Skip to main content
The West Indian Medical Journal logoLink to The West Indian Medical Journal
. 2015 Jul 28;63(6):650–654. doi: 10.7727/wimj.2013.241

Monoclonal Antibodies in Cancer Therapy: Mechanisms, Successes and Limitations

Los Anticuerpos Monoclonales en el Tratamiento del Cáncer: Mecanismos, Éxitos y Limitaciones

A Coulson 1, A Levy 1, M Gossell-Williams 1,
PMCID: PMC4663950  PMID: 25803383

ABSTRACT

Rituximab was the first chemotherapeutic monoclonal antibody (CmAb) approved for clinical use in cancer therapeutics in 1997 and has significantly improved the clinical outcomes in non-Hodgkin's lymphoma. Since then, numerous CmAbs have been developed and approved for the treatment of various haematologic and solid human cancers. In this review, the classification, efficacy and significantly reduced toxicity of CmAbs available for use in the United States of America are presented. Finally, the limitations of CmAbs and future considerations are explored.

Keywords: Antigenic targets, cancer therapy, clinical benefits, monoclonal antibodies

INTRODUCTION

Kohler and Milstein (1) revolutionized anti-cancer therapeutics in the late 19th century with the development of hybridoma technology, now used to produce monoclonal antibodies. Since then, chemotherapeutic monoclonal antibodies (CmAbs) have emerged as standard therapeutic agents for many haematological and solid cancers in humans in the last decade. Cancer cells share many similarities with the normal host cells and this presents a challenge for achieving high levels of selective cytotoxicity. Chemotherapeutic monoclonal antibodies were engineered with the predicted advantage of specificity, thus acting as ‘targeting missiles’ toward cancer cells (2).

Classification of chemotherapeutic monoclonal antibodies

Advances in genetic engineering techniques have resulted in the development of four main types of CmAbs: murine, chimeric, humanized and human CmAbs. Murine CmAbs, derived exclusively from mouse, were the first to be applied in cancer chemotherapeutics. Utilization, however, was rapidly revoked because of inability to effectively interact with components of the human immune system due to their foreign nature and subsequent limited recognition by the host immune system. Chimeric CmAbs typically comprise variable regions derived from a murine source and constant regions (65%) derived from a human source (3). Chimeric CmAbs can also be non-humanized (chimeric trifunctional CmAbs), rat-mouse hybrid monoclonal antibodies that have three different antigen-binding specificities: for tumour cells, T lymphocyte cells and one for accessory cells (4). The development of chimeric CmAbs that possess a fully human Fc portion provided considerably less immunogenic and more efficient interaction with human effector cells and the complement system than murine CmAbs (5). Humanized CmAbs are predominantly (90%) engineered from a human source with the exception that the complementarity-determining regions of the Fab portion are of murine origin; they are even less immunogenic than chimeric CmAbs. Human CmAbs, which are 100% human, are engineered from transgenic mice, and compared to chimeric and humanized CmAbs, have higher affinity values toward human antigens and minimal or no hypersensitivity responses (6).

Chemotherapeutic monoclonal antibodies may be conjugated to other forms of cancer therapy and this facilitates greater efficacy. More importantly, conjugation provides targeted attack at cancer cells and therefore reduced widespread systemic toxicities to normal cells. There are three types of conjugated CmAbs: radiolabelled CmAbs which are linked to radionuclide particles (7), chemolabelled CmAbs which are attached to anti-neoplastic drugs (8) and immunotoxin CmAbs which are attached to plant and bacterial toxins (9). Table 1 lists the CmAbs approved by the United States of America (USA) Food and Drug Administration (FDA) for use in oncology by type and year approved.

Table 1. Unconjugated and conjugated monoclonal antibodies currently approved by the Food and Drug Administration (FDA) for cancer therapy.

CmAb Type Year approved
Unconjugated
Rituximab Chimeric IgG1 1997
Trastuzumab Humanized IgG1 1998
Alemtuzumab Humanized IgG1 2001
Tositumomab Murine IgG2a 2003
Cetuximab Chimeric IgG1 2004
Bevacizumab Humanized IgG1 2004
Panitumumab Human IgG2 2006
Catumaxomab* Chimeric mouse-rat hybrid 2009
Ofatumumab Human IgG1 2009
Ipilimumab Human IgG1 2011
Pertuzumab Humanized IgG1 2012
Denosumab Human IgG2 2013
Conjugated
Ibritumomab tiuxetan Murine IgG1
Radionucleotide (Yttrium90 or Indium111) 2002
Tositumomab Murine IgG2a
Radionucleotide (Iodine131) 2003
Brentuximab vedotin Chimeric IgG1
Drug (Auristatin E) 2011
Trastuzumab emtansine Humanized IgG1
Drug (Mertansine) 2013
*

Approved by European Medicines Agencies and undergoing trials in the USA. CmAb: chemotherapeutic monoclonal antibody.

Source: 10, 37

Mechanisms of action of CmAbs: a targeted approach with a promising future

Chemotherapeutic monoclonal antibodies target cancer cells by binding to cell surface antigens. Cell surface antigens include antigens associated with growth and differentiation, such as cluster of differentiation (CD; eg CD20, CD 30, CD 33 and CD52), carcino-embryonic antigen (CEA), epidermal growth factor receptor (EGFR), receptor activator of nuclear factor kappa-B ligand (RANKL), human epidermal growth factor receptor 2 (HER2), vascular endothelial growth factor (VEGF), VEGF receptor (VEGFR), integrins (eg αVβ3 and α5β1), fibroblast activation protein (FAP) and extracellular matrix metalloproteinase inducers [EMMPRIN] (1013). Once CmAbs attach to the specific target antigen tumour, cell destruction is effected through three main mechanisms:

  • direct tumour cell death by mechanisms such as targeting and inhibition of cell survival signalling, the induction of apoptosis or through the direct delivery of cytotoxic drugs or radioisotope modalities by conjugated antibodies (1416).

  • immune mediated tumour cell killing by engaging antibody-dependent-cell-mediated-cytotoxicity, complement-mediated-cytotoxicity and activating cellular phagocytosis (4, 17, 18). Additionally, immunostimulatory CmAbs can activate T lymphocyte cells through the inhibition of T lymphocyte inhibitory receptors (19).

  • vascular ablation and disruption of stromal interaction with cancer cells. This denies tumours of blood supply and supporting network promotes tumour regression (20).

Clinical successes

Table 2 lists FDA approved CmAbs for haematologic and solid cancers. Among the most noteworthy candidates for haematologic tumours are rituximab, alemtuzumab and ofatumumab, brentuximab vedotin, 131I-tositumomab and 90Y-iIbritumomab tiuxetan. Rituximab was the first CmAb approved for clinical use in cancer therapeutics and has proven to be highly effective in increasing the overall survival rates in lymphoproliferative disorders. In a study involving 48 patients with chronic or small lymphocytic leukaemia, rituximab therapy resulted in an overall response rate of 58%, with 9% complete responses (21). Similar success has been reported for treatment of follicular lymphoma (22) and diffuse large B-cell lymphoma (23). Brentuximab vedotin is indicated in Hodgkin's lymphoma and anaplastic large cell lymphoma where it has produced a response rate of greater than 50% in both malignancies and achieved 57% remission in patients with anaplastic large cell lymphoma (24). 131I-tositumomab and 90Y-ibritumomab tiuxetan are indicated for treatment of relapsed or refractory low-grade non-Hodgkin's lymphoma and have achieved high response rates (25).

Table 2. Antigenic targets, cancer indication and mechanism of action (MOA) of the chemotherapeutic monoclonal antibodies (CmAbs) currently approved by the Food and Drug Administration (FDA) for cancer therapy.

CmAb Antigenic target MOA Main cancer indication(s)
Rituximab CD20 ADCC, CMC, induces apoptosis Non-Hodgkin's lymphoma
Alemtuzumab CD52 Induces apoptosis, CMC, ADCC Chronic lymphocytic leukaemia
Tositumomab CD20 ADCC, induces apoptosis Non-Hodgkin's lymphoma
Cetuximab EGFR ADCC, inhibition of EGFR signalling Colorectal cancer, head and neck cancer
Bevacizumab VEGF Inhibition of VEGF signalling Lung cancer, renal cancer, colorectal cancer, brain cancer, breast cancer
Panitumumab EGFR Inhibition of EGFR signalling Colorectal cancer
Catumaxomab* EpCAM ADCC, T-cell mediated lysis, phagocytosis via FcγR accessory cells Malignant ascites in patients with EpCAM +ve cancers
Ofatumumab CD20 ADCC, CMC Chronic lymphocytic leukaemia
Denosumab RANKL Inhibition of RANKL signalling Breast cancer, prostate cancer
Ipilimumab CTLA-4 Melanoma
Pertuzumab HER2 Inhibition of HER2 signalling Breast cancer
90Y-ibritumomab tiuxetan CD20 Radioisotope (90-Yttrium) delivery Non-Hodgkin's lymphoma
131-I tositumomab CD20 Radioisotope (131-Iodine) delivery Non-Hodgkin's lymphoma
Brentuximab vedotin CD30 Cytotoxic drug (auristatin E) delivery Hodgkin's lymphoma, anaplastic large cell lymphoma
Trastuzumab emtansine HER2 Inhibition of HER2 signalling, ADCC Breast cancer
*

Approved by European Medicines Agency and undergoing trials in the USA.

Source: 10, 3740

Trastuzumab, cetuximab, panitumumab, bevacizumab, catumaxomab, ipilimumab and denosumab are FDA approved for use across several solid cancers. Trastuzumab emtansine selectively targets HER2; in a clinical trial involving 137 patients with HER2-positive breast cancer, trastuzumab proved to significantly improve progressive-free survival time (14.2 months) compared to 9.2 months with the conventional chemotherapeutic agent, docetaxel (26). The clinical improvement in response rate, progression-free survival and overall survival of bevacizumab in non-small cell lung cancer were confirmed in a recent meta-analysis which included 15 650 patients collected from 30 randomized clinical trials (27).

Limitations and promising future

Compared with conventional chemotherapy, the adverse effects of unconjugated CmAbs are usually mild, while conjugated CmAbs precipitate severe adverse effects (3). These adverse effects are commonly related to the antigens they target and the intravenous route of administration. For example, bevacizumab targets tumour blood vessel growth and causes adverse effects such as hypertension and kidney damage (28). More than 90% of patients on rituximab therapy experience infusion-related reactions such as cytokine release syndrome and tumour lysis syndrome (23). Intravenous administration of alemtuzumab is associated with lymphopenia and concomitant immunosuppression (29). Conjugated agents such as brentuximab vedotin precipitate cumulative peripheral neuropathy (30) while myelosuppression is the main toxicity of 131I-tositumomab and 90Y-ibritumomab tiuxetan (31). Other adverse effects common to most CmAbs are chills, weakness, headache, nausea, vomiting, diarrhoea, hypotension and rashes.

Although great strides have been made in antibody engineering and cancer therapy, production cost is estimated at twice that required for conventional drugs (32). Production requires the use of very large cultures of cells, which are expensive to maintain, primarily as a consequence of high turnover of disposables, such as media, and the continuous requirement for sophisticated purification steps to ensure clinical quality (33). Thus, the cost to the users is restrictive. In 2012, the calculated per patient cost of treatment of colorectal cancer with CmAbs (bevacizumab, cetuximab and panitumumab) was US$30 400 in comparison to US$17 500 for the use of conventional chemotherapeutic drugs [oxaliplatin, irinotecan, fluorouracil and leucovorin] (34).

Moreover, while the introduction of substitutes (generics) for innovator brands of small drug molecules provides cost savings of 80% to USA medical expense, no such benefit occurs with the biological substitutes (biosimilars), where the savings amounts to 30% at best (35, 36).

There is no debating that the engineering of CmAbs signified a key milestone in cancer therapeutics, and with the success rate of bringing these drugs to the market being better than that of small molecular drugs, it is expected that pharmaceutical companies will continue to progress toward more specific, less toxic and more cost-effective CmAbs.

REFERENCES

  • 1.Köhler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–497. doi: 10.1038/256495a0. [DOI] [PubMed] [Google Scholar]
  • 2.Yoon S, Kim YS, Shim H, Chung J. Current perspectives on therapeutic antibodies. Biotechnol Bioprocess Eng. 2010;15:709–715. [Google Scholar]
  • 3.Hansel TT, Kropshofer H, Singer T, Mitchell JA, George AJ. The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov. 2010;9:325–338. doi: 10.1038/nrd3003. [DOI] [PubMed] [Google Scholar]
  • 4.Seimetz D. Novel monoclonal antibodies for cancer treatment: the trifunctional antibody catumaxomab (removab) J Cancer. 2011;2:309–309. doi: 10.7150/jca.2.309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Reichert JM, Rosensweig CJ, Faden LB, Dewitz MC. Monoclonal antibody successes in the clinic. Nat Biotechnol. 2005;23:1073–1078. doi: 10.1038/nbt0905-1073. [DOI] [PubMed] [Google Scholar]
  • 6.Lonberg N. Human monoclonal antibodies from transgenic mice. In: Chernajovsky Y, Nissim A, editors. Therapeutic anti-bodies. Handbook of experimental pharmacology. Vol. 181. Berlin Heidelberg: Springer-Verlag; 2008. pp. 69–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Steiner M, Neri D. Antibody-radionuclide conjugates for cancer therapy: historical considerations and new trends. Clin Cancer Res. 2011;17:6406–6416. doi: 10.1158/1078-0432.CCR-11-0483. [DOI] [PubMed] [Google Scholar]
  • 8.Flygare JA, Pillow TH, Aristoff P. Antibody-drug conjugates for the treatment of cancer. Chem Biol Drug Des. 2013;81:113–121. doi: 10.1111/cbdd.12085. [DOI] [PubMed] [Google Scholar]
  • 9.Choudhary S, Mathew M, Verma RS. Therapeutic potential of anticancer immunotoxins. Drug Discov Today. 2011;16:495–503. doi: 10.1016/j.drudis.2011.04.003. [DOI] [PubMed] [Google Scholar]
  • 10.Scott AM, Wolchok JD, Old LJ. Antibody therapy of cancer. Nat Rev Cancer. 2012;12:278–287. doi: 10.1038/nrc3236. [DOI] [PubMed] [Google Scholar]
  • 11.Schliemann C, Neri D. Antibody-based targeting of the tumor vasculature. Biochim Biophys Acta. 2007;1776:175–192. doi: 10.1016/j.bbcan.2007.08.002. [DOI] [PubMed] [Google Scholar]
  • 12.Hudis CA. Trastuzumab – mechanism of action and use in clinical practice. New Engl J Med. 2007;357:39–51. doi: 10.1056/NEJMra043186. [DOI] [PubMed] [Google Scholar]
  • 13.Hofmeister V, Vetter C, Schrama D, Bröcker EB, Becker JC. Tumor stroma-associated antigens for anti-cancer immunotherapy. Cancer Immunol Immunother. 2006;55:481–494. doi: 10.1007/s00262-005-0070-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kaminski MS, Estes J, Zasadny KR, Francis IR, Ross CW, Tuck M, et al. Radioimmunotherapy with iodine 131I tositumomab for relapsed or refractory B-cell non-Hodgkin lymphoma: updated results and long-term follow-up of the University of Michigan experience. Blood. 2000;96:1259–1266. [PubMed] [Google Scholar]
  • 15.Nguyen TH, Havari E, McLaren R, Zhang M, Jiang Y, Madden SL, et al. Alemtuzumab induction of intracellular signaling and apoptosis in malignant B lymphocytes. Leuk Lymphoma. 2012;53:699–709. doi: 10.3109/10428194.2011.623253. [DOI] [PubMed] [Google Scholar]
  • 16.Vaklavas C, Forero-Torres A. Safety and efficacy of brentuximab vedotin in patients with Hodgkin lymphoma or systemic anaplastic large cell lymphoma. Ther Adv Hematol. 2012;3:209–225. doi: 10.1177/2040620712443076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Weiner GJ. Rituximab: mechanism of action. Semin Hematol. 2010;47:115–123. doi: 10.1053/j.seminhematol.2010.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hörl S, Banki Z, Huber G, Ejaz A, Müllauer B, Willenbacher E, et al. Complement factor H-derived short consensus repeat 18–20 enhanced complement-dependent cytotoxicity of ofatumumab on chronic lymphocytic leukemia cells. Haematologica. 2013;98:1939–1947. doi: 10.3324/haematol.2013.089615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yang JC, Hughes M, Kammula U, Royal R, Sherry RM, Topalian SL, et al. Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J Immunother. 2007;30:825–825. doi: 10.1097/CJI.0b013e318156e47e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Willett CG, Boucher Y, di Tomaso E, Duda DG, Munn LL, Tong RT, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med. 2004;10:145–147. doi: 10.1038/nm988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hainsworth JD, Litchy S, Barton JH, Houston GA, Hermann RC, Bradof JE, et al. Single-agent rituximab as first-line and maintenance treatment for patients with chronic lymphocytic leukemia or small lymphocytic lymphoma: a phase II trial of the Minnie Pearl Cancer Research Network. J Clin Oncol. 2003;21:1746–1751. doi: 10.1200/JCO.2003.09.027. [DOI] [PubMed] [Google Scholar]
  • 22.Hiddemann W, Kneba M, Dreyling M, Schmitz N, Lengfelder E, Schmits R, et al. Frontline therapy with rituximab added to the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) significantly improves the outcome for patients with advanced-stage follicular lymphoma compared with therapy with CHOP alone: results of a prospective randomized study of the German Low-Grade Lymphoma Study Group. Blood. 2005;106:3725–3732. doi: 10.1182/blood-2005-01-0016. [DOI] [PubMed] [Google Scholar]
  • 23.Coiffier B, Lepage E, Briere J, Herbrecht R, Tilly H, Bouabdallah R, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. New Engl J Med. 2002;346:235–342. doi: 10.1056/NEJMoa011795. [DOI] [PubMed] [Google Scholar]
  • 24.Terriou L, Bonnet S, Debarri H, Demarquette H, Morschhauser F. Brentuximab vedotin: new treatment for CD30+ lymphomas. Bull Cancer. 2013;100:775–779. doi: 10.1684/bdc.2013.1778. In French. [DOI] [PubMed] [Google Scholar]
  • 25.Bodet-Milin C, Ferrer L, Pallardy A, Eugene T, Rauscher A, Faivre-Chauvet A, et al. Radioimmunotherapy of B-cell non-Hodgkin's lymphoma. Front Oncol. 2013;3:177–177. doi: 10.3389/fonc.2013.00177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hurvitz SA, Dirix L, Kocsis J, Bianchi GV, Lu J, Vinholes J, et al. Phase II randomized study of trastuzumab emtansine versus trastuzumab plus docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer. J Clin Oncol. 2013;31:1157–1163. doi: 10.1200/JCO.2012.44.9694. [DOI] [PubMed] [Google Scholar]
  • 27.Cui J, Cai X, Zhu M, Liu T, Zhao N. The efficacy of bevacizumab compared with other targeted drugs for patients with advanced NSCLC: a meta-analysis from 30 randomized controlled clinical trials. PLoS One. 2013;8: doi: 10.1371/journal.pone.0062038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gressett SM, Shah SR. Intricacies of bevacizumab-induced toxicities and their management. Ann Pharmacother. 2009;43:490–501. doi: 10.1345/aph.1L426. [DOI] [PubMed] [Google Scholar]
  • 29.Gibbs SD, Westerman DA, McCormack C, Seymour JF, Miles Prince H. Severe and prolonged myeloid haematopoietic toxicity with myelodysplastic features following alemtuzumab therapy in patients with peripheral T-cell lymphoproliferative disorders. Br J Haematol. 2005;130:87–91. doi: 10.1111/j.1365-2141.2005.05570.x. [DOI] [PubMed] [Google Scholar]
  • 30.van de Donk NW, Dhimolea E. Brentuximab vedotin. MAbs. 2012;4:458–465. doi: 10.4161/mabs.20230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ansell SM, Ristow KM, Habermann TM, Wiseman GA, Witzig TE. Subsequent chemotherapy regimens are well tolerated after radio-immunotherapy with yttrium-90 ibritumomab tiuxetan for non-Hodgkin's lymphoma. J Clin Oncol. 2002;20:3885–3890. doi: 10.1200/JCO.2002.10.143. [DOI] [PubMed] [Google Scholar]
  • 32.Craik DJ, Fairlie DP, Liras S, Price D. The future of peptide-based drugs. Chem Biol Drug Des. 2013;81:136–147. doi: 10.1111/cbdd.12055. [DOI] [PubMed] [Google Scholar]
  • 33.Farid SS. Process economics of industrial monoclonal antibody manufacture. J Chromatogr B Analyt Technol Biomed Life Sci. 2007;848:8–18. doi: 10.1016/j.jchromb.2006.07.037. [DOI] [PubMed] [Google Scholar]
  • 34.Leung HW, Chan AL, Leung MS, Lu CL. Systematic review and quality assessment of cost-effectiveness analysis of pharmaceutical therapies for advanced colorectal cancer. Ann Pharmacother. 2013;47:506–518. doi: 10.1345/aph.1R152. [DOI] [PubMed] [Google Scholar]
  • 35.Walsh G. Biopharmaceutical benchmarks 2010. Nat Biotechnol. 2010;28:917–924. doi: 10.1038/nbt0910-917. [DOI] [PubMed] [Google Scholar]
  • 36.Chow SC, Ju C. Assessing biosimilarity and interchangeability of biosimilar products under the Biologics Price Competition and Innovation Act. Generics and Biosimilars Initiative (GaBI) Journal. 2013;2:20–25. doi: 10.5639/gabij.2013.0201.004. [DOI] [Google Scholar]
  • 37.Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, et al. Trial watch: monoclonal antibodies in cancer therapy. Oncoimmunology. 2013;2: doi: 10.4161/onci.22789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wen F, Tang R, Sang Y, Li M, Hu Q, Du Z, et al. Which is false: oxaliplatin or fluoropyrimidine? An analysis of patients with KRAS wild-type metastatic colorectal cancer treated with first-line epidermal growth factor receptor monoclonal antibody. Cancer Sci. 2013;104:1330–1338. doi: 10.1111/cas.12224. Epub 2013 Jul 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rampino M, Bacigalupo A, Russi E, Schena M, Lastrucci L, Iotti C, et al. Efficacy and feasibility of induction chemotherapy and radiotherapy plus cetuximab in head and neck cancer. Anticancer Res. 2012;32:195–199. [PubMed] [Google Scholar]
  • 40.Poulin-Costello M, Azoulay L, Van Cutsem E, Peeters M, Siena S, Wolf M. An analysis of the treatment effect of panitumumab on overall survival from a phase 3, randomized, controlled, multicenter trial (20020408) in patients with chemotherapy refractory metastatic colorectal cancer. Target Oncol. 2013;8:127–136. doi: 10.1007/s11523-013-0271-z. Epub 2013 Apr 27. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The West Indian Medical Journal are provided here courtesy of The University of the West Indies

RESOURCES