WELCOME TO HEALTH WORLD!!!

Search 2.0


The generally accepted definition of health is "a state of complete physical, mental, and social well-being and not merely the absence of disease or infirmity"

Monday, June 8, 2009

Monoclonal antibody therapy

Each antibody binds only one specific antigen.

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, where a radioactive dose localizes on target cell line, delivering lethal chemical doses to the target.


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.

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).

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.

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.


Origins of monoclonal antibody therapy

Monoclonal antibodies for cancer. ADEPT, antibody directed enzyme prodrug therapy; ADCC, antibody dependent cell-mediated cytotoxicity; CDC, complement dependent cytotoxicity; MAb, monoclonal antibody; scFv, single-chain Fv fragment.

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. 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. 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. Initial therapeutic antibodies were simple murine analogues, which contributed to the early lack of success. 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. 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. 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 circumstances. 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.

Chimeric and humanized monoclonal antibodies

To reduce murine antibody immunogenicity, murine molecules were engineered to remove immunogenic content and to increase their immunologic efficiency. 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. Increases in antibody-antigen binding strength have been achieved by introducing mutations into the complementarity determining regions (CDR), 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.

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 desired antigen, leading to the production of monoclonal antibodies. Phage display libraries allow the transformation of murine antibodies in vitro into fully human antibodies.


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. 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


Antibody Brand name Approval date Type Target Approved treatment(s) Abciximab
ReoPro 1994 chimeric inhibition 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
Basiliximab
Simulect 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 Efalizumab
Raptiva 2002 humanized CD11a
Inflammatory diseases (psoriasis) Ibritumomab tiuxetan
Zevalin 2002 murine CD20
Non-Hodgkin lymphoma (with yttrium-90 or indium-111) Infliximab
Remicade 1998 chimeric inhibition of TNF-a signalling Inflammatory diseases (mostly auto-immune disorders) Muromonab-CD3
Orthoclone OKT3 1986 murine T cell CD3 Receptor Transplant rejection Natalizumab
Tysabri 2006 humanized T cell VLA4 receptor Inflammatory 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. However it holds 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.


Therapeutic Monoclonal Antibody Market Future

Since 2000, the therapeutic market for monoclonal antibodies has grown exponentially. The current “big 5” therapeutic antibodies on the market: Avastin, Herceptin (both oncology), Humira, Remicade (both Autoimmune and Infectious Disease ‘AIID’) and Rituxan (oncology and AIID) accounted for 80% of revenues in 2006.

In the immediate future, it is likely that Genentech/Roche will retain their control over the market (due to ownership of 3 of the “big 5” products), oncology and AIID will remain the mAb segment therapeutic focus (because these are the disease areas addressed by the big 5) and the three most commercially important ‘targets’ for the mAb class will be VEGF (Avastin), TNF-alpha (Remicade and Humira) and CD20 (Rituxan). Experts forecast that the therapeutic antibody market will continue to be dominated by Oncology and AIID segments (82-84 percent) from 2004 to 2011. Furthermore, experts note a potential for change in the balance between Oncology and AIID in the coming years. While Oncology therapeutics dominated the market in 2004, AIID is expected to dominate by 2011.

1 comment:

  1. Hello,

    This is the perfect blog for anyone who wants to know about this topic. Monoclonal antibody therapy is the use of monoclonal antibodies to specifically target cells. These proteins are designed to attach to areas on the surface of cancer cells and interfere with their growth and spread...

    pSIVA

    ReplyDelete

Powered By Blogger