My watch list
my.bionity.com  
Login  

Monoclonal antibody therapy



  Monoclonal antibody therapy is the use of monoclonal antibodies (or mAb) to specifically target cells. The main objective is stimulating the patient's immune system to attack the malignant tumor cells and the prevention of tumor growth by blocking specific cell receptors. Variations exist within this treatment, e.g.radioimmunotherapy a radioactive dose directly to the target cell, and lethal chemical doses to the target. [1]

Contents

Structure and function of human and therapeutic antibodies

Immunoglobulin G (IgG) antibodies are large heterodimeric molecules, approximately 150 kDa and are composed of two different kinds of polypeptide chain, called the heavy (~50kDa) and the light chain (~25kDa). There are two types of light chains, kappa (κ) and lambda (λ). By cleavage with enzyme papain, the Fab (fragment-antigen binding) part can be separated from the Fc (fragment crystalline) part of the molecule (see image). The Fab fragments contain the variable domains, which consist of three hypervariable amino acid domains responsible for the antibody specificity embedded into constant regions. There are four known IgG subclasses all of which are involved in Antibody-dependent cellular cytotoxicity.[2]

The immune system responds to the environmental factors it encounters on the basis of discrimination between self and non-self. Tumor cells are not specifically targeted by one's immune system since tumor cells are the patient's own cells. Tumor cells, however are highly abnormal, and many display unusual antigens that are either inappropriate for the cell type, its environment, or are only normally present during the organisms' development (e.g. fetal antigens).[2]

Other tumor cells display cell surface receptors that are rare or absent on the surfaces of healthy cells, and which are responsible for activating cellular signal transduction pathways that cause the unregulated growth and division of the tumor cell. Examples include ErbB2, a constitutively active cell surface receptor that is produced at abnormally high levels on the surface of approximately 30% of breast cancer tumor cells. Such breast cancer is known a HER2 positive breast cancer.[3]

Antibodies are a key component of the adaptive immune response, playing a central role in both in the recognition of foreign antigens and the stimulation of an immune response to them. The advent of monoclonal antibody technology has made it possible to raise antibodies against specific antigens presented on the surfaces of tumors.[3]

Origins of monoclonal antibody therapy

 

Immunotherapy developed as a technique with the discovery of the structure of antibodies and the development of hybridoma technology, which provided the first reliable source of monoclonal antibodies.[5] These advances allowed for the specific targeting of tumors both in vitro and in vivo. Initial research on malignant neoplasms found Mab therapy of limited and generally short-lived success with malignancies of the blood.[6][7] Furthermore treatment had to be specifically tailored to each individual patient, thus proving to be impracticable for the routine clinical setting.

Throughout the progression of monoclonal drug development there have been four major antibody types developed: murine, chimeric, humanised and human. That initial therapeutic antibodies were simple murine analogues, probably led to these failures. It has since been shown that these antibodies have; a short half-life in vivo (due to immune complex formation), limited penetration into tumour sites, and that they inadequately recruit host effector functions.[8]To overcome these difficulties the technical issues initially experienced had to be surpassed. Chimeric and humanized antibodies have generally replaced murine antibodies in modern therapeutic antibody applications. Hybridoma technology has been replaced by recombinant DNA technology, transgenic mice and phage display.[9] Understanding of proteomics has proven essential in identifying novel tumour targets.

Murine monoclonal antibodies

Initially, murine antibodies were obtained by hybridoma technology, for which Kohler and Milstein received a Nobel prize, however the dissimilarity between murine and human immune systems led to the clinical failure of these antibodies, except in some specific indications. Major problems associated with murine antibodies included reduced stimulation of cytotoxicity and the formation complexes after repeated administration, which resulted in mild allergic reactions and sometimes anaphylactic shock.[8]

Chimeric and humanized monoclonal antibodies

To reduce murine antibody immunogenicity, murine molecules were engineered to remove immunogenic content and to increase their immunologic efficiency.[8] This was initially achieved by the production of chimeric and humanized antibodies. Chimeric antibodies are composed of murine variable regions fused onto human constant regions. Human gene sequences, taken from the kappa light chain and the IgG1 heavy chain, results in antibodies that are approximately 65% human. This reduces immunogenicity, and thus increases serum half-life.

Humanised antibodies are produced by grafting murine hypervariable amino acid domains into human antibodies. This results in a molecule of approximately 95% human origin. However it has been shown in several studies that humanised antibodies bind antigen much more weakly than the parent murine monoclonal antibody, with reported decreases in affinity of up to several hundredfold.[10][11] Increases in antibody-antigen binding strength have been achieved by introducing mutations into the complementarity determining regions (CDR)[12], using techniques such as chain-shuffling, randomization of complementarity determining regions and generation of antibody libraries with mutations within the variable regions by error-prone PCR, E-coli mutator strains, and site-specific mutagenesis.[1]

Human monoclonal antibodies

Human monoclonal antibodies are produced using transgenic mice or phage display libraries. Human monoclonal antibodies are produced by transferring human immunoglobulin genes into the murine genome, after which the transgenic mouse is vaccinated against the immunoglobulin, leading to the production of monoclonal antibodies.[9] Phage display libraries allow the transformation of murine antibodies in vitro into fully human antibodies.[3]

FDA approved therapeutic antibodies

The first FDA-approved therapeutic monoclonal antibody was a murine IgG2a CD3 specific transplant rejection drug, Muromonab (OKT-3), in 1986. This drug found use in solid organ transplant recipients who became steroid resistant.[13] Currently, Twenty-one FDA-approved therapies exist, and hundreds of therapies are undergoing clinical trials. Most are concerned with immunological and oncological targets.

FDA approved monoclonal antibodies[1]
AntibodyBrand name Approval date Type Target Approved treatment(s)
Abciximab ReoPro 1994 chimericinhibition of glycoprotein IIb/IIIa Cardiovascular disease
Adalimumab Humira 2002 human inhibition of TNF-a signalling Inflammatory diseases (mostly auto-immune disorders)
Alemtuzumab Campath 2001 humanized CD52 Chronic lymphocytic leukemia
BasiliximabSimulect 1998 chimeric IL-2 receptor a Transplant rejection
Bevacizumab Avastin 2004 humanized vascular endothelial growth factor Colorectal cancer
Cetuximab Erbitux 2004 chimeric epidermal growth factor receptor Colorectal cancer
Daclizumab Zenapax 1997 humanized IL-2 receptor a Transplant rejection
Eculizumab Soliris 2007 humanized complement system protein C5 Inflammatory diseases including paroxysmal nocturnal hemoglobinuria
EfalizumabRaptiva 2002 humanized CD11a Inflammatory diseases (psoriasis)
Ibritumomab tiuxetan Zevalin 2002 murine CD20 Non-Hodgkin lymphoma (with yttrium-90 or indium-111)
InfliximabRemicade 1998 chimeric inhibition of TNF-a signallingInflammatory diseases (mostly auto-immune disorders)
Muromonab-CD3Orthoclone OKT3 1986 murine T cell CD3 Receptor Transplant rejection
Natalizumab Tysabri 2006 humanized T cell VLA4 receptorInflammatory diseases (mainly autoimmune-related multiple sclerosis therapy)
Omalizumab Xolair 2004 humanized immunoglobulin E (IgE) Inflammatory diseases (mainly allergy-related asthma therapy)
Palivizumab Synagis 1998 humanized an epitope of the F protein of RSV Viral infection (especially Respiratory Syncytial Virus (RSV)
Panitumumab Vectibix 2006 human epidermal growth factor receptor Colorectal cancer
Ranibizumab Lucentis 2006 humanized vascular endothelial growth factor Macular degeneration
Gemtuzumab ozogamicin Mylotarg 2000 humanized CD33 Acute myelogenous leukemia (with calicheamicin)
Rituximab Rituxan, Mabthera 1997 chimeric CD20 Non-Hodgkin lymphoma
Tositumomab Bexxar 2003 murine CD20 Non-Hodgkin lymphoma
Trastuzumab Herceptin 1998 humanized ErbB2 Breast cancer

Radioimmunotherapy

Radioimmunotherapy involves the use of radioactively conjugated murine antibodies against cellular antigens. Most research currently involved their application to lymphomas, as these are highly radio-sensitive malignancies. To limit radiation exposure, murine antibodies were especially chosen, as their high immunogenicity promotes rapid clearance from the body. Tositumomab is an exemplar used for non-Hodgkins lymphoma.

Antibody-directed enzyme prodrug therapy (ADEPT)

ADEPT involves the application of cancer associated monoclonal antibodies which are linked to a drug-activating enzyme. Subsequent systemic administration of a non-toxic agent results in its conversion to a toxic drug, and resulting in a cytotoxic effect which can be targeted at malignant cells. The clinical success of ADEPT treatments has been limited to date.[14] However it does hold great promise, and recent reports suggest that it will have a role in future oncological treatment.

Drug and gene therapy: Immuno-liposomes

Immunoliposomes are antibody-conjugated liposomes. Liposomes can carry drugs or therapeutic nucleotides and when conjugated with monoclonal antibodies, may be directed against malignant cells. Although this technique is still in its infancy, significant advances have been made. Immunoliposomes have been successfully used in vivo to achieve targeted delivery of tumour-suppressing genes into tumours, using an antibody fragment against the human transferrin receptor. Tissue-specific gene delivery using immunoliposomes has also been achieved in brain, and breast cancer tissue.[15]

References

  1. ^ a b c Waldmann, Thomas A. (2003). "Immunotherapy: past, present and future". Nature Medicine 9: 269–277.
  2. ^ a b Janeway, Charles; Paul Travers, Mark Walport, and Mark Shlomchik (2001). Immunobiology; Fifth Edition. New York and London: Garland Science. ISBN 0-8153-4101-6. .
  3. ^ a b c Janeway CA, Jr. et al (2005). Immunobiology., 6th ed., Garland Science. ISBN 0-443-07310-4. 
  4. ^ Modified from Carter P: Improving the efficacy of antibody-based cancer therapies. Nat Rev Cancer 2001;1:118-129
  5. ^ Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975; 256: 495-497
  6. ^ Nadler LM, Stashenko P, Hardy R, et al. Serotherapy of a patient with a monoclonal antibody directed against a human lymphoma associated antigen. Cancer Res 1980; 40: 3147-3154.
  7. ^ Ritz J, Schlossman SF. Utilization of monoclonal antibodies in the treatment of leukemia and lymphoma. Blood 1982; 59: 1-11.
  8. ^ a b c Stern M, Herrmann R. Overview of monoclonal antibodies in cancer therapy: present and promise. Crit Rev Oncol Hematol 2005; 54: 11-29.
  9. ^ a b Hudson PJ, Souriau C. Engineered antibodies. Nat Med 2003; 9: 129-134.
  10. ^ Carter P, Presta L, Gorman CM, et al. Humanization of an Anti-p185HER2 Antibody for Human Cancer Therapy. Proc Natl Acad Sci USA 1992; 89: 4285-4289.
  11. ^ Presta LG, Lahr SJ, Shields RL, et al. Humanization of an antibody directed against IgE. J Immunol 1993; 151: 2623-2632.
  12. ^ Chothia C, Lesk AM, Tramontano A, et al. Conformations of immunoglobulin hypervariable regions. Nature 1989; 342: 877-883.
  13. ^ Hooks MA, Wade CS, Millikan WJ. Muromonab CD-3: a review of its pharmacology, pharmacokinetics, and clinical use in transplantation. Pharmacotherapy 1991; 11: 26-37.
  14. ^ Francis RJ, Sharma SK, Springer C, et al. A phase I trial of antibody directed enzyme prodrug therapy (ADEPT) in patients with advanced colorectal carcinoma or other CEA producing tumours. Br J Cancer 2002; 87: 600-607.
  15. ^ Krauss WC, Park JW, Kirpotin DB, Hong K, Benz CC. Emerging antibody-based HER2 (ErbB-2/neu) therapeutics. Breast Dis 2000; 11: 113-124.

See also

 
This article is licensed under the GNU Free Documentation License. It uses material from the Wikipedia article "Monoclonal_antibody_therapy". A list of authors is available in Wikipedia.
Your browser is not current. Microsoft Internet Explorer 6.0 does not support some functions on Chemie.DE