REVIEW
Infectious diseases in patients receiving monoclonal antibodies
 
More details
Hide details
1
Pneumonologist, Assistant Professor of Critical Care Medicine, 2nd Critical Care Medicine Department, National and Kapodistrian University of Athens Medical School, University Hospital “Attiko”, Chaidari, Attiki, Greece
 
2
Pneumonologist, Critical Care Consultant, 2nd Critical Care Medicine Department, National and Kapodistrian University of Athens Medical School, University Hospital “Attiko”, Chaidari, Attiki, Greece
 
3
Pneumonologist, Critical Care Fellow, 2nd Critical Care Medicine Department, National and Kapodistrian University of Athens Medical School, University Hospital “Attiko”, Chaidari, Attiki, Greece
 
4
Pneumonologist, Critical Care Fellow, 2nd Critical Care Medicine Department, National and Kapodistrian University of Athens Medical School, University Hospital “Attiko”, Chaidari, Attiki, Greece.
 
5
General Surgeon, Critical Care Consultant, 2nd Critical Care Medicine Department, National and Kapodistrian University of Athens Medical School, University Hospital “Attiko”, Chaidari, Attiki, Greece
 
6
Pneumonologist, Professor of Critical Care Medicine, 2nd Critical Care Medicine Department, National and Kapodistrian University of Athens Medical School, University Hospital “Attiko”, Chaidari, Attiki, Greece
 
 
Corresponding author
George Dimopoulos   

University of Athens, University Hospital “Attiko”, Rimini 1, 12462, Chaidari
 
 
Pneumon 2012;25(4):402-409
 
KEYWORDS
ABSTRACT
Monoclonal antibodies (MA) act against specific antigens and are indicated for the treatment of inflammatory and neoplastic diseases as well as in the control of immune responses in transplant patients. More than 30 MA are currently in use in clinical practice, including rituximab, targeting the CD-20 antigen of B lymphocytes, bevacizumab against vascular endothelial growth factor (VEGF), infliximab and adalimubab against tumour necrosis factor alpha (TNF-α) and omalizumab, which neutralizes circulating IgE. The use of MA has been associated with infective complications due to their immunomodulatory action. The prevalence of herpes virus infections, Pneumocystis jirovecii pneumonia and Mycoplasma tuberculosis infection is increased in patients receiving MA. Prevention of these infective complications is based on the clinical evaluation of patients before treatment with MA and close monitoring of the parameters associated with infections during treatment.
 
REFERENCES (50)
1.
Yamada T. Therapeutic Monoclonal Antibodies. J Med 2011; 60 (2):37–46.
 
2.
Goldstein G. Overview of the development of Octhoclone OKT3: monoclonal antibody for therapeutic use in transplantation. Transplant Proc 1987;2(suppl I):1-6.
 
3.
Lanini S, Molloy AC, Fine E, Prentice A, Ippolito G, Kibbler C. Risk of infection in patients with lymphoma receiving rituximab: systematic review and meta-analysis. BMC Medicine 2011;9;36:2-14.
 
4.
Demidem A, Lam T, Alas S, Hariharan K, Hanna N, Bonavida B: Chimeric anti-CD20 (IDEC-C2B8) monoclonal antibody sensitizes a B cell lymphoma cell line to cell killing by cytotoxic drugs. Cancer Biother Radiopharm 1997; 12: 177–186.
 
5.
Maloney G, Liles M, Czerwinski K, et al. Phase I clinical trial using escalating single-dose infusion of chimeric anti-CD20 monoclonal antibody (IDEC-C2B8) in patients with recurrent B-cell lymphoma. Blood 1994; 84: 2457-66.
 
6.
van Der Velden H, te Marvelde G, Hoogeveen G, et al. Targeting of the CD33-calicheamicin immunoconjugate Mylotarg (CMA676) in acute myeloid leukemia: in vivo and in vitro saturation and internalization by leukemic and normal myeloid cells. Blood 2001;97:3197–3204.
 
7.
Dedon C, Salzberg A, Xu J: Exclusive Larson A, Sievers L, Stadtmauer A, et al: Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence. Cancer 2005;104: 1442–1452.
 
8.
Salnikov V, Heldin E, Stuhr B, et al. Inhibition of carcinoma cellderived VEGF reduces inflammatory characteristics in xenograft carcinoma. Int J Cancer 2006; 119:2795–2802.
 
9.
Sullivan A, Carbon G, Roland L, et al. r84, a novel therapeutic antibody against mouse and human VEGF with potent antitumor activity and limited toxicity induction. PLoS One 2010; 5:e12031.
 
10.
Osbourn J, Groves M, Vaughan T. From rodent reagents to human therapeutics using antibody guided selection. Methods 2005;36: 61–68.
 
11.
Voulgari V, Drosos A. Adalimumab for rheumatoid arthritis. Expert Opin Biol Ther 2006;6: 1349–60.
 
12.
Shealy D, Cai A, Staquet K, et al. Characterization of golimumab, a human monoclonal antibody specific for human tumor necrosis factor alpha. MAbs 2010;2:428–439.
 
13.
Pollack P, Groothuis JR. Development and use of palivizumab (Synagis): a passive immunoprophylactic agent for RSV. J Infect Chemother 2002; 8: 201–206.
 
14.
Gruppo A, Rother P. Eculizumab for congenital atypical hemolytic–uremic syndrome. N Engl J Med 2009;360:544-6.
 
15.
Rodrigo GJ, Neffen H, Castro-Rodriguez JA. Efficacy and Safety of Subcutaneous Omalizumab vs Placebo as Add-on Therapy to Corticosteroids for Children and Adults With Asthma. Chest 2011;139:28-35.
 
16.
Haldar P, Brightling E, Hargadon B, et al. Mepolizumab and Exacerbations of Refractory Eosinophilic Asthma. N Engl J Med 2009;360:973-984.
 
17.
Wenzel S, Wilbraham D, Fuller R, Getz B, Longphre M. Effect of aninterleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. The Lancet 2007;370:1422-31.
 
18.
Busse W, Israel E, Nelson S, et al and the Daclizumab Asthma Study Goup: Daclizumab Improves Asthma Control in Patients with Moderate to Severe Persistent Asthma: A Randomized, Controlled Trial. Am J Respir Crit Care Med 2008;178:1002-8.
 
19.
Yamada K, Hayashi M, Du W, et al. Localization of CD26/DPPIV in nucleus and its nuclear translocation enhanced by antiCD26 monoclonal antibody with anti-tumor effect. Cancer Cell Int 2009; 9: 17.
 
20.
Gentille G, Foà R. Viral infections associated with the clinical use of monoclonal antibodies. Clin Microb Infect 2011;17(12):1768-75.
 
21.
Shale M, Seow C, Coffin C, Kaplan G, Panaccione R, Ghosh S. Review article: chronic viral infection in the anti-tumour necrosis factor thearpy era in inflammatory bowel disease. Aliment Pharmacol Ther 2010;31:20-34.
 
22.
Ljungman P, de la Camara R, Milpied N et al. Randomized study of valaciclovir prophylaxis against cytomegalovirus reactivation in recipients of allogenic bone marrow transplantation. Blood 2002;99:3050-56.
 
23.
Ιssa N, Fishman J. Infectious complications of antilymphocyte therapies in solid organ transplantation. Clin Infect Dis 2009;48:772-86.
 
24.
Styczynski J, Einsele H, Gil L, Ljungman P. Outcome of treatment of Epstein-Barr virus-related lymphoproliferative disorder in hematopoietic stem cell recipients: a comprehensive review of the reported cases. Transpl Infect Dis 2009;11:383-92.
 
25.
Stranfeld A, Listing J, Herzer P et al. Risk of herpes zoster in patients with rheumatoid arthritis treated with anti-TNF-alpha agents. JAMA 2009;301:737-44.
 
26.
Scemla A, Loupy A, Candon S et al. Incidence of infectious complications in highly sensitized renal transplant recipients treated by rirituximab: a case-controlled study. Transplantation 2010;90:1180-84.
 
27.
Look ASF, McMahon BJ. Chronic hepatitis B update 2009. Hepatology 2009;50:1-35.
 
28.
Ferri C, Govoni M, Calabrese L. The ABCs of viral hepatitis in the biologic era. Curr Opin Rheumatol 2010;22:443-50.
 
29.
Lin PL, Plessner HL, Voitenok NN, et al. Tumor necrosis factor and tuberculosis. J Investig Dermatol Sym Proc 2007; 12: 22–25.
 
30.
Harris J, Keane J. How tumour necrosis factor blockers interfere with tuberculosis immunity. Clinical and Experimental Immunology 2010;161: 1–9.
 
31.
Villiger M, Zellweger P, Moller B. Novel screening tools for latent tuberculosis: time to leave an old friend? Curr Opin Rheumatol 2009; 21: 238–24.
 
32.
Solovic Ι, Sester M, Gomez-Reino J et al. The risk of tuberculosis related to tumour necrosis factor antagonist therapies: a TBNET consensus statement. Eur Respir J 2010; 36: 1185–1206.
 
33.
Morrison VA. Update on prophylaxis and therapy of infection in patients with chronic lymphocytic leukemia. Expert Rev Anticancer Ther 2001;1:84–90.
 
34.
Kamel S, O’Connor S, Lee N, Tadmor T, McLaughlin P, Polliack A. A resurgence of Pneumocystis in aggressive lymphoma treated with R-CHOP 14: the price of a dose-dense regimen? Leuk Lymphoma 2010;51:737–738.
 
35.
Brusamolino E, Rusconi C, Montalbetti L, et al. Dose-dense R-CHOP-14 supported by pegfilgrastim in patients with diffuse large B-cell lymphoma: a phase II study of feasibility and toxicity. Haematologica 2006;91:496–502.
 
36.
Kolstad A, Holte H, Fossa A, Lauritzsen GF, Gaustad P, Torfoss D Pneumocystis jirovecii pneumonia in B-cell lymphoma patients treated with the rituximab-CHOEP-14 regimen. Haematologica 2007;92:139–140.
 
37.
Venhuizen AC, Hustinx WN, van Houte AJ, Veth G, van der Griend R. Three cases of Pneumocystis jirovecii pneumonia (PCP) during first-line treatment with rituximab in combination with CHOP-14 for aggressive B-cell non-Hodgkin’s lymphoma. Eur J Haematol 2008;80:275–276.
 
38.
Pfreundschuh M, Trumper L, Kloess M, et al. Two-weekly or 3-weekly CHOP chemotherapy with or without etoposide for the treatment of elderly patients with aggressive lymphomas: results of NHL-B2 trial of the DSHNHL. Blood 2004;104: 634–641.
 
39.
Hamilton CD. Infectious complications of treatment with biologic agents. Curr Opin Rheumatol. 2004;16(4):393-398.
 
40.
Cabanillas F, Liboy I, Pavia O, Rivera E. High incidence of nonneutropenic infections induced by rituximab plus fludarabine and associated with hypogammaglobulinemia:a frequently unrecognized and easily treatable complication. Ann Oncol. 2006;17:1424–7.
 
41.
Lim SH, Esler WV, Zhang Y, et al. B-cell depletion for 2 years after autologous stem cell transplant for NHLinduces prolonged hypogammaglobulinemia beyond the rituximab maintenance period. Leuk Lymphoma 2008;49:152–3.
 
42.
Tsiodras S, Samonis G, Boumpas D, Kontoyiannis D. Fungal Infections Complicating Tumor Necrosis Factor α Blockade TherapyMayo Clin Proc 2008;83(2):181-194.
 
43.
Busca A, Locatelli F, Marmont F, Ceretto C, Falda M. Recombinant human soluble tumor necrosis factor receptor fusion protein as treatment for steroid refractory graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. Am J Hematol. 2007;82(1):45-52.
 
44.
Singh P, Taylor SF, Murali R, Gomes LJ, Kanthan GL, Maloof AJ. Disseminated mucormycosis and orbital ischaemia in combination immunosuppression with a tumour necrosis factor-α inhibitor. Clin Experiment Ophthalmol. 2007;35(3):275-280.
 
45.
Belda A, Hinojosa J, Serra B, Garcia L, Merino C, Moles JR. Systemic candidiasis and infliximab therapy. Gastroenterol Hepatol. 2004;27(6):365-367.
 
46.
Hoang JK, Burruss J. Localized cutaneous Cryptococcus albidus infection in a 14-year-old boy on etanercept therapy. Pediatr Dermatol. 2007; 24(3):285-288.
 
47.
Jain VV, Evans T, Peterson MW. Reactivation histoplasmosis after treatment with anti-tumor necrosis factor α in a patient from a non-endemic area. Respir Med. 2006 Jul;100(7):1291-93.
 
48.
Sawalha AH, Lutz BD, Chaudhary NA, Kern W, Harley JB, Greenfield RA. Panniculitis: a presenting manifestation of disseminated histoplasmosis in a patient with rheumatoid arthritis. J Clin Rheumatol. 2003;9(4): 259-262.
 
49.
Gottlieb GS, Lesser CF, Holmes KK, Wald A. Disseminated sporotrichosis associated with treatment with immunosuppressants and tumor necrosis factor-α antagonists. Clin Infect Dis. 2003 Sep 15;37(6):838-840.
 
50.
Khoury JA, Dubberke ER, Devine SM. Fatal case of protothecosis in a hematopoietic stem cell transplant recipient after infliximab treatment for graft-versus-host disease. Blood 2004;104(10):3414-15.
 
eISSN:1791-4914
ISSN:1105-848X
Journals System - logo
Scroll to top