Immunotherapy for Infectious Diseases

Immunotherapy for Infectious Diseases Immunotherapy for Infectious Diseases

ibvacunas.com
from ibvacunas.com More from this publisher
10.02.2013 Views

Tuberculosis and Other Mycobacterial Infections 293 and Iran also suggested activity in TB patients with drug-susceptible and drugresistant tuberculosis (149–152). These studies suffered from methodologic problems including insufficient sample sizes, nonrandom treatment allocation, high losses to follow-up, and the use of various TB drug treatment regimens (153). Three recent clinical trials have examined the role of immunotherapy with heat-killed M. vaccae in a more rigorous fashion. In Romania, 206 previously untreated patients with pulmonary TB were randomized to receive M. vaccae immunotherapeutic agent or placebo 1 month after the beginning of anti-TB treatment (151). In this trial, which included patients with both drug-susceptible and drug-resistant TB, sputum cultures 1 month after the administration of M. vaccae (i.e., 2 months after the onset of anti-TB chemotherapy) were negative in 86% of patients in the immunotherapy group compared with 76% of the placebo arm ( p � 0.08). Patients who received M. vaccae had significantly greater weight gain after 2 and 6 months of TB treatment and decrement in cavitary disease at 6 months. In a companion study of 102 patients with chronic or relapsed TB, 60% of whom were infected with bacilli resistant to at least one first-line drug, 77% patients treated with M. vaccae had successful treatment outcomes at 1 year compared with 52% of patients treated with chemotherapy only (p � 0.02) (150). Sputum culture negativity at 2 months was significantly higher in M. vaccae than placebo recipients. In contrast, a randomized clinical trial from South Africa found no difference in the rate of sputum culture conversion, weight gain, radiographic improvement, survival, or decrease in erythrocyte sedimentation rate after 2 months of TB treatment (154). This study included 374 HIV-infected and HIV-noninfected patients with pulmonary TB, treated with standard short-course chemotherapy and a single intradermal injection of heat-killed M. vaccae or placebo 1 week after the onset of anti-TB treatment. In a similar clinical trial done in HIV-noninfected TB patients in Uganda, the rate of sputum culture conversion after 1 month of TB treatment was twofold higher in the M. vaccae group compared with the placebo arm after 1 month of TB treatment ( p � 0.01) and was comparable between the groups thereafter (155). Weight gain and improvement in cough and chest pain did not differ between treatment groups. Treatment with M. vaccae was also associated with greater improvement in radiographic extent of disease at the end of anti-TB treatment and at 1-year follow-up. The reasons underlying the disparate results of these studies are unclear but may reflect differences in exposure and sensitization to environmental mycobacteria between the trial sites that might obscure any potential benefit from the immunotherapeutic agent (148). At the present time, immunotherapy with M. vaccae should continue to be regarded as experimental therapy. The promising results in several studies suggest that further research with M. vaccae is warranted. Another large study of M. vaccae immunotherapy is currently under way in Zambia. CONCLUSIONS The evolution of Mycobacterium tuberculosis as an intracellular pathogen has led to a complex relationship between the organism and its host, the human mononuclear phagocyte. The products of M. tuberculosis-specific T-lymphocytes, particularly IFN-�, are essential for macrophage activation for intracellular mycobacterial killing. However, some cytokines, including products of both lymphocytes and phagocytic cells, may contribute to disease pathogenesis, by enhancing mycobacterial survival and by causing many of the pathologic features of the disease. In HIV-associated mycobacterial

294 Wallis and Johnson infections, cytokines may mediate accelerated progression of HIV disease. The objectives of adjunctive immunotherapy for tuberculosis are also complex. In some situations, such as MDR disease, clearance of bacilli may be enhanced by administration of IL-2, IL-12, or IFN-�, or possibly by using inhibitors of the deactivating cytokines TGF-� and IL-10. In other circumstances, such as in HIV coinfection, it may be desirable to reduce the nonspecific inflammatory response—and thereby reduce HIV expression—using inhibitors of TNF-� such as pentoxifylline, prednisone, or soluble TNF receptor. Further clinical trials are needed to define the clinical role for immunotherapy of tuberculosis and other mycobacterial infections. REFERENCES 1. Global Tuberculosis Programme. Global Tuberculosis Control WHO Report 1999. Geneva: WHO, 1999. 2. Global Tuberculosis Programme. Anti-tuberculosis drug resistance in the world. 1998. 3. Villarino ME, Dooley SW, Geiter LJ, Castro KG, Snider DE Jr. Management of persons exposed to multidrug-resistant tuberculosis. MMWR 1992; 41:61–71. 4. Selwyn PA, Alcabes P, Hartel D, et al. Clinical manifestations and predictors of disease progression in drug users with human immunodeficiency virus infection. N Engl J Med 1992; 327:1697–1703. 5. Di Perri G, Cruciani M, Danzi MC, et al. Nosocomial epidemic of active tuberculosis among HIV-infected patients. Lancet 1989; 2:1502–1504. 6. Daley CL, Small PM, Schecter GF, et al. An outbreak of tuberculosis with accelerated progression among persons infected with the human immunodeficiency virus. An analysis using restriction-fragment-length polymorphisms. N Engl J Med 1992; 326:231–235. 7. Crowle AJ, Elkins N. Relative permissiveness of macrophages from black and white people for virulent tubercle bacilli. Infect Immun 1990; 58:632–638. 8. Skamene E, Forget A. Genetic basis of host resistance and susceptibility to intracellular pathogens. Adv Exp Med Biol 1988; 239:23–37. 9. Radzioch D, Hudson T, Boule M, Barrera L, Urbance JW, Varesio L, Skamene E. Genetic resistance/susceptibility to mycobacteria: phenotypic expression in bone marrow derived macrophage lines. J Leukoc Biol 1991; 50:263–272. 10. Stach JL, Gros P, Forget A, Skamene E. Phenotypic expression of genetically-controlled natural resistance to Mycobacterium bovis (BCG). J Immunol 1984; 132:888–892. 11. Goto Y, Buschman E, Skamene E. Regulation of host resistance to Mycobacterium intracellulare in vivo and in vitro by the bcg gene. Immunogenetics 1989; 30:218–221. 12. Bellamy R, Ruwende C, Corrah T, McAdam KP, Whittle HC, Hill AV. Variations in the NRAMP1 gene and susceptibility to tuberculosis in West Africans [see comments]. N Engl J Med 1998; 338:640–644. 13. Bermudez LE, Wu M, Young LS. Interleukin-12-stimulated natural killer cells can activate human macrophages to inhibit growth of M. avium. Infect Immun 1995; 63:4099–4104. 14. Fujiwara H, Kleinhenz ME, Wallis RS, Ellner JJ. Increased interleukin-1 production and monocyte suppressor cell activity associated with human tuberculosis. Am Rev Respir Dis 1986; 133:73–77. 15. Takashima T, Ueta C, Tsuyuguchi I, Kishimoto S. Production of tumor necrosis factor alpha by monocytes from patients with pulmonary tuberculosis. Infect Immun 1990; 58:3286–3292. 16. Chensue SW, Warmington KS, Berger AE, Tracey DE. Immunohistochemical demonstration of interleukin-1 receptor antagonist protein and interleukin-1 in human lymphoid tissue and granulomas. Am J Pathol 1992; 140:269–275. 17. Kindler V, Sappino AP. The beneficial effects of localized tumor necrosis factor production in BCG infection. Behring Inst Mitt 1991; 88:120–124.

294 Wallis and Johnson<br />

infections, cytokines may mediate accelerated progression of HIV disease. The objectives<br />

of adjunctive immunotherapy <strong>for</strong> tuberculosis are also complex. In some<br />

situations, such as MDR disease, clearance of bacilli may be enhanced by administration<br />

of IL-2, IL-12, or IFN-�, or possibly by using inhibitors of the deactivating<br />

cytokines TGF-� and IL-10. In other circumstances, such as in HIV coinfection, it may<br />

be desirable to reduce the nonspecific inflammatory response—and thereby reduce<br />

HIV expression—using inhibitors of TNF-� such as pentoxifylline, prednisone, or<br />

soluble TNF receptor. Further clinical trials are needed to define the clinical role <strong>for</strong><br />

immunotherapy of tuberculosis and other mycobacterial infections.<br />

REFERENCES<br />

1. Global Tuberculosis Programme. Global Tuberculosis Control WHO Report 1999. Geneva:<br />

WHO, 1999.<br />

2. Global Tuberculosis Programme. Anti-tuberculosis drug resistance in the world. 1998.<br />

3. Villarino ME, Dooley SW, Geiter LJ, Castro KG, Snider DE Jr. Management of persons<br />

exposed to multidrug-resistant tuberculosis. MMWR 1992; 41:61–71.<br />

4. Selwyn PA, Alcabes P, Hartel D, et al. Clinical manifestations and predictors of disease<br />

progression in drug users with human immunodeficiency virus infection. N Engl J Med<br />

1992; 327:1697–1703.<br />

5. Di Perri G, Cruciani M, Danzi MC, et al. Nosocomial epidemic of active tuberculosis<br />

among HIV-infected patients. Lancet 1989; 2:1502–1504.<br />

6. Daley CL, Small PM, Schecter GF, et al. An outbreak of tuberculosis with accelerated progression<br />

among persons infected with the human immunodeficiency virus. An analysis<br />

using restriction-fragment-length polymorphisms. N Engl J Med 1992; 326:231–235.<br />

7. Crowle AJ, Elkins N. Relative permissiveness of macrophages from black and white people<br />

<strong>for</strong> virulent tubercle bacilli. Infect Immun 1990; 58:632–638.<br />

8. Skamene E, Forget A. Genetic basis of host resistance and susceptibility to intracellular<br />

pathogens. Adv Exp Med Biol 1988; 239:23–37.<br />

9. Radzioch D, Hudson T, Boule M, Barrera L, Urbance JW, Varesio L, Skamene E. Genetic<br />

resistance/susceptibility to mycobacteria: phenotypic expression in bone marrow derived<br />

macrophage lines. J Leukoc Biol 1991; 50:263–272.<br />

10. Stach JL, Gros P, Forget A, Skamene E. Phenotypic expression of genetically-controlled<br />

natural resistance to Mycobacterium bovis (BCG). J Immunol 1984; 132:888–892.<br />

11. Goto Y, Buschman E, Skamene E. Regulation of host resistance to Mycobacterium intracellulare<br />

in vivo and in vitro by the bcg gene. Immunogenetics 1989; 30:218–221.<br />

12. Bellamy R, Ruwende C, Corrah T, McAdam KP, Whittle HC, Hill AV. Variations in the<br />

NRAMP1 gene and susceptibility to tuberculosis in West Africans [see comments].<br />

N Engl J Med 1998; 338:640–644.<br />

13. Bermudez LE, Wu M, Young LS. Interleukin-12-stimulated natural killer cells can activate<br />

human macrophages to inhibit growth of M. avium. Infect Immun 1995; 63:4099–4104.<br />

14. Fujiwara H, Kleinhenz ME, Wallis RS, Ellner JJ. Increased interleukin-1 production and<br />

monocyte suppressor cell activity associated with human tuberculosis. Am Rev Respir Dis<br />

1986; 133:73–77.<br />

15. Takashima T, Ueta C, Tsuyuguchi I, Kishimoto S. Production of tumor necrosis factor<br />

alpha by monocytes from patients with pulmonary tuberculosis. Infect Immun 1990;<br />

58:3286–3292.<br />

16. Chensue SW, Warmington KS, Berger AE, Tracey DE. Immunohistochemical demonstration<br />

of interleukin-1 receptor antagonist protein and interleukin-1 in human lymphoid tissue<br />

and granulomas. Am J Pathol 1992; 140:269–275.<br />

17. Kindler V, Sappino AP. The beneficial effects of localized tumor necrosis factor production<br />

in BCG infection. Behring Inst Mitt 1991; 88:120–124.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!