the antibody’s<br />

sequence. 27 Interestingly, the binding affinity, specificity,<br />

signal perturbation capability, and effector functions<br />

of trastuzumab remain intact despite DM1 conjugation. 28<br />

Maytansinoids are natural products that are potent antimitotic<br />

agents, which like the vinca alkaloids, prevent<br />

microtubule assembly. 27 It is postulated that following<br />

receptor-mediated endocytosis, T-DM1 undergoes intralysosomal<br />

proteolytic degradation resulting in the release of<br />

Lys-MCC-DM1 and subsequent antitubulin-associated cell<br />

death. 27 In the phase I dose escalation study of T-DM1,<br />

thrombocytopenia was found to be a dose-limiting toxicity.<br />

29 In two phase II studies in patients with heavily<br />

pretreated ERBB2-positive advanced cancers who had progressed<br />

on trastuzumab and chemotherapy, or both<br />

trastuzumab- and lapatinib-based regimens in the metastatic<br />

setting, T-DM1 produced response rates between<br />

33.8% and 41%. 13,30 In one study, higher response rates and<br />

longer response durations were seen in patients with centrally<br />

confirmed ERBB2-amplified disease, once again reinforcing<br />

the importance of accurate ERBB2 testing in<br />

patients receiving ERBB2-targeted therapies. In a random-<br />

EDUCATIONAL SUMMARIES<br />

ized study in previously untreated patients with ERBB2positive<br />

breast cancer, T-DM1 also produced higher<br />

response rates and had a favorable toxicity profile compared<br />

with free trastuzumab when given in combination with free<br />

docetaxel. 31 Importantly, in terms of therapeutic index, the<br />

incidence of grade 3 or higher adverse events in the T-DM1<br />

arm was just half that observed in the trastuzumab plus<br />

docetaxel arm (37% vs. 75%). No grade 3 neutropenia was<br />

observed with T-DM1 and, remarkably, only 1.5% of patients<br />

experienced alopecia. Fortunately, in this study<br />

trastuzumab-DM1 was not associated with an increased risk<br />

of cardiotoxicity compared with trastuzumab plus docetaxel.<br />

EMILIA (NCT00829166) is a phase III, multinational,<br />

multicenter, open-label study evaluating the efficacy and<br />

safety of T-DM1 compared with capecitabine plus lapatinib<br />

(XL), an approved combination for trastuzumab-refractory<br />

ERBB2-positive metastatic BC. Pts were randomized to<br />

receive T-DM1 (3.6 mg/kg IV q3w) or X (1,000 mg/m 2 bid,<br />

days 1–14 q3w) � L (1,250 mg PO qd, days 1-21) until<br />

disease progression. Key eligibility criteria included central<br />

confirmation of ERBB2 status (IHC 3� or FISH-positive)<br />

and prior treatment with trastuzumab and a taxane. 32 Primary<br />

endpoints were PFS by independent review, OS, and<br />

safety. Baseline demographics, prior therapy, and disease<br />

characteristics were balanced between the T-DM1 arm (495<br />

patients) and XL arm (496 patients). Median durations of<br />

follow-up were 12.9 months (T-DM1) and 12.4 months (XL).<br />

Patients in the T-DM1 arm had significantly longer median<br />

PFS than those in the XL arm (9.6 vs. 6.4 mos; HR � 0.650<br />

[95% CI, 0.549-0.771]; p � 0.0001). Median OS was not<br />

reached in the T-DM1 arm compared with 23.3 mos in the<br />

XL arm (HR � 0.621 [95% CI, 0.475-0.813]; p � 0.0005);<br />

however the interim efficacy O’Brien-Fleming statistical<br />

boundary for OS was not crossed. In a planned analysis,<br />

1-year survival rates (85% [95% CI, 81%-89%] compared<br />

with 77% [72%-82%]) and 2-year survival rates (65% [59%-<br />

72%] compared with 48% [39%-56%]) were higher with<br />

T-DM1. There were fewer grade 3 or higher adverse events<br />

with T-DM1 (41% vs. 57%) and fewer dose reductions (16%<br />

with T-DM1 vs. 53% with X and 23% with L). 32 This impressive<br />

result from an ADC therapeutic approach offers a new<br />

paradigm for treatment of ERBB2-positive advanced BC,<br />

and suggests that further exploration of this ADC is warranted<br />

in (1) earlier lines of treatment for advanced disease<br />

and (2) early-stage, ERBB2-postive breast cancer. The<br />

MARIANNE study (BO22589/TDM4788g) is an ongoing<br />

randomized, phase III, placebo-controlled study in first line<br />

ERBB2-positive metastatic BC (1,092 patients) that randomly<br />

assigns patients to receive trastuzumab plus a taxane,<br />

T-DM1 plus placebo, or T-DM1 plus pertuzumab. 33<br />

Primary endpoints are PFS assessed by independent review<br />

and safety, while secondary endpoints include OS, patientreported<br />

outcomes, and an exploratory biomarker analysis.<br />

It is hoped that the result of this trial will help to inform<br />

potential regimens to be considered for future adjuvant<br />

T-DM1 campaigns.<br />

57


EDUCATIONAL SUMMARIES<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Pegram, Mark D. Dava Oncology (I) Bristol-Myers<br />

Squibb;<br />

Genentech;<br />

GlaxoSmithKline;<br />

Novartis; Pfizer/<br />

Wyeth (U);<br />

Sanofi<br />

References<br />

1. Slamon DJ, Clark GM, Wong SG, et al. Science. 1987;235:177-<br />

182.<br />

2. Slamon DJ, Godolphin W, Jones LA, et al. Science. 1989;244:707-<br />

712.<br />

3. Pegram M, Slamon D. Semin Oncol. 2000;27:13-19.<br />

4. Slamon DJ, Leyland-Jones B, Shak S, et al. N Engl J Med.<br />

2001;344:783-792.<br />

5. Romond EH, Perez EA, Bryant J, et al. N Engl J Med. 2005;353:<br />

1673-1684.<br />

6. Perez EA, Romond EH, Suman VJ, et al. J Clin Oncol. 2011;29:<br />

3366-3373.<br />

7. Slamon D, Eiermann W, Robert N, et al. N Engl J Med. 2011;<br />

365:1273-1283.<br />

8. Geyer CE, Forster J, Lindquist D, et al., N Engl J Med. 2006;355:<br />

2733-2743.<br />

9. Cameron D, Casey M, Oliva C, et al. Oncologist. 2010;15:924-934.<br />

10. Gianni L, Pienkowski T, Im YH, et al. Lancet Oncol. <strong>2012</strong>;13:25-<br />

32.<br />

11. Baselga J, Bradbury I, Eidtmann H, et al. Lancet. <strong>2012</strong>;379:633-<br />

640.<br />

12. Baselga J, Cortés J, Kim SB, et al. N Engl J Med. <strong>2012</strong>;366:109-<br />

119.<br />

13. Burris HA III, Rugo HS, Vukelja SJ, et al. J Clin Oncol. 2011;29:<br />

398-405.<br />

14. Fendly BM, Winget M, Hudziak RM, et al. <strong>Cancer</strong> Res. 1990;50:<br />

1550-1558.<br />

15. Franklin MC, Carey KD, Vajdos FF, et al. <strong>Cancer</strong> Cell. 2004;5:<br />

317-328.<br />

16. Adams CW, Allison DE, Flagella K, et al. <strong>Cancer</strong> Immunol Immunother.<br />

2006;55:717-727.<br />

17. Agus DB, Akita RW, Fox WD, et al. <strong>Cancer</strong> Cell. 2002;2:127-137.<br />

18. Makhija S, Amler LC, Glenn D, et al. J Clin Oncol. 2010;28:1215-<br />

1223.<br />

58<br />

Stock<br />

Ownership Honoraria<br />

Genentech;<br />

GlaxoSmithKline;<br />

Sanofi<br />

Research<br />

Funding<br />

Expert<br />

Testimony<br />

Sanofi Novartis<br />

Other<br />

Remuneration<br />

19. Walshe JM, Denduluri N, Berman AW, et al. Clin <strong>Breast</strong> <strong>Cancer</strong>.<br />

2006;6:535-539.<br />

20. Agus DB, Sweeney CJ, Morris MJ, et al. J Clin Oncol. 2007;25:<br />

675-681.<br />

21. Herbst RS, Davies AM, Natale RB, et al. Clin <strong>Cancer</strong> Res. 2007;<br />

13:6175-6181.<br />

22. Nahta R, Hung MC, Esteva FJ. <strong>Cancer</strong> Res. 2004;64:2343-2346.<br />

23. Baselga J, Gelmon KA, Verma S, et al. J Clin Oncol. 2010;28:<br />

1138-1144.<br />

24. Arteaga CL, Sliwkowski MX, Osborne CK, et al. Nat Rev Clin<br />

Oncol. 2011;9:16-32.<br />

25. Gilbert CW, McGowan EB, Seery GB, et al. <strong>Breast</strong> <strong>Cancer</strong> Res<br />

Treat. 2011;128:347-356.<br />

26. Lewis Phillips GD, Li G, Dugger DL, et al. <strong>Cancer</strong> Res. 2008;68:<br />

9280-9290.<br />

27. Krop IE, Beeram M, Modi S, et al. J Clin Oncol. 2010;28:2698-<br />

2704.<br />

28. Krop I, LoRusso P, Miller KD, et al. [abstract 277O]. Presented<br />

at: European Society for Medical Oncology Congress; Milan,<br />

Italy, October 8-12, 2010.<br />

29. Perez EA, Dirix L, Kocsis J, et al. [abstract LBA3]. Presented at:<br />

European Society of Medical Oncology; Milan, Italy, October<br />

8-12, 2010.<br />

30. Blackwell K, Miles D, Gianni L, et al. Primary results from<br />

EMILIA, a phase III study of trastuzumab emtansine (T-DM1)<br />

versus capecitabine (X) and lapatinib (L) in HER2-positive locally<br />

advanced or metastatic breast cancer (MBC) previously<br />

treated with trastuzumab (T) and a taxane. J Clin Oncol. <strong>2012</strong>;<br />

30 (suppl; abstr LBA1).<br />

31. LoRusso PM, Weiss D, Guardino E, et al. Clin <strong>Cancer</strong> Res.<br />

2011;17:6437-6447.<br />

32. Hubbard SR. EGF receptor inhibition: attacks on multiple<br />

fronts. <strong>Cancer</strong> Cell. 2005;7:287-288.


GENERAL SESSION XI:<br />

Debates on Current Controversies in Systemic Therapy<br />

CO-CHAIRS<br />

Gabriel N. Hortobagyi, MD<br />

University of Texas M. D. Anderson <strong>Cancer</strong> Center<br />

Minetta C. Liu, MD<br />

Georgetown University<br />

SPEAKERS<br />

Julie Gralow, MD<br />

Fred Hutchinson <strong>Cancer</strong> Research Center<br />

Clifford Hudis, MD<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center and Weill Cornell Medical College<br />

Stephen Jones, MD<br />

US Oncology Research<br />

Catherine Van Poznak, MD<br />

University of Michigan<br />

After this session, attendees should be able to<br />

● Evaluate the benefits and risks of using anthracyclines in patients with HER2-positive and HER2-negative disease and<br />

compare this therapeutic option with other forms of treatment to maximize efficacy and quality of life<br />

● Assess the use of bisphosphonates to halt bone metastasis and determine whether the use of these agents can effectively<br />

prevent disease progression in patients with early-stage breast cancer


Bone Health and the Role of Biphosphonates in Early-Stage<br />

<strong>Breast</strong> <strong>Cancer</strong>: A Debate<br />

Julie Gralow, MD, and Catherine Van Poznak, MD<br />

Fred Hutchinson <strong>Cancer</strong> Research Center, Seattle, WA, and<br />

University of Michigan, Ann Arbor, MI<br />

A<br />

djuvant systemic therapy is administered in an attempt<br />

to eradicate occult tumor cells and decrease the<br />

risk of cancer recurrence. Although the vast majority of<br />

women diagnosed with operable breast cancer will not experience<br />

recurrent disease, the risk of developing metastases<br />

remains, even after optimal standard local and systemic<br />

therapies. When breast cancer metastasizes, bone is the<br />

most commonly affected site. 1 Osteoclast inhibition has<br />

been studied as a means to interrupt the vicious cycle of cell<br />

signaling between breast cancer cells and the bone microenvironment<br />

to affect the metastatic process.<br />

Preclinical studies suggest that bisphosphonates may inhibit<br />

critical steps in the development of bone metastases in<br />

addition to inhibiting resorption. Bisphosphonates inhibit<br />

the adhesion of breast cancer cells to extracellular bone<br />

matrix, inhibit tumor cell invasion, 2-4 and induce apoptosis.<br />

5 Pretreatment of nude mice with risedronate before<br />

inoculation of tumor cells reduced the development of osteolytic<br />

lesions. 6 Clinically, there is epidemiologic evidence that<br />

bisphosphonates used in the management of postmenopausal<br />

osteoporosis may have anticancer effects. 7,8 It is<br />

important to note that use of osteoclast-inhibiting therapy<br />

to prevent osteoporotic fractures is a separate, but related,<br />

concern in managing patients with breast cancer.<br />

Adjuvant Osteoclast-Targeted Therapy in<br />

Early-Stage <strong>Breast</strong> <strong>Cancer</strong>: Clinical Evidence<br />

Studies investigating whether bisphosphonates could prevent<br />

bone metastases and improve survival in women with<br />

early-stage breast cancer have produced conflicting results.<br />

Four randomized trials of oral clodronate and one trial of<br />

oral ibandronate have been reported. Two of the four clodronate<br />

trials showed benefit for the clodronate arm. 9-11<br />

The primary endpoint of these two studies was the occurrence<br />

of bone metastases and both studies met this<br />

endpoint. 9-11 In contrast, a third study demonstrated that<br />

bone metastases were detected similarly in the clodronate<br />

and the control group. 12,13 A meta-analysis using the 5-year<br />

data from these three adjuvant clodronate trials did not<br />

show a statistically significant difference in overall survival<br />

(OS) or bone metastasis-free survival, though heterogeneity<br />

among the trials was noted. 14<br />

Two additional trials of oral bisphosphonates were reported<br />

in December 2011. The National Surgical Adjuvant<br />

<strong>Breast</strong> and Bowel Project (NSABP) B-34 trial compared 3<br />

years of daily 1,600 mg oral clodronate with placebo. The<br />

primary end point, disease-free survival (DFS), was not<br />

statistically significant between clodronate and placebo<br />

arms. 15 Subgroup analysis suggested benefits among patients<br />

age 50 or older at diagnosis. Oral ibandronate was<br />

evaluated in the German Intergroup Node-Positive (GAIN)<br />

60<br />

study in which patients receiving one of two dose-dense<br />

chemotherapy regimens were further randomly assigned to<br />

ibandronate 50 mg orally daily versus placebo. 16 In the first<br />

interim efficacy analysis, there was no difference in 3-year<br />

DFS or overall survival (OS).<br />

Studies with the more potent intravenous aminobisphosphonate,<br />

zoledronic acid, have also shown conflicting results.<br />

In the ABCSG-12 trial, premenopausal women with<br />

estrogen receptor (ER)-positive breast cancer received ovarian<br />

suppression for 3 years with goserelin. 17 Patients were<br />

randomly assigned in a two-by-two design to receive tamoxifen<br />

versus anastrozole, and zoledronic acid (4 mg, every 6<br />

months) or not. After a median follow-up of 84 months,<br />

patients receiving zoledronic acid had a statistically significant<br />

28% reduced risk of recurrence, the primary study<br />

endpoint (hazard ratio [HR]: 0.72; p � 0.014), and a 36%<br />

reduction in risk of death. 18 The recently presented post<br />

hoc, intent to treat analysis of the Zometa-Femara Adjuvant<br />

Synergy Trials (ZO-FAST), which randomly assigned postmenopausal<br />

women receiving adjuvant letrozole to either<br />

upfront or delayed zoledronic acid, showed a 34% improvement<br />

in DFS in favor of the upfront use arm (p � 0.037). 19<br />

In contrast, the Adjuvant Zoledronic Acid to Reduce Recurrence<br />

(AZURE) trial, which randomly assigned patients to<br />

standard adjuvant systemic therapy with or without an<br />

intensified regimen of zoledronic acid for 5 years, showed no<br />

benefit for adjuvant zoledronic acid. 20 At a median follow-up<br />

of 59 months, there was no significant difference in DFS or<br />

OS. In a prespecified subgroup analysis of patients who<br />

were more than 5 years from menopause prior to randomization,<br />

the 5-year OS rate was 84.6% in the zoledronic acid<br />

group and 78.7% in the control group (adjusted HR: 0.74;<br />

95% CI [0.55-0.98]; p � 0.04). There are several important<br />

differences between the study populations of these adjuvant<br />

zoledronic acid trials including hormone receptor status,<br />

menopausal status, and systemic therapies administered. A<br />

number of hypotheses to account for the different study<br />

outcomes can be generated by assessing the differences in<br />

study populations.<br />

Additional recently closed and ongoing trials will provide<br />

further data on the utility of bisphosphonates in the adjuvant<br />

breast cancer setting. The Southwest Oncology Group<br />

(SWOG) S0307 trial randomly assigned 6,000 women with<br />

stage I-III breast cancer receiving standard adjuvant therapy<br />

to oral clodronate versus oral ibandronate versus zoledronic<br />

acid, all for 3 years duration.<br />

Additional strategies for osteoclast inhibition are also in<br />

evaluation. The Denosumab as Adjuvant Treatment for<br />

Women with High Risk Early <strong>Breast</strong> <strong>Cancer</strong> Receiving Neoadjuvant<br />

or Adjuvant Therapy (D-CARE) trial is ongoing<br />

and looks at the adjuvant effects of RANK ligand-targeted<br />

therapy. ABCSG-18 will evaluate the effect of denosumab


on fracture risk in patients with breast cancer who receive<br />

AI therapy, with DFS and OS as secondary endpoints. The<br />

S0307 and D-CARE trials include a broad and high-risk<br />

patient population of both pre- and postmenopausal women<br />

and all tumor subsets, including ER-positive and -negative<br />

tumors. The results of these trials will be critical in better<br />

defining the efficacy of bone-modifying agents for preventing<br />

recurrence and may provide additional data regarding<br />

which patients and tumors are most likely to benefit from<br />

treatment.<br />

Authors’ Disclosures of Potential Conflicts of Interest<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Conclusion<br />

The results of the studies to date have not provided sufficient<br />

evidence to universally incorporate osteoclast inhibition<br />

into adjuvant care, although there may be subsets of<br />

patients for whom a bisphosphonate offers a potential anticancer<br />

affect. The ongoing adjuvant phase III studies investigating<br />

bisphosphonates and denosumab are likely to<br />

provide important information, and it is anticipated that<br />

correlative studies will aid in defining those most likely to<br />

gain benefit from adjuvant osteoclast inhibition.<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Gralow, Julie Amgen;<br />

Genentech;<br />

Novartis; Roche;<br />

Sanofi<br />

Van Poznak, Catherine Amgen, Novartis<br />

References<br />

1. Coleman RE, Rubens RD. The clinical course of bone metastases<br />

from breast cancer. Brit J <strong>Cancer</strong>. 1987;55:61-66.<br />

2. van der Pluijm G, Vloedgraven H, van Beek E, et al. Bisphosphonates<br />

inhibit the adhesion of breast cancer cells to bone matrices<br />

in vitro. J Clin Invest. 1996;98:698-705.<br />

3. Boissier S, Ferreras M, Peyruchaud O, et al. Bisphosphonates<br />

inhibit breast and prostate carcinoma cell invasion, an early<br />

event in the formation of bone metastases. <strong>Cancer</strong> Res.2000;60:<br />

2949-2954.<br />

4. Boissier S, Magnetto S, Frappart L, et al. Bisphosphonates inhibit<br />

prostate and breast carcinoma cell adhesion to unmineralized<br />

and mineralized bone extracellular matrices. <strong>Cancer</strong> Res.<br />

1997;57:3890-3894.<br />

5. Senaratne SG, Mansi JL, Colston KW. The bisphosphonate zoledronic<br />

acid impairs Ras membrane localisation and induces<br />

cytochrome c release in breast cancer cells. Br J <strong>Cancer</strong>. 1002;<br />

86:1479-1486.<br />

6. Sasaki A, Boyce BF, Story B, et al. Bisphosphonate risedronate<br />

reduces metastatic human breast cancer burden in bone in nude<br />

mice. <strong>Cancer</strong> Res. 1995;55:3551-3557.<br />

7. Chlebowski RT, Chen Z, Cauley JA, et al. Oral bisphosphonate<br />

use and breast cancer incidence in postmenopausal women.<br />

J Clin Oncol. 2010;28:3582-3590.<br />

8. Rennert G, Pinchev M, Rennert HS. Use of bisphosphonates<br />

and risk of postmenopausal breast cancer. J Clin Oncol. 2010;28:<br />

3577-3581.<br />

9. Powles T, Paterson A, McCloskey E, et al. Reduction in bone<br />

relapse and improved survival with oral clodronate for adjuvant<br />

treatment of operable breast cancer. <strong>Breast</strong> <strong>Cancer</strong> Res. 2006;8:<br />

R13.<br />

10. Diel IJ, Solomayer EF, Costa SD, et al. Reduction in new metastases<br />

in breast cancer with adjuvant clodronate treatment.<br />

N Engl J Med. 1998;339:357-363.<br />

11. Diel IJ, Jaschke A, Solomayer EF, et al. Adjuvant oral clodronate<br />

improves the overall survival of primary breast cancer patients<br />

with micrometastases to the bone marrow: a long-term followup.<br />

Ann Oncol. 2008;19:2007-2011.<br />

12. Saarto T, Blomqvist C, Virkkunen P, et al. Adjuvant clodronate<br />

EDUCATIONAL SUMMARIES<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

treatment does not reduce the frequency of skeletal metastases<br />

in node-positive breast cancer patients: 5-year results of a randomized<br />

controlled trial. J Clin Oncol. 2001;19:10-17.<br />

13. Saarto T, Vehmanen L, Virkkunen P, et al. Ten-year follow-up of<br />

a randomized controlled trial of adjuvant clodronate treatment<br />

in node-positive breast cancer patients. Acta Oncol. 2004;43:650-<br />

656.<br />

14. Ha TC, Li H. Meta-analysis of clodronate and breast cancer<br />

survival. Br J <strong>Cancer</strong>. 2007;96:1797-1801.<br />

15. Paterson AHG, Anderson SJ, Lembersky BC, et al. NSABP<br />

Protocol B-34: A Clinical Trial Comparing Adjuvant Clodronate<br />

vs. Placebo In Early Stage <strong>Breast</strong> <strong>Cancer</strong> Patients Receiving<br />

Systemic Chemotherapy and/or Tamoxifen or No Therapy–Final<br />

Analysis. Paper presented at: San Antonio <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong>;<br />

December 2011; San Antonio, TX.<br />

16. Möbus V DI, Elling D, Harbeck N, et al. GAIN study: a phase III<br />

trial to compare ETC vs. EC-TX and ibandronate vs observation<br />

in patients with node positive primary breast cancer-1st interim<br />

efficacy analysis. Paper presented at: San Antonio <strong>Breast</strong> <strong>Cancer</strong><br />

<strong>Symposium</strong>; December 2011; San Antonio, TX.<br />

17. Gnant M, Mlineritsch B, Schippinger W, et al. Endocrine therapy<br />

plus zoledronic acid in premenopausal breast cancer. N Engl<br />

J Med. 2009;360:679-691.<br />

18. Gnant M, Luschin-Ebengreuth G, Stoeger H, et al. Long-term<br />

follow-up in ABCSG-12: significantly improved overall survival<br />

with adjuvant zoledronic acid in premenopausal patients with<br />

endocrine-receptor–positive early breast cancer. Paper presented<br />

at: San Antonio <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong>; December<br />

2011; San Antonio, TX.<br />

19. de Boer RH, Eidtmann H, Neven P, et al. Long-term survival<br />

outcomes among postmenopausal women with hormone<br />

receptor-positive early breast cancer receiving adjuvant letrozole<br />

and zoledronic acid: 5-year follow-up of ZO-FAST. Paper presented<br />

at: San Antonio <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong>; December<br />

2011; San Antonio, TX.<br />

20. Coleman R, Cameron D, Dodwell D, et al. <strong>Breast</strong>-cancer adjuvant<br />

therapy with Zoledronic Acid. N Engl J Med. 2011;365:1396-<br />

1405.<br />

61


In Favor of the Use of Anthracyclines in Patients with<br />

HER2-Positive and HER2-Negative <strong>Breast</strong> <strong>Cancer</strong><br />

Clifford Hudis, MD<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center and<br />

Weill Cornell Medical College, New York, NY<br />

A<br />

nthracycline antibiotics are among the most effective<br />

cytotoxic drugs for a wide range of malignancies. In<br />

the 1950s, the Italian-based Farmitalia Research Laboratories<br />

explored soil-based microbes and found the redpigmented<br />

Streptomyces peucetius producing a cytotoxic<br />

antibiotic called daunorubicin at a similar time as a French<br />

group. 1 The Italians generated a mutated bacterium producing<br />

a red antibiotic named Adriamycin (doxorubicin or<br />

more precisely, hydroxydaunorubicin) for the Adriatic Sea.<br />

These agents (daunorubicin and doxorubicin) served as prototypes<br />

and there are now thousands of analogs, including<br />

idarubicin, epirubicin, as well as novel intravenous delivery<br />

systems such as liposomes. 2,3<br />

The anthracyclines interact with DNA by intercalation<br />

and inhibit the biosynthesis of macromolecules and specifically<br />

the relaxation of DNA supercoils by topoisomerase II,<br />

a step needed for effective transcription. By stabilizing<br />

topoisomerase II following interruption of the DNA, these<br />

agents prevent replication. 4<br />

In the last decades of the 20th century, investigators built<br />

on the demonstrations of activity in breast cancer using<br />

cyclophosphamide, methotrexate, and 5-fluorouracil (CMF)<br />

by adding doxorubicin and later epirubicin as components<br />

of palliative and curative systemic treatment. The key challenges<br />

with these drugs include their risks of congestive<br />

heart failure, secondary leukemia, and a host of short-term<br />

and generally reversible toxicities such as neutropenia,<br />

mucositis, nausea, and vomiting. Importantly, the worldwide<br />

overview (meta-analysis) of adjuvant chemotherapy<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Hudis, Clifford*<br />

*No relevant relationships to disclose.<br />

Employment or<br />

Leadership<br />

Positions<br />

References<br />

Consultant or<br />

Advisory Role<br />

1. Arcamone F, Cassinelli G, Fantini G, et al. Adriamycin, 14hydroxydaunomycin,<br />

a new antitumor antibiotic from S. peucetius<br />

var. caesius. Biotechnol Bioeng. 1969;11:1101-1110.<br />

2. Bonfante V, Bonadonna G, Villani, F, et al. Preliminary clinical<br />

experience with 4-epidoxorubicin in advanced human neoplasia.<br />

Recent Results in <strong>Cancer</strong> Research. 1980;74:192–199.<br />

3. Symon Z, Peyser A, Tzemach D, et al. Selective delivery of<br />

doxorubicin to patients with breast carcinoma metastases by<br />

Stealth liposomes. <strong>Cancer</strong>. 1999;86:72-78.<br />

4. Fornari FA, Randolph JK, Yalowich JC, et al. Interference by<br />

doxorubicin with DNA unwinding in MCF-7 breast tumor cells.<br />

Mol Pharmacol. 1994;45:649-656.<br />

5. Early <strong>Breast</strong> <strong>Cancer</strong> Trialists’ Collaborative Group. Comparisons<br />

between different polychemotherapy regimens for early<br />

62<br />

regimens performed approximately every 5 years in Oxford,<br />

England, provides evidence of superiority for anthracyclinecontaining<br />

adjuvant regimens in 18,000 randomly assigned<br />

patients (with node-negative, and node-positive disease, all<br />

hormone receptor profiles, and of all ages and menopausal<br />

status). 5 Here, there was no evidence that four doses of<br />

standard doxorubicin (60 mg/m 2 ) and cyclophosphamide<br />

(600 mg/m 2 ) (AC) were superior to CMF regimens. However,<br />

regimens with greater cumulative doses, such as<br />

cyclophosphamide, doxorubicin, and 5-fluorouracil or cyclophosphamide,<br />

epirubicin, and 5-fluorouracil, showed reductions<br />

in breast cancer and all-cause mortality of 22% and<br />

36%, respectively. Not explored fully in the study were<br />

optimal regimens, such as dose-dense AC-paclitaxel and<br />

their comparative toxicities. 6,7 Despite this, there are biological<br />

factors (e.g., topoisomerase II expression), clinical<br />

factors (age, use of additional targeted therapies, prior<br />

treatments), and specific phase III trials (such as docetaxel/<br />

cyclophamide or TC compared to AC) that have raised questions<br />

about the ongoing role of the anthracyclines. 8-13<br />

The challenge for medical oncologists in <strong>2012</strong> is to identify<br />

situations where the anthracyclines remain critical and<br />

those where they might be avoided with no compromise in<br />

overall outcomes. Continued discussion and research is<br />

needed to better identify the most appropriate and optimal<br />

role for these highly effective agents but the superior overall<br />

outcomes in nearly all randomized studies testing these<br />

agents makes it clear that they cannot simply be abandoned.<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

breast cancer: meta-analysis of long-term outcome among<br />

100,000 women in 123 randomised trials. Lancet. <strong>2012</strong>;379:432–<br />

444.<br />

6. Citron ML, Berry DA, Cirincione C, et al. Randomized trial of<br />

dose-dense versus conventionally scheduled and sequential versus<br />

concurrent combination chemotherapy as postoperative adjuvant<br />

treatment of node-positive primary breast cancer: first<br />

report of Intergroup Trial C9741/<strong>Cancer</strong> and Leukemia Group B<br />

Trial 9741. J Clin Oncol. 2003;21:431-439.<br />

7. Morris P, Dickler M, McArthur H, et al. Dose-dense adjuvant<br />

doxorubicin and cyclophosphamide is not associated with frequent<br />

short-term changes in left ventricular ejection fraction.<br />

J Clin Oncol. 2009;27:6117-6123.<br />

8. Perez EA, Romond EH, Suman VJ, et al. Four-year follow-up of<br />

trastuzumab plus adjuvant chemotherapy for operable human<br />

epidermal growth factor receptor 2-positive breast cancer: joint


analysis of data from NCCTG N9831 and NSABP B-31. J Clin<br />

Oncol. 2011;29:3366-3373.<br />

9. Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant<br />

chemotherapy for operable HER2-positive breast cancer.<br />

N Engl J Med. 2005;353:1673-1684.<br />

10. Slamon D, Eiermann W, Robert N, et al. Adjuvant trastuzumab<br />

in HER2-positive breast cancer. N Engl J Med. 2011;365:1273-<br />

1283.<br />

11. Press M, Sauter G, Boyce M, et al. Alteration of topoisomerase<br />

II-alpha gene in human breast cancer: association with respon-<br />

EDUCATIONAL SUMMARIES<br />

siveness to anthracycline-based chemotherapy. J Clin Oncol.<br />

2011;29:859-867.<br />

12. Jones SE, Holmes FA, O’Shaughnessy JA, et al. Docetaxel with<br />

cyclophosphamide is associated with an overall survival benefit<br />

compared to doxorubicin and cyclophosphamide: 7-year follow-up<br />

of US Oncology Research Trial 9735. J Clin Oncol. 2009;27:1177-<br />

1183.<br />

13. Gianni L, Norton L, Wolmark N, et al. Role of anthracyclines in<br />

the treatment of early breast cancer. J Clin Oncol. 2009;27:4798-<br />

4808.<br />

63


Marker-Driven Personalized Therapy for <strong>Breast</strong> <strong>Cancer</strong><br />

Gabriel N. Hortobagyi, MD<br />

University of Texas M. D. Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Optimal treatment for primary breast cancer is multidisciplinary<br />

and adapted to the clinical and molecular<br />

characteristics of the tumor. 1 Wide surgical excision<br />

remains a cornerstone of curative treatment and postoperative<br />

radiotherapy is known to improve survival. 2 Increasingly,<br />

the molecular subtype of breast cancer is known to<br />

influence local-regional control, and because of this there is<br />

a large interest in determining optimal local-regional therapeutic<br />

interventions. Systemic treatments reduce annual<br />

odds of recurrence by 50% to 65% and annual odds of death<br />

by 40% to 50%. 3<br />

<strong>Breast</strong> cancer includes several molecularly defined subgroups,<br />

defined by gene expression or protein profiling. 4,5 At<br />

the clinical level, determination of hormone-receptor (HR)<br />

expression, human epidermal growth factor receptor 2<br />

(HER2), grade, and proliferation markers is considered a<br />

practical method to identify clinically relevant subsets of<br />

patients for whom distinct therapeutic decisions are made. 1<br />

Patients with HR-positive, HER2-negative tumors with low<br />

proliferation rate (or low grade) benefit from adjuvant endocrine<br />

therapy; chemotherapy provides limited or no benefit<br />

for this group but does result in incremental benefit for<br />

patients with high-grade or highly proliferative tumors.<br />

Patients with HER2-positive tumors benefit from trastuzumab<br />

combined with chemotherapy, although the magnitude<br />

of this benefit is modulated by hormone-receptor<br />

expression. Patients with triple-negative breast cancer (absent<br />

HR and normal HER2 expression) benefit from chemotherapy<br />

and probably from antivascular endothelial growth<br />

factor therapy such as bevacizumab. However, no specifically<br />

targeted therapeutics exist for this subtype. 6<br />

Adjuvant! Online is widely utilized to estimate risk of<br />

recurrence and death and Oncotype DX and Mammaprint<br />

are increasingly used to define the early breast cancer populations<br />

that benefit from adjuvant systemic therapy. 1<br />

These assays have been validated retrospectively in multiple<br />

datasets, some of them from prospective clinical trials.<br />

From these early validations, only the OncotypeDX seems<br />

to have the ability to predict benefit from specific treatment<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

Hortobagyi, Gabriel N. Allergan;<br />

Genentech;<br />

Novartis (U);<br />

Sanofi<br />

References<br />

1. Hortobagyi GN. Towards individualized breast cancer therapy:<br />

translating biological concepts to the bedside. Oncologist. <strong>2012</strong>;<br />

17:577-584.<br />

2. Early <strong>Breast</strong> <strong>Cancer</strong> Trialists’ Collaborative Group (EBCTCG),<br />

Darby S, McGale P, et al. Effect of radiotherapy after breast-<br />

64<br />

modalities. Both the OncotypeDx and the Mammaprint are<br />

undergoing definitive evaluation in large, multicenter clinical<br />

trials (TAILORx, RxPONDER, and MINDACT). 7,8<br />

In general, these first-generation genomic assays work<br />

very well for groups of patients but less precisely for individuals.<br />

With technological advances, these approaches are<br />

being overtaken by full sequencing strategies in an attempt<br />

to individualize treatment strategies by finding specific<br />

drivers within tumors. Several companies have developed<br />

commercial products that are currently being assessed in<br />

clinical trials (e.g., Foundation Medicine). Genomic assays<br />

are being studied to determine whether specific therapeutic<br />

interventions would be effective in reducing risk of tumor<br />

cell dissemination and death. Preliminary evidence suggests<br />

that some assays might predict benefit from chemotherapy<br />

whereas others might predict benefit from<br />

endocrine therapy within estrogen receptor–positive subtypes.<br />

9<br />

Widely used biomarkers (HR and HER2) have limited<br />

utility in selecting optimal therapy. The absence of HR and<br />

HER2 is a sensitive indicator of resistance to endocrine and<br />

HER2-directed therapies, respectively, but these assays<br />

have limited ability to predict response to those same treatments.<br />

There is interest in understanding mechanisms of<br />

resistance to commonly used drugs; thus, coactivation of<br />

membrane-based growth factor receptor systems (EGFR,<br />

HER2, insulin-like growth factor receptor, etc.) has been<br />

linked to resistance to endocrine agents, and dysregulation<br />

of the PI3K/AKT/mTOR-signaling pathways is associated<br />

with resistance to endocrine and trastuzumab therapy.<br />

Combinations of targeted agents are currently under assessment.<br />

10,11 There is ample evidence that dual inhibition<br />

of commonly dysregulated cell-signaling pathways is more<br />

successful than the use of monotherapy in the metastatic<br />

setting, and this concept is currently under evaluation in<br />

the adjuvant setting. As our understanding of driving molecular<br />

anomalies expands, we will get closer to individualized<br />

cancer therapy.<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

Novartis<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

conserving surgery on 10-year recurrence and 15-year breast<br />

cancer death: meta-analysis of individual patient data on 10,801<br />

women in 17 randomised trials. Lancet. 2011;378:1707-1716.<br />

3. Early <strong>Breast</strong> <strong>Cancer</strong> Trialists’ Collaborative Group (EBCTCG).<br />

Effects of chemotherapy and hormonal therapy for early breast<br />

cancer on recurrence and 15-year survival: an overview of the<br />

randomised trials. Lancet. 2005;365:1687-1717.


4. Sørlie T, Perou CM, Tibshirani R, et al. Gene expression patterns<br />

of breast carcinomas distinguish tumor subclasses with clinical<br />

implications. Proc Natl Acad Sci USA.2001;98:10869-10874.<br />

5. Gonzalez-Angulo AM, Hennessy BT, Meric-Bernstam F, et al.<br />

Functional proteomics can define prognosis and predict pathologic<br />

complete response in patients with breast cancer. Clin<br />

Proteomics. 2011;8:11.<br />

6. Gelmon K, Dent R, Mackey JR, et al. Targeting triple-negative<br />

breast cancer: optimising therapeutic outcomes. Ann Oncol.<br />

Epub <strong>2012</strong> Apr 19.<br />

7. Sparano JA. TAILORx: trial assigning individualized options for<br />

treatment (Rx). Clin <strong>Breast</strong> <strong>Cancer</strong>. 2006;7:347-350.<br />

8. Cardoso F, Van’t Veer L, Rutgers E, et al. Clinical application of<br />

EDUCATIONAL SUMMARIES<br />

the 70-gene profile: the MINDACT trial. J Clin Oncol. 2008;26:<br />

729-735.<br />

9. Tang G, Shak S, Paik S, et al. Comparison of the prognostic and<br />

predictive utilities of the 21-gene recurrence score assay and<br />

Adjuvant! for women with node-negative, ER-positive breast<br />

cancer: results from NSABP B-14 and NSABP B-20. <strong>Breast</strong><br />

<strong>Cancer</strong> Res Treat. 2011;127:133-142.<br />

10. Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal<br />

hormone-receptor-positive advanced breast cancer.<br />

N Engl J Med. <strong>2012</strong>;366:520-529.<br />

11. Baselga J, Cortés J, Kim SB, et al. Pertuzumab plus trastuzumab<br />

plus docetaxel for metastatic breast cancer. N Engl J Med.<br />

<strong>2012</strong>;366:109-119.<br />

65


“Just Say No” to Anthracyclines for Early <strong>Breast</strong> <strong>Cancer</strong><br />

T<br />

he use of anthracyclines for adjuvant therapy of early<br />

breast cancer is associated with the potential for serious<br />

long-term consequences for some women treated with<br />

curative intent. These include late bone-marrow effects<br />

(e.g., leukemia or myelodysplastic syndromes) and myocardial<br />

damage (e.g., congestive heart failure [CHF]). Anthracyclines<br />

have been a mainstay of adjuvant regimens since<br />

early work with the regimen in the early 1970s, 1 including<br />

more modern regimens like anthracycline taxane, dosedense<br />

anthracycline/paclitaxel; docetaxel, doxorubicin hydrochloride,<br />

and cyclophosphamide (TAC); and fluorouracil,<br />

epirubicin, and cyclophosphamide (FEC)/docetaxel. But<br />

these commonly used regimens still have the potential to<br />

cause late toxicities of bone-marrow and heart damage. This<br />

is well illustrated by long-term follow-up of the BCIRG 001<br />

study comparing TAC with doxorubicin in combination<br />

with fluorouracil and cyclophosphamide (FAC). 2 With 10<br />

years of follow-up, TAC remained significantly superior to<br />

FAC in terms of disease-free survival (DFS) and overall<br />

survival (OS). However, rates of CHF continued to increase<br />

over the 10 years of follow-up with both of these anthracyline<br />

regimens with rates of CHF of 3.5% with TAC and<br />

2.3% with FAC. 2 Beyond the diagnosis of CHF, 15 to 17% of<br />

patients exhibited decreases of cardiac ejection fractions of<br />

over 20%. In addition, there were nine cases of hematologic<br />

malignancies often associated with anthracycline usage.<br />

Similar data have been observed in other trials using<br />

anthracycline if long-term follow-up was performed (it is<br />

not always performed). We would all accept the risks to our<br />

patients from use of anthracycline regimens if there were<br />

no alternative nonanthracycline treatment options. Fortunately,<br />

that is not the case.<br />

Pivotal Nonanthracycline Trials<br />

There are two studies that strongly support the use of<br />

anthracycline-free treatment for early breast cancer. The<br />

first is the study we conducted at US Oncology Research<br />

comparing classic anthracycline chemotherapy to a nonanthracycline<br />

regimen of docetaxel/cyclophosphamide (TC). 3<br />

In this study, women with early breast cancer were randomized<br />

to four cycles of chemotherapy; at 7 years of follow-up,<br />

we reported superior overall survival (OS) (87% TC vs. 82%<br />

anthracycline, hazard ration [HR]: 0.69) as well as diseasefree<br />

survival (DFS) (81% TC vs. 75% anthracycline, HR:<br />

0.74) with fewer instances of CHF or bone marrow damage<br />

in the TC arm. These results were seen in HER2-positive<br />

disease as well as in HER2-negative disease, as the study<br />

was conducted at a time when adjuvant trastuzumab was<br />

not routinely utilized. Based on this study, one must consider<br />

the use of TC for node-negative or node-positive early<br />

breast cancer, particularly those with more limited nodal<br />

involvement and HER2-negative disease. In fact, since the<br />

publication of this study, the use of the TC regimen in the<br />

66<br />

Stephen Jones, MD<br />

US Oncology Research, The Woodlands, TX<br />

United States has increased dramatically, thereby sparing<br />

many women the exposure to anthracyclines.<br />

The second pivotal trial is the recently published BCIRG<br />

006, 4 which deals with the addition of trastuzumab to either<br />

an anthracycline-based regimen (AC-TH) or a nonanthracycline<br />

regimen of docetaxel/carboplatin (TCH) for women<br />

with HER2-overexpressed early breast cancer. This study<br />

compared DFS at 5 years by regimen and found rates of 75%<br />

for ACT and rates of 84% and 81% for AC-TH and TCH,<br />

respectively. Both trastuzumab regimens were significantly<br />

superior to the arm without trastuzumab (ACT) and not<br />

different between the two trastuzumab-containing arms.<br />

Those who favor use of anthracyclines for HER2-positive<br />

disease cite the 84% 5-year rate of DFS with AC-TH as<br />

“numerically different” than 81% for TCH. In fact, this<br />

is not a valid medical or statistical conclusion and is only<br />

an argument used by those who want to promote anthracyclines<br />

for early breast cancer. In addition, there was significantly<br />

more cases of acute leukemia and cardiac<br />

dysfunction in the anthracycline arms compared with the<br />

TCH arm. For example, there were eight cases of leukemia<br />

with seven in the anthracycline arms and one in the TCH<br />

arm. Based on this randomized prospective trial for women<br />

with early HER2-positive disease, the TCH regimen has<br />

also been widely adopted, sparing many women the added<br />

risk of anthracyclines.<br />

Both of these trials provide convincing arguments for<br />

treating women with anthracycline-free regimens, but<br />

there is additional supporting science to be reviewed.<br />

Supporting Science<br />

The reputed primary target for anthracyclines is topoisomerase<br />

II alpha (TOP2A). Although this may not be the only<br />

target, it is the predominant mechanism of action. Over the<br />

years, there has been increasing evidence of the role of<br />

TOP2A in predicting sensitivity to anthracyclines, but probably<br />

the most impressive body of work was reported by<br />

Press et al. 5 In that study, the laboratory analyzed alterations<br />

in nearly 5,000 breast cancers from trials of chemotherapy<br />

with or without trastuzumab using data from a<br />

metastatic trial as the test set and data from BCIRG 005<br />

and 006 as the validation set. In the BCIRG 005 trial (a<br />

study of HER2-negative breast cancer where the HER2<br />

testing was also done by Press and colleagues), there were<br />

no cases of TOP2A amplification in 1,614 breast cancers. 6 In<br />

the BCIRG 006 study of HER2-overexpressed breast cancer<br />

summarized above, Press and colleagues found in 2,990<br />

cases that overamplification of TOP2A occurred in 35%<br />

whereas TOP2A-normal was seen in 60% of cases and<br />

TOP2A-deletions was seen in 5% of cases. In this trial, since<br />

there were 2 anthracycline arms (AC-T and AC-TH) as well<br />

as a nonanthracycline arm (TCH), the investigators could<br />

compare outcomes related to alterations in a gene copy of


TOP2A. In the AC-T arm, there was a clear-cut benefit with<br />

anthracycline use without trastuzumab only in the group of<br />

women where TOP2A overexpression existed. And in the<br />

presence of trastuzumab, the use of anthracyclines did not<br />

make a difference regardless of TOP2A gene copy or anthracycline<br />

use. This is the largest study from a single laboratory<br />

with excellent quality controls to examine this issue. If one<br />

looks at all invasive breast cancer based on this data—since<br />

almost all of anthracycline sensitivity relates to TOP2A<br />

overexpression and such overexpression occurs almost<br />

entirely in the HER2-positive population—less than 10% of<br />

breast cancer will actually benefit from the use of anthracyclines.<br />

That 10% resides almost entirely in the HER2positive<br />

population, which is now being treated with<br />

trastuzumab-based regimens. This strongly argues that<br />

few, if any, women will benefit from anthracycline regimens.<br />

Unanswered Questions<br />

In the original AC (doxorubicin/cyclophosphamide) versus<br />

TC trial, we used four courses of therapy. Recent data from<br />

the BCIRG 005 trial 6 and the NSABP trial 7 (which did not<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Employment or<br />

Leadership Consultant or Stock<br />

Author<br />

Positions Advisory Role Ownership Honoraria<br />

Jones, Stephen Genentech Genentech<br />

References<br />

1. Jones SE, Durie BG, Salmon SE. Combination chemotherapy with<br />

adriamycin and cyclophosphamide for advanced breast cancer.<br />

<strong>Cancer</strong>. 1975;36:90-97.<br />

2. Martin M, Mackey J, Pienkowski T, et al. Ten-year follow-up<br />

analysis of the BCIRG 001, trial confirms superior DFS and OS<br />

benefit of adjuvant TAC (docetaxel, doxorubicin, cyclophosphamide)<br />

over FAC (fluorouracil, doxorubicin, cyclophosphamide) in<br />

women with operable node-positive breast cancer. <strong>Cancer</strong> Research.<br />

Presented at: 33 rd Annual San Antonio <strong>Breast</strong> <strong>Cancer</strong><br />

<strong>Symposium</strong>, San Antonio, TX, December 2010; abstr S4–3.<br />

3. Jones S, Holmes FA, O’Shaughnessy J, et al. Docetaxel with<br />

cyclophosphamide is associated with an overall survival benefit<br />

compared with doxorubicin and cyclophosphamide: 7-Year<br />

follow-up of US Oncology Research Trial 9735. J Clin Oncol.<br />

2009;27:1177-1183. Epub 2009 Feb 9.<br />

study TC) suggested that four courses of therapy might not<br />

be enough for women with node-positive disease. For this<br />

reason, the ideal number of cycles of TC chemotherapy has<br />

still not been determined, but in the prospective studies 6 is<br />

being used. The other issue for those who still support the<br />

use of anthracyclines would be data from a randomized trial<br />

of TC versus an anthracycline regimen. In US Oncology<br />

06-090, six cycles of TC versus six cycles of TAC were<br />

investiaged. This trial has been modified twice to include<br />

bevacizumab (B46 and USON 06-090) and more recently to<br />

take out the bevacizumab arm (NSABP B49). Ultimately,<br />

about 4,000 women will be randomly assigned to a nonanthracycline<br />

regimen (TC) or an anthracycline-based regimen<br />

(TAC or a version of dose-dense ACT). Because of the<br />

data described above and the putative target of TOP2A, I<br />

believe that the TC regimen will be equal to anthracycline<br />

regimens for most women with HER-negative disease.<br />

However, whether or not there is a subset of patients who<br />

benefit from anthracyclines (e.g., triple-negative disease)<br />

based on some mechanism of action other than TOP2A gene<br />

expression still remains to be answered, and this trial will<br />

provide definite answers to these questions.<br />

Research<br />

Funding<br />

EDUCATIONAL SUMMARIES<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

4. Slamon D, Eiermann W, Robert N, et al. Adjuvant trastuzumab<br />

in HER2-positive breast cancer. N Engl J Med. 2011;365:1273-<br />

1283.<br />

5. Press MF, Sauter G, Buyse M, et al. Alteration of topoisomerase<br />

II-alpha gene in human breast cancer: association with responsiveness<br />

to anthracycline-based chemotherapy. J Clin Oncol.<br />

2011;29:859-867. Epub 2010 Dec 28.<br />

6. Eiermann W, Pienkowski T, Crown J, et al. Phase III study of<br />

doxorubicin/cyclophosphamide with concomitant versus sequential<br />

docetaxel as adjuvant treatment in patients with human<br />

epidermal growth factor receptor 2-normal, node-positive breast<br />

cancer: BCIRG-005 trial. J Clin Oncol. 2011;29:3877-3884. Epub<br />

2011 Sep 12.<br />

7. Swain SM, Jeong JH, Geyer CE Jr., et al. Longer therapy, iatrogenic<br />

amenorrhea, and survival in early breast cancer. N Engl<br />

J Med. 2010;362:2053-2065.<br />

67


Reassessing Local Treatment in the Context of Increasingly<br />

Effective Systemic Therapy<br />

Jay R. Harris, MD<br />

Dana-Farber <strong>Cancer</strong> Institute and Brigham and Women’s Hospital,<br />

Harvard Medical School, Boston, MA<br />

T<br />

he current status of local treatment is that (1) improved<br />

local treatment is now clearly linked to longterm<br />

survival, (2) radiation therapy (RT) is generally used<br />

after breast-conserving surgery (BCS) and selectively used<br />

after mastectomy, and (3) RT for breast cancer has become<br />

technically advanced and safer. However, the main focus in<br />

research is clearly on further improvements in the effectiveness<br />

of systemic therapy. Given this, understanding the role<br />

of local treatment in the context of systemic therapy is<br />

important.<br />

In 2007, we hypothesized that the benefit of local therapy<br />

on survival would have a parabolic relationship to increasing<br />

effectiveness of systemic therapy (Fig. 1). 1 In the absence<br />

of effective (adjuvant) systemic therapy, local therapy<br />

will have no effect on survival in the many patients with<br />

micrometastatic disease. Initially, as systemic therapy becomes<br />

able to eradicate micrometastatic disease, local therapy<br />

will have a bigger impact on survival. Ultimately, as we<br />

hope, systemic therapy will become so effective that local<br />

therapy will not be required.<br />

The latest report from the Early <strong>Breast</strong> <strong>Cancer</strong> Clinical<br />

Trialists’ Collaborative Group (EBCTCG) (also<br />

known as the Oxford Overview) on the impact of adding<br />

RT to BCS was published in 2011. 2 Seven trials of<br />

lumpectomy in low-risk patients were added, and the<br />

prior 10 trials were updated for a total of 10,801 women<br />

with a median woman-year at risk of 9.5. Of note, the<br />

EBCTCG moved from assessing the effect of RT on LR<br />

Fig. 1. Hypothetical relationship between increasing<br />

effectiveness of systemic therapy and the benefit<br />

of local therapy on survival.<br />

68<br />

to its effect on first failure (either local recurrence [LR]<br />

or distant metastases [DM]). It turns out that actuarial<br />

calculation of time to LR is not statistically valid. 3 RT<br />

reduced the annual rate of any failure (DM or LR) over<br />

the first 10 years by about 1/2 (response rate [RR] �<br />

0.52). RT reduced the annual rate of breast cancer<br />

death by about 1/6 (RR � 0.82). RT, of course, mostly<br />

reduced first LR failure. Also published in Lancet in<br />

2011 was the EBCTCG meta-analysis of the impact of<br />

adjuvant tamoxifen taken for 5 years. 4 Tamoxifen reduced<br />

the annual rate of any failure over the first 10<br />

years by about 1/2 (RR � 0.53), very similar to the<br />

impact of RT after BCS. Tamoxifen reduced the annual<br />

rate of death from breast cancer by about 1/3 (RR �<br />

0.68), greater than the impact of RT since tamoxifen<br />

reduced first LR and DM failures about equally. The<br />

updated results from the EBCTCG continue to show<br />

that improved local treatment is still linked to improved<br />

long-term survival. However, the new “4:1 ratio” is<br />

between the reduction in first failure at 10 years (not<br />

the reduction in local-regional recurrence [LRR] at 5<br />

years) and the reduction in mortality at 15 years.<br />

There have been two recent sets of observations of<br />

importance. One is that adjuvant systemic therapy developed<br />

to address the problem of micrometastatic disease<br />

serendipitously has served to make RT much more<br />

effective. This is seen most clearly in National Surgical<br />

Adjuvant <strong>Breast</strong> and Bowel Project (NSABP) trials B-14<br />

and B-13. In the NSABP B-14 trial, women with nodenegative,<br />

estrogen-receptor (ER) positive tumors were<br />

randomly assigned to tamoxifen or placebo; the 10-year<br />

rate of local recurrence after BCS was reduced from<br />

14.7% in the placebo group to 4.3% in the tamoxifen<br />

group. 5 Similarly, in the NSABP B-13 trial, women with<br />

node-negative, ER-negative tumors were randomly assigned<br />

to methotrexate and 5-fluorouracil or to no treatment;<br />

a reduction in 10-year local recurrence rate from<br />

13.4% to 2.6% was observed. 6 This was also seen in the<br />

updated EBCTCG results where RT reduced first failure<br />

by 62% in patients with ER-positive cancers treated<br />

with tamoxifen but only by 35% in patients with ERnegative<br />

cancers (without chemotherapy or trastuzumab).<br />

2 The second set of observations of importance<br />

is that the molecular subtype of breast cancer, approximated<br />

by hormone receptor, HER2, and grade (or Ki-<br />

67), is the major determinant of the success of RT. 7<br />

Finally, the results of MA.20 were provocative. In this<br />

trial, node-positive or high-risk, node-negative patients<br />

(85% had one to three positive nodes) treated with BCS<br />

were randomly assigned to whole breast irradiation<br />

(WBI) or WBI and regional nodal irradiation (RNI). RNI<br />

involved irradiation of the internal mammary nodes, supra-


clavicular, and level 3 axillary nodes. Ninety-one percent<br />

of patients were treated with adjuvant chemotherapy<br />

and all hormone receptor–positive patients were<br />

treated with adjuvant hormonal therapy. Nine hundred<br />

sixteen patients were randomly assigned to each arm.<br />

RNI, in addition to WBI, increased disease-free survival<br />

at 5 years with reductions in both local-regional and<br />

distant recurrence. There was a trend in improvement<br />

in overall survival. A similar randomization was performed<br />

in a European Organisation for Research and<br />

Treatment of <strong>Cancer</strong> (EORTC) trial with results due<br />

in 2014. If the MA.20 results are seen in the EORTC<br />

trial, the results can be hypothesized to demonstrate<br />

that with increasingly effective systemic therapy, local<br />

therapy assumes greater benefit in survival as shown in<br />

Fig. 2.<br />

Author’s Disclosures of Potential Conflicts of Interest<br />

Author<br />

Harris, Jay R.*<br />

*No relevant relationships to disclose.<br />

Employment or<br />

Leadership<br />

Positions<br />

Consultant or<br />

Advisory Role<br />

References<br />

1. Punglia R, Morrow M, Winer E, et al. Local therapy and survival<br />

in breast cancer. N Engl J Med. 2007: 356:2399-2405.<br />

2. Darby S, McGale P, Correa C, et al. Effect of radiotherapy after<br />

breast-conserving surgery on 10-year recurrence and 15-year<br />

breast cancer death: meta-analysis of individual patient data for<br />

10,801 women in 17 randomised trials. Lancet. 2011;378:1707-<br />

1716.<br />

3. Gelman R, Gelber R, Henderson IC, et al. Improved methodology<br />

for analyzing local and distant recurrence. J Clin Oncol. 1990:8;<br />

548-555.<br />

4. Early <strong>Breast</strong> <strong>Cancer</strong> Trialists’ Collaborative Group, Davies C,<br />

Godwin J, et al. Relevance of breast cancer hormone receptors and<br />

other factors to the efficacy of adjuvant tamoxifen: patient-level<br />

meta-analysis of randomised trials. Lancet. 2011;27:771-784.<br />

5. Fisher B, Dignam J, Bryant J, et al. Five versus more than five<br />

Stock<br />

Ownership Honoraria<br />

Research<br />

Funding<br />

EDUCATIONAL SUMMARIES<br />

Fig. 2. Hypothetical relationship between increasing<br />

effectiveness of systemic therapy and the benefit<br />

of local therapy on survival.<br />

Expert<br />

Testimony<br />

Other<br />

Remuneration<br />

years of tamoxifen therapy for breast cancer patients with negative<br />

lymph nodes and estrogen receptor-positive tumors. J Natl<br />

<strong>Cancer</strong> Inst. 1996;88:1529-1542.<br />

6. Fisher B, Dignam J, Mamounas EP, et al. Sequential methotrexate<br />

and fluorouracil for the treatment of node-negative breast<br />

cancer patients with estrogen receptor-negative tumors: eightyear<br />

results from National Surgical Adjuvant <strong>Breast</strong> and Bowel<br />

Project (NSABP) B-13 and first report of findings from NSABP<br />

B-19 comparing methotrexate and fluorouracil with conventional<br />

cyclophosphamide, methotrexate, and fluorouracil. J Clin Oncol.<br />

1996;14:1982-1992.<br />

7. Arvold ND, Taghian AG, Niemierko A, et al. Age, breast cancer<br />

subtype approximation, and local recurrence after breastconserving<br />

therapy. J Clin Oncol. 2011;29:3885-3891.<br />

8. Whelan TJ, Olivotto I, Ackerman I, et al. NCIC-CTG MA.20: An<br />

intergroup trial of regional nodal irradiation in early breast cancer.<br />

J Clin Oncol. 2001;29 (suppl; LBA1003).<br />

69

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!