Smaller Font Default Font Larger Font

You are here:

Research Resources: Therapeutic Antibody Discovery & Development

 

The potential clinical utility of antisera (a polyclonal mixture of antibodies) in the treatment of human diseases has been recognized for more than a century. However, the difficulty of isolating specific antibodies against the antigen from antisera in a reproducible manner was a major hurdle to their clinical exploitation. With the advent of murine monoclonal antibody technology, it seemed that the antibody’s therapeutic promise would be finally fulfilled. However, mouse monoclonal antibodies are foreign to human and provoke a strong immune response when administered to patients. The Human Anti-Mouse Antibody (HAMA) response limited both the dose and the number of times that could be repeated. Other important factors such as short serum half-life and poor recruitment of human immune effector functions also set back the clinical application of murine antibodies.

 

 

Therapeutic Antibody Development

 

The 1st attempt to engineer mouse antibodies to facilitate therapeutic use was chimerization. This involves genetically replacing mouse constant regions with the corresponding human constant regions, while retaining the mouse variable regions responsible for antigen binding. Antibody humanization (also known as CDR-grafting or reshaping) was invented as a more elegant solution to the immunogenicity problem of murine antibodies. Antibody humanization involves the design and synthesis of composite variable regions, which contain the amino acids of mouse CDRs integrated into the FWRs of a human antibody variant. The resulting antibody retains both the specificity and binding affinity of the original mouse antibody, and is sufficiently human to deceive the patient’s immune system. Both chimerization and humanization strategies have been proven successful in clinic.

 

 

A schematic diagram of mouse (top left), chimeric (top right), humanized (bottom left), and fully human (bottom right) antibodies are shown above. Human parts are shown in red, while non-human parts in blue. The International Nonproprietary Names (INN) recommends to name murine antibodies to end in “–omab”, chimeric antibodies to end in “-ximab”, humanized antibodies to end in “–zumab”, and fully human antibodies with end in “–umab”. 

 

There are ~300 monoclonal antibodies currently in clinic and on market for various therapeutic, diagnostic, and preventive applications. Among them,  about 40% (114) are humanized antibodies, 34% (99) are fully human antibodies, and 10% (30) are chimeric antibodies (see the pie chart below).

 

  • Examples of approved chimeric antibodies: Basiliximab(Simulect®), Cetuximab(Erbitux®), Infliximab (Remicade®), and Rituximab (Rituxan®)
  • Examples of approved humanized antibodies: Alemtuzumab (Campath®), Atlizumab (Actemra®), Bevacizumab (Avastin®), Daclizumab (Zanapax®), Natalizumab (Tysabri®), Omalizumab (Xolair®), Palivizumab (Synagis®), Pertuzumab (Omnitarg®), and Trastuzumab (Herceptin®)
  • Examples of approved fully human antibodies: Adalimumab (Humira®), Belimumab (Benlysta®), Denosumab (Prolia®), Ipilimumab (Yervoy®), Ofatumumab (Arzerra®), Panitumumab(Vectibix®), and Ustekinumab (Stelara®)

 

Click here to see "a complete list of monoclonal antibodies for clinical use" (that includes therapeutic, diagnostic and preventive monoclonal antibodies that are approved, investigational drugs, and those withdrawn from the market.)

 

 

 

Antibody Humanization & Engineering

 

The antibody humanization process usually includes the creation of a mouse-human chimera in an initial step.  Thereafter the chimera is further humanized by the selective alteration of the sequence of amino acids in the variable region of the molecule. The process must be "selective" to retain the specificity for which the antibody was originally developed. It normally involves the design and synthesis of composite variable regions which contain mouse CDRs integrated into human framework regions, whose role is to support CDRs in the same orientation as that of a corresponding human antibody variant. Ideally a humanized antibody should be essentially identical to that of a human variant, containing the only non-human origin of CDRs responsible for antigen binding. In reality the mouse sequences make up 5-10% of the humanized antibody (for more information and examples, please visit our "Antibody Services").

 

 

The feasibility of this process is, to a large extent, due to the inter-species conserved nature of antibody variable region genes between animals and humans. Within any species, variable region gene sequences can be grouped into a number of families according to amino acid or nucleotide sequence homology. In some cases inter-species homology can be higher than intra-species homology. For example, inter-species homologies can range from 40% to 80% between mouse and human variable region sequences. Moreover the highly conserved and well defined CDR loop structures seen in mouse antibodies are also observed across the species. It is thus the sequence and structural conservation throughout the variable regions of antibodies from different species that makes antibody humanization feasible.

 

 

Success Hallmarks of Therapeutic Antibody

 

Over the last decade, Biotechnology & Biopharmaceutical industry has concentrated on developing new technologies for antibody production and engineering with the aims to optimize or enhance its manufacturability and therapeutic efficacy. Several key “success hallmarks” have been proposed for an effective antibody-based therapeutic for human conditions such as cancers, inflammatory and infectious diseases:

 

• High affinity & specificity to target antigen
• Minimal immunogenicity and/or toxicity
• Efficient & selective recruitment of immune effectors
• Robust therapeutic intervening activity  

 

All approved antibody drugs are thought to work through a Fab-mediated or the combination of Fab- and Fc-mediated action.  However the proposed mechanisms are largely based upon data generated in vitro or in animal models. The clinical mechanisms of action of many approved antibody drugs are in actuality complicated and remain poorly understood.  In addition, antibodies frequently fail to activate ADCC or CDC and show little or no efficacy even with optimal binding to the target antigen or recruiting immune effectors.  This suggests that the clinical outcome is driven by a more complex interplay between the antibody, the cognate antigen that for example is expressed on stromal or tumor cells, and our immune system (see the figure below).  In addition there are many approaches to improving the clinical performance for antibody-based therapeutics, including antibody-drug conjugate (ADC) and bispecific antibody. 

 

 

Therapeutic Antibodies Discovery

 

There are technologies that completely avoid the use of mice or other animals in the discovery of antibodies for human therapeutic applications. Examples include various "display" methods that employ the selective principles of specific antibody production but exploit microorganisms (such as in phage display and yeast display) or even cell free extracts (as in ribosome display). These systems rely on the creation of antibody gene "libraries", which are usually derived from RNA of human peripheral blood. Each antibody gene is linked to a product (e.g., antibody fragment Fab or scFv) displayed by the system, allowing rapid screening for antigen-specific binders. Adalimumab (Humira) is an example of an antibody approved for human therapy that was created through phage display.

 

 

Antibody and Immunomodulation

 

The immune system has the intrinsic power to detect and eliminate abnormal cells, such as those derived from tumors.  This process, commonly referred to as immune surveillance, takes advantage of numerous biological features that distinguish tumor cells from their normal counterparts.  For example, tumor cells display aberrant functional behaviors and an altered surface antigen composition, typically resulting from a myriad of genetic and epigenetic changes. Abnormal cytokine and growth factor expression patterns are also common hallmarks of certain types of cancer, eliciting to either support growth or counteract local inflammation, particularly during cellular invasion and metastasis. With more advanced disease, tumor cells eventually develop active mechanisms to escape immune surveillance and induce tolerance.  There is ample evidence that B cell-driven antibody responses can trigger autologous tumor regression in animals and humans. 

 

Approved Antibody-based Therapeutics for Cancer and Target Antigens. 

The antigens are in black while the approved antibody therapeutics are in red.

See the list of "monoclonal antibodies for clinical use" for more details and examples.

 

 

 

Therapeutic Antibodies from Humans

 

It is possible to exploit human immune response in the discovery of truly human antibodies for therapeutic applications. Human immune response works essentially in the same way as that in a mouse. Therefore, persons experiencing a challenge to their immune system, such as an infectious virus, a passive vaccination, or abnormal tumor cells are a potential source of discovering antibodies directed against that challenge. This approach seems especially useful for the development of anti-viral and anti-cancer (of particular types) therapies that exploit the principles of passive immunity. Variants of this approach have been demonstrated with proof-of-principle in preclinical studies and several are finding way into clinical development.

 

 

Antibody R&D Capability at G&P Biosciences

 

We offer a range of antibody production and engineering services that may complement your research and accelerate the progress of development towards clinical use. Our services are offered as stand-alone services and also as part of a complete suite of antibody custom solution package. We offer a proprietary antibody humanization service. Using our technology, the sequences of the antibody variable domains, which determine its binding specificity, are incorporated into human donor sequences properly, thus creating a panel of humanized antibodies for expression. We also provide bundled services, starting from antigen preparation, hybridoma screening, recombinant antibody generation, affinity determination, antibody humanization and engineering to custom-scale production. We can drive your antibody R&D from any stage to the delivery of a 100% royalty free drug candidate that can be moved into clinical development rapidly (please visit our "Antibody Services" to learn more and request a quote).

 

 

 

 

«  Return to the previous topic: Antibody Structure, Function and Production

 

«  Go back to Technical Support main page

 

Research Resources: Antibody

 

The International Nonproprietary Names (INN) recommends to name murine antibodies to end in “–omab”, chimeric antibodies to end in “-ximab”, humanized antibodies to end in “–zumab”, and fully human antibodies with end in “–umab”. The list includes therapeutic, diagnostic, and preventive monoclonal antibodies that are approved, investigational drugs, and drugs that have been withdrawn from the market. 

 

List of Monoclonal Antibodies for Clinical Use

 
Name Trade Name Type Source Target Antigen Clinical Use
Abagovomab   mab mouse CA-125 (imitation) ovarian cancer
Abciximab ReoPro® Fab chimeric CD41 (integrin α-IIb) platelet aggregation inhibitor
Actoxumab   mab human Clostridium difficile Clostridium difficile infection
Adalimumab Humira® mab human TNF-α rheumatoid arthritis, Crohn's Disease, Plaque Psoriasis, Psoriatic Arthritis, Ankylosing Spondylitis, Juvenile Idiopathic Arthritis
Adecatumumab   mab human EpCAM prostate and breast cancer
Ado-trastuzumab emtansine Kadcyla®

mab (ADC)

humanized HER2/neu breast cancer
Afelimomab   F(ab')2 mouse TNF-α sepsis
Afutuzumab   mab humanized CD20 lymphoma
Alacizumab pegol   F(ab')2 humanized VEGFR2 cancer
Alemtuzumab Campath-1H®, MabCampath® mab humanized CD52 CLL, CTCL
Alirocumab   mab human NARP-1 hypercholesterolemia
Altumomab pentetate Hybri-ceaker® mab mouse CEA colorectal cancer (diagnosis)
Amatuximab   mab chimeric Mesothelin cancer
Anatumomab mafenatox   Fab mouse TAG-72 non-small cell lung carcinoma
Anifrolumab   mab human Interferon α/β receptor systemic lupus erythematosus
Anrukinzumab   mab humanized IL-13  ?
Apolizumab   mab humanized HLA-DR hematological cancers
Arcitumomab CEA-Scan® Fab' mouse CEA gastrointestinal cancers (diagnosis)
Aselizumab   mab humanized L-selectin (CD62L) severely injured patients
Atezolizumab Tecentriq® mab humanized PD-L1 lung cancer, bladder cancer
Atinumab   mab human RTN4  ?
Atlizumab (Tocilizumab) Actemra®, RoActemra® mab humanized IL-6 receptor rheumatoid arthritis
Atorolimumab   mab human Rhesus factor hemolytic disease of the newborn
Bapineuzumab   mab humanized β-amyloid Alzheimer's disease
Basiliximab Simulect® mab chimeric CD25 prevention of organ transplant rejections
Bavituximab   mab chimeric Phosphatidylserine cancer, viral infections
Bectumomab LymphoScan® Fab' mouse CD22 non-Hodgkin's lymphoma (detection)
Belimumab Benlysta®, LymphoStat-B mab human BAFF SLE, non-Hodgkin lymphoma
Benralizumab   mab humanized CD125 asthma
Bertilimumab   mab human CCL11 (eotaxin-1) severe allergic disorders
Besilesomab Scintimun® mab mouse CEA-related antigen inflammatory lesions and metastases (detection)
Bevacizumab Avastin® mab humanized VEGF-A metastatic cancer
Bezlotoxumab   mab human Clostridium difficile Clostridium difficile infection
Biciromab FibriScint® Fab' mouse Fibrin II, β chain thromboembolism (diagnosis)
Bimagrumab   mab human ACVRIIB myostatin inhibitor
Bivatuzumab mertansine   mab humanized CD44 v6 squamous cell carcinoma
Blinatumomab  Blincyto®  scFv (BiTE) mouse CD19, CD3 cancer
Blosozumab   mab humanized SOST osteoporosis
Bococizumab   mab humanized PCSK9 hypocholesterolemia
Brentuximab vedotin  Adcetris® mab (ADC) chimeric CD30 (TNFRSF8) hematologic cancers (Hodgkin lymphoma)
Briakinumab   mab human IL-12, IL-23 psoriasis, rheumatoid arthritis, inflammatory bowel diseases, multiple sclerosis
Brodalumab   mab human IL-17 inflammatory diseases
Canakinumab Ilaris® mab human IL-1 rheumatoid arthritis
Cantuzumab mertansine   mab humanized Mucin CanAg colorectal cancer etc.
Cantuzumab ravtansine   mab humanized MUC1 cancers
Caplacizumab   mab humanized VWF  ?
Capromab pendetide Prostascint® mab mouse PMSA prostate cancer (detection)
Carlumab   mab human MCP-1 oncology/immune indications
Catumaxomab Removab® 3funct rat/mouse hybrid EpCAM, CD3 ovarian cancer, malignant ascites, gastric cancer
Cedelizumab   mab humanized CD4 prevention of organ transplant rejections, treatment of autoimmune diseases
Certolizumab pegol Cimzia® Fab' humanized TNF-α Crohn's disease
Cetuximab Erbitux® mab chimeric EGFR metastatic colorectal cancer and head and neck cancer
Citatuzumab bogatox   Fab humanized EpCAM ovarian cancer and other solid tumors
Cixutumumab   mab human IGF1R solid tumors
Clazakizumab   mab humanized Oryctolagus cuniculus rheumatoid arthritis
Clenoliximab   mab chimeric CD4 rheumatoid arthritis
Clivatuzumab tetraxetan hPAM4-Cide mab humanized MUC1 pancreatic cancer
Conatumumab   mab human TRAIL-R2 cancer
Concizumab   mab humanized TFPI bleeding
Crenezumab   mab humanized 1-40-β-amyloid Alzheimer's disease
Dacetuzumab   mab humanized CD40 hematologic cancers
Daclizumab Zenapax® mab humanized CD25 prevention of organ transplant rejections
Dalotuzumab   mab humanized IGF1R cancer etc.
Daratumumab Darzalex® mab human CD38 multiple myeloma
Demcizumab   mab humanized DLL4 cancer
Denosumab Prolia®/Xgeva® mab human RANKL osteoporosis, bone metastases etc.
Detumomab   mab mouse B-lymphoma cell lymphoma
Dorlimomab aritox   F(ab')2 mouse  ?  ?
Drozitumab   mab human DR5 cancer etc.
Duligotumab   mab human HER3  ?
Dupilumab   mab human IL4 atopic diseases
Dusigitumab   mab human ILGF2 cancer
Ecromeximab   mab chimeric GD3 ganglioside malignant melanoma
Eculizumab Soliris® mab humanized C5 paroxysmal nocturnal hemoglobinuria
Edobacomab   mab mouse Endotoxin sepsis caused by Gram-negative bacteria
Edrecolomab Panorex® mab mouse EpCAM colorectal carcinoma
Efalizumab Raptiva® mab humanized LFA-1 (CD11a) psoriasis (blocks T-cell migration)
Efungumab Mycograb® scFv human Hsp90 invasive Candida infection
Eldelumab   mab human Interferon γ-induced protein Crohn's disease, ulcerative colitis
Elotuzumab Elmplicit® mab humanized SLAMF7 multiple myeloma
Elsilimomab   mab mouse IL-6  ?
Enavatuzumab   mab humanized TWEAK receptor cancer etc.
Enlimomab pegol   mab mouse ICAM-1 (CD54)  ?
Enokizumab   mab humanized IL9 asthma
Enoticumab   mab human DLL4  ?
Ensituximab   mab chimeric 5AC cancer
Epitumomab cituxetan   mab mouse Episialin  ?
Epratuzumab   mab humanized CD22 cancer, SLE
Erlizumab   F(ab')2 humanized ITGB2 (CD18) heart attack, stroke, traumatic shock
Ertumaxomab Rexomun® 3funct rat/mouse hybrid HER2/neu, CD3 breast cancer etc.
Etaracizumab Abegrin® mab humanized Integrin αvβ3 melanoma, prostate cancer, ovarian cancer etc.
Etrolizumab   mab humanized Integrin α7 β7 inflammatory bowel disease
Evolocumab   mab human PCSK9 hypocholesterolemia
Exbivirumab   mab human Hepatitis B surface antigen hepatitis B
Fanolesomab NeutroSpec® mab mouse CD15 appendicitis (diagnosis)
Faralimomab   mab mouse Interferon receptor  ?
Farletuzumab   mab humanized Folate receptor 1 ovarian cancer
Fasinumab   mab human HNGF  ?
Felvizumab   mab humanized Respiratory syncytial virus respiratory syncytial virus infection
Fezakinumab   mab human IL-22 rheumatoid arthritis, psoriasis
Ficlatuzumab   mab humanized HGF cancer etc.
Figitumumab   mab human IGF-1 receptor adrenocortical carcinoma, non-small cell lung carcinoma etc.
Flanvotumab   mab human Glycoprotein 75 melanoma
Fontolizumab HuZAF® mab humanized IFN-γ Crohn's disease etc.
Foralumab   mab human CD3ε  ?
Foravirumab   mab human Rabies virus glycoprotein rabies (prophylaxis)
Fresolimumab   mab human TGF-β idiopathic pulmonary fibrosis, focal segmental glomerulosclerosis, cancer
Fulranumab   mab human NGF pain
Futuximab   mab chimeric EGFR  ?
Galiximab   mab chimeric CD80 B-cell lymphoma
Ganitumab   mab human IGF-I cancer
Gantenerumab   mab human β-amyloid Alzheimer's disease
Gavilimomab   mab mouse CD147 (basigin) graft versus host disease
Gemtuzumab ozogamicin Mylotarg® mab (ADC) humanized CD33 acute myelogenous leukemia (withdrawn from the market)
Gevokizumab   mab humanized IL-1β diabetes etc.
Girentuximab Rencarex® mab chimeric Carbonic anhydrase 9 (CA-IX) clear cell renal cell carcinoma
Glembatumumab vedotin   mab human GPNMB melanoma, breast cancer
Golimumab Simponi® mab human TNF-α rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis
Gomiliximab   mab chimeric CD23 (IgE receptor) allergic asthma
Guselkumab   mab human IL23p19 psoriasis
Ibalizumab   mab humanized CD4 HIV infection
Ibritumomab tiuxetan Zevalin® mab mouse CD20 non-Hodgkin's lymphoma
Icrucumab   mab human VEGFR-1 cancer etc.
Igovomab Indimacis-125® F(ab')2 mouse CA-125 ovarian cancer (diagnosis)
Imciromab Myoscint® mab mouse Cardiac myosin cardiac imaging
Imgatuzumab   mab humanized EGFR cancer
Inclacumab   mab human Selectin P  ?
Indatuximab ravtansine   mab chimeric SDC1 cancer
Infliximab Remicade® mab chimeric TNF-α rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, psoriasis, Crohn's disease, ulcerative colitis
Intetumumab   mab human CD51 solid tumors (prostate cancer, melanoma)
Inolimomab   mab mouse CD25 graft versus host disease
Inotuzumab ozogamicin   mab humanized CD22 cancer
Ipilimumab Yervoy® mab human CD152 melanoma
Iratumumab   mab human CD30 (TNFRSF8) Hodgkin's lymphoma
Itolizumab   mab humanized CD6  ?
Ixekizumab   mab humanized IL-17A autoimmune diseases
Keliximab   mab chimeric CD4 chronic asthma
Labetuzumab CEA-Cide® mab humanized CEA colorectal cancer
Lambrolizumab (Pembrolizumab) Keytruda® mab humanized PD-1 antineoplastic agent
Lampalizumab   mab humanized CFD  ?
Lebrikizumab   mab humanized IL-13 asthma
Lemalesomab   mab mouse NCA-90 (granulocyte antigen) diagnostic agent
Lerdelimumab   mab human TGFβ2 reduction of scarring after glaucoma surgery
Lexatumumab   mab human TRAIL-R2 cancer
Libivirumab   mab human Hepatitis B surface antigen hepatitis B
Ligelizumab   mab humanized IGHE  ?
Lintuzumab   mab humanized CD33 cancer
Lirilumab   mab human KIR2D  ?
Lodelcizumab   mab humanized PCSK9 hypercholesterolemia
Lorvotuzumab mertansine   mab humanized CD56 cancer
Lucatumumab   mab human CD40 multiple myeloma, non-Hodgkin's lymphoma, Hodgkin's lymphoma
Lumiliximab   mab chimeric CD23 (IgE receptor) chronic lymphocytic leukemia
Mapatumumab   mab human TRAIL-R1 cancer
Margetuximab   mab humanized ch4D5 cancer
Maslimomab    ? mouse T-cell receptor  ?
Mavrilimumab   mab human GMCSF receptor α-chain rheumatoid arthritis
Matuzumab   mab humanized EGFR colorectal, lung and stomach cancer
Mepolizumab Bosatria® mab humanized IL-5 asthma and white blood cell diseases
Metelimumab   mab human TGF beta 1 systemic scleroderma
Milatuzumab   mab humanized CD74 multiple myeloma and other hematological malignancies
Minretumomab   mab mouse TAG-72  ?
Mitumomab   mab mouse GD3 ganglioside small cell lung carcinoma
Mogamulizumab   mab humanized CCR4 cancer
Morolimumab   mab human Rhesus factor  ?
Motavizumab Numax® mab humanized Respiratory syncytial virus respiratory syncytial virus (prevention)
Moxetumomab pasudotox   mab mouse CD22 cancer
Muromonab-CD3 Orthoclone OKT3 mab mouse CD3 prevention of organ transplant rejections
Nacolomab tafenatox   Fab mouse C242 antigen colorectal cancer
Namilumab   mab human CSF2  ?
Naptumomab estafenatox   Fab mouse 5T4 non-small cell lung carcinoma, renal cell carcinoma
Narnatumab   mab human RON cancer
Natalizumab Tysabri® mab humanized Integrin α4 multiple sclerosis, Crohn's disease
Nebacumab   mab human Endotoxin sepsis
Necitumumab Portrazza® mab human EGFR non-small cell lung carcinoma
Nerelimomab   mab mouse TNF-α  ?
Nesvacumab   mab human Angiopoietin 2 cancer
Nimotuzumab Theracim®, Theraloc® mab humanized EGFR squamous cell carcinoma, head and neck cancer, nasopharyngeal cancer, glioma
Nivolumab Opdivo® mab human PD-1 melanoma, lung cancer, kidney cancer
Nofetumomab merpentan Verluma® Fab mouse  ? cancer (diagnosis)
Obinutuzumab Gazyva® mab humanized CD20 B-CLL, lymphoma
Ocaratuzumab   mab humanized CD20 cancer
Ocrelizumab   mab humanized CD20 rheumatoid arthritis, lupus erythematosus etc.
Odulimomab   mab mouse LFA-1 (CD11a) prevention of organ transplant rejections, immunological diseases
Ofatumumab Arzerra® mab human CD20 chronic lymphocytic leukemia etc.
Olaratumab Lartruvo® mab human PDGF-Rα cancer
Olokizumab   mab humanized IL6  ?
Omalizumab Xolair® mab humanized IgE Fc region allergic asthma
Onartuzumab   mab humanized (monovalent) MET, c-Met, HGF/SF-R cancer
Ontuxizumab   mab chimeric/humanized TEM1 cancer
Oportuzumab monatox   scFv humanized EpCAM cancer
Oregovomab OvaRex® mab mouse CA-125 ovarian cancer
Orticumab   mab human oxLDL  ?
Otelixizumab   mab chimeric/humanized CD3 diabetes mellitus type 1
Oxelumab   mab human OX-40 asthma
Ozanezumab   mab humanized NOGO-A ALS and multiple sclerosis
Ozoralizumab   mab humanized TNF-α inflammation
Pagibaximab   mab chimeric Lipoteichoic acid sepsis (Staphylococcus)
Palivizumab Synagis®, Abbosynagis® mab humanized F protein of respiratory syncytial virus respiratory syncytial virus (prevention)
Panitumumab Vectibix® mab human EGFR colorectal cancer
Panobacumab   mab human Pseudomonas aeruginosa Pseudomonas aeruginosa infection
Parsatuzumab   mab human EGFL7 cancer
Pascolizumab   mab humanized IL-4 asthma
Pateclizumab   mab humanized LTA ?
Patritumab   mab human HER3 cancer
Pembrolizumab Keytruda® mab humanized PD-1 Melanoma, lung cancer
Pemtumomab Theragyn®  ? mouse MUC1 cancer
Perakizumab   mab humanized IL17A arthritis
Pertuzumab Omnitarg/Perjeta® mab humanized HER2/neu cancer
Pexelizumab   scFv humanized C5 reduction of side effects of cardiac surgery
Pidilizumab   mab humanized PD-1 cancer and infectious diseases
Pinatuzumab vedotin   mab humanized CD22 cancer
Pintumomab   mab mouse Adenocarcinoma antigen adenocarcinoma (imaging)
Placulumab   mab human TNF  ?
Polatuzumab vedotin   mab humanized CD79B  ?
Ponezumab   mab humanized β-amyloid Alzheimer's disease
Priliximab   mab chimeric CD4 Crohn's disease, multiple sclerosis
Pritoxaximab   mab chimeric E. coli shiga toxin type-1  ?
Pritumumab   mab human Vimentin brain cancer
Quilizumab   mab humanized IGHE  ?
Racotumomab   mab mouse N-glycolylneuraminic acid cancer
Radretumab   mab human Fibronectin extra domain-B cancer
Rafivirumab   mab human Rabies virus glycoprotein rabies (prophylaxis)
Ramucirumab Cyramza® mab human VEGFR2 solid tumors (NSCLC, gastric)
Ranibizumab Lucentis® Fab humanized VEGF-A macular degeneration (wet form)
Raxibacumab   mab human Anthrax toxin, protective antigen anthrax (prophylaxis and treatment)
Regavirumab   mab human Cytomegalovirus glycoprotein B cytomegalovirus infection
Reslizumab   mab humanized IL-5 inflammations of the airways, skin and gastrointestinal tract
Rilotumumab   mab human HGF solid tumors
Rituximab MabThera, Rituxan® mab chimeric CD20 lymphomas, leukemias, some autoimmune disorders
Robatumumab   mab human IGF-1 receptor cancer
Roledumab   mab human RHD  ?
Romosozumab   mab humanized Scleroscin osteoporosis
Rontalizumab   mab humanized IFN-α systemic lupus erythematosus
Rovelizumab LeukArrest® mab humanized CD11, CD18 haemorrhagic shock etc.
Ruplizumab Antova® mab humanized CD154 (CD40L) rheumatic diseases
Samalizumab   mab humanized CD200 cancer
Sarilumab   mab human IL6 rheumatoid arthritis, ankylosing spondylitis
Satumomab pendetide   mab mouse TAG-72 cancer (diagnosis)
Secukinumab   mab human IL-17A uveitis, rheumatoid arthritis psoriasis
Seribantumab   mab human ERBB3, HER3 cancer
Setoxaximab   mab chimeric E. coli shiga toxin type-1  ?
Sevirumab    ? human Cytomegalovirus cytomegalovirus infection
Sibrotuzumab   mab humanized FAP cancer
Sifalimumab   mab humanized IFN-α SLE, dermatomyositis, polymyositis
Siltuximab Sylvant® mab chimeric IL-6 Multicentric Castleman's disease, cancer
Simtuzumab   mab humanized LOXL2  ?
Siplizumab   mab humanized CD2 psoriasis, graft-versus-host disease (prevention)
Sirukumab   mab human IL-6 rheumatoid arthritis
Solanezumab   mab humanized β-amyloid Alzheimer's disease
Solitomab   mab mouse EpCAM  ?
Sonepcizumab    ? humanized Sphingosine-1-phosphate choroidal and retinal neovascularization
Sontuzumab   mab humanized Episialin  ?
Stamulumab   mab human Myostatin muscular dystrophy
Sulesomab LeukoScan® Fab' mouse NCA-90 (granulocyte antigen) osteomyelitis (imaging)
Suvizumab   mab humanized HIV-1 viral infections
Tabalumab   mab human BAFF B-cell cancers
Tacatuzumab tetraxetan AFP-Cide® mab humanized α-fetoprotein cancer
Tadocizumab   Fab humanized Integrin αIIbβ3 percutaneous coronary intervention
Talizumab   mab humanized IgE allergic reaction
Tanezumab   mab humanized NGF pain
Taplitumomab paptox   mab mouse CD19 cancer[citation needed]
Tefibazumab Aurexis® mab humanized Clumping factor A Staphylococcus aureus infection
Telimomab aritox   Fab mouse  ?  ?
Tenatumomab   mab mouse Tenascin C cancer
Teneliximab   mab chimeric CD40  ?
Teplizumab   mab humanized CD3 diabetes mellitus type 1
Teprotumumab   mab human CD221 hematologic tumors
Ticilimumab (Tremelimumab)   mab human CTLA-4 cancer
Tildrakizumab   mab humanized IL23 immunologically mediated inflammatory disorders
Tigatuzumab   mab humanized TRAIL-R2 cancer
Tocilizumab (Atlizumab) Actemra®, RoActemra® mab humanized IL-6 receptor rheumatoid arthritis
Toralizumab   mab humanized CD154 (CD40L) rheumatoid arthritis, lupus nephritis etc.
Tositumomab Bexxar® mab mouse CD20 follicular lymphoma
Tovetumab   mab human CD140a cancer
Tralokinumab   mab human IL-13 asthma etc.
Trastuzumab Herceptin® mab humanized HER2/neu breast cancer
Tregalizumab   mab humanized CD4  ?
Tremelimumab   mab human CTLA-4 cancer
Tucotuzumab celmoleukin   mab humanized EpCAM cancer
Tuvirumab    ? human Hepatitis B virus chronic hepatitis B
Ublituximab   mab chimeric MS4A1 cancer
Urelumab   mab human 4-1BB cancer etc.
Urtoxazumab   mab humanized Escherichia coli diarrhoea caused by E. coli
Ustekinumab Stelara® mab human IL-12, IL-23 multiple sclerosis, psoriasis, psoriatic arthritis
Vantictumab   mab human Frizzled receptor cancer
Vapaliximab   mab chimeric AOC3 (VAP-1)  ?
Vatelizumab   mab humanized ITGA2  ?
Vedolizumab  Entyvio®
mab humanized Integrin α4β7 Crohn's disease, ulcerative colitis
Veltuzumab   mab humanized CD20 non-Hodgkin's lymphoma
Vepalimomab   mab mouse AOC3 (VAP-1) inflammation
Vesencumab   mab human NRP1  ?
Visilizumab Nuvion® mab humanized CD3 Crohn's disease, ulcerative colitis
Volociximab   mab chimeric Integrin α5β1 solid tumors
Vorsetuzumab mafodotin   mab humanized CD70 cancer
Votumumab HumaSPECT® mab human Tumor antigen CTAA16.88 colorectal tumors
Zalutumumab HuMax-EGFr mab human EGFR squamous cell carcinoma of the head and neck
Zanolimumab HuMax-CD4 mab human CD4 rheumatoid arthritis, psoriasis, T-cell lymphoma
Zatuximab   mab chimeric EGFR, HER1 cancer
Ziralimumab   mab human CD147 (basigin)  ?
Zolimomab aritox   mab mouse CD5 systemic lupus erythematosus, graft-versus-host disease

 


» Return to the previous topic: Therapeutic Antibodu Discovery & Development

 

» Return to Technical Support Main Page

Research Resources: Antibody Structure, Function and Production

 

Antibodies or immunoglobulins are a group of structurally & functionally similar glycoproteins that confer humoral immunity in humans and animals. In 1975, Köhler and Milstein developed mouse hybridoma technology to immortalize individual B cell clones producing a single (monoclonal) antibody. Since then, antibody structure and function have been studied extensively. The antibody backbone typically consists of two identical heavy chains and two identical light chains. Five antibody classes or isotypes (IgG, IgA, IgM, IgD, IgE) are recognized in mice and humans on the basis of different constant regions in the heavy chains. In the following sections, we will focus on the IgG subclasses (using human IgGs as examples), which are widely utilized for research, diagnostic and therapeutic applications.

 

 

Antibody Structure and Fucntion

 

Human IgG is a tetrameric protein comprising two identical 50-kDa heavy chains and two identical 25-kDa light chains. Each light chain is covalently linked to the N-terminal region of one heavy chain, while the two heavy chains associate covalently via disulfide bridges located in the hinge region, endowing an IgG with a characteristic Y-shaped structure. In addition intra-chain disulfide bonds are responsible for the formation of loops, leading to the compact, discrete folding of Ig domain-like structure of ~110 amino acids (see the domain structure of human IgG1 below).

 

Each light chain or heavy contains a variable region (VL, VH) and one or three constant regions (CL, CH1-3).  The amino acid sequences of these N-terminal regions are much more variable than the constant regions, which make up the rest of the IgG molecule. The variable regions and first constant region form the so-called fragment for antigen binding (Fab), while the remainder of the molecule constitutes the fragment crystalline (Fc), a region displaying little subclass variability. 

 

IgG is a truly “bifunctional” molecule, working through the coordinated actions of their two arms, Fab (or Fv) and Fc:

 

Fab, through specific antigen binding, usually neutralizes or antagonizes the biological activities of the target antigen. 
 
Fc, through interacting with Fcγ receptors (FcγRs) or the 1st complement component C1q, recruits and activates the immune effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), ultimately causing lysis of target cells. 

 

 

 

 

Antibody Variable Regions

 

The coding sequences for the variable regions are assembled from a number of mini-gene families (V and J for the light chain; V, D and J for the heavy chain), which are present in multiple variant copies in our genome. The DNA sequence of these mini-genes is modified further during the recombination and hypermutation events ("somatic hypermutation"), which occur during the development of antibody-producing B cells. The combination of heavy and light chain variable regions (VH+VL) at each N-terminal arm of a whole IgG is also known as Fv.

 

Within each variable region, there are three non-contiguous regions, which are exceptionally variable and thus referred to as "complementarity determining regions" (CDRs). CDRs provide the repertoire of complementary surfaces for recognizing different antigens or epitopes. The binding affinity is essentially mediated via the six CDRs, which fold as independent loops and together form the large surface patch making direct contact with the antigen. The variable region residues that are not part of the CDR's constitute the "framework" regions (FWRs) and generally do not interact with the antigen.  However certain residues in FWRS are key to positioning CDRs and therefore contribute to the binding affinity and specificity of an antibody.

 

 

Human IgG Subclasses & Properties

 

There are four subclasses of human IgG (IgG1-4), which can be distinguished by their heavy chain’s constant regions. The primary sequences of these constant regions are >95% homologous. Major differences are found in the hinge region in terms of the numbers of residues and interchain disulfide bonds (see the table below). The hinge region is the most diverse structural feature of different IgGs. It connects two heavy chains through disulfide bonds in the middle. It also links the two Fab arms to the Fc portion and provides flexibilities to the IgG molecules. The flexibility is important for the Fab arm to interact with differently spaced epitopes and for Fc to adopt different conformations to induce immune effector functions. 

 

 

Human IgG Subclass

IgG1

IgG2

IgG3

IgG4

Heavy Chain (HC)

γ1

γ2

γ3

γ4

Light Chain (LC)

κ, λ

κ, λ

κ, λ

κ, λ

Light Chain (κ:λ) Ratio

2.4

1.1

1.4

8.0

Calculated M.W. (kDa)

146

146

170

146

Functional Valency

2

2

2

2

Hinge Region Amino Acid#

15

12

62

12

Interchain Disulfide Bonds#

2

4

11

2

pI range (mean±SD)

8.6±0.4

7.4±0.6

8.3±0.7

7.2±0.8

Average Serum Conc. (mg/ml)

8

4

0.8

0.4

Circulating B Cell Distribution (%)

40

48

8

1

Plasma Cell Distribution (%)

64

26

8

1

Half-Life (days)

21-23

20-23

7-8

21-23

Complement Fixation (classic)

++

+

++

-

ADCC Activation

++

+/-

++

-

Binding to Protein A

++

++

+/-

++

Binding to Protein G

++

++

++

++

 

 

There are two types of light chains termed kappa (κ) and lambda (λ) chains.  The ratio of kappa and lambda light chain varies from species to species (e.g., 20:1 in mice vs. 2:1 in humans) and is also a characteristic of different IgG subclass (e.g., 2.4:1 for human IgG1 vs. 8:1 for IgG4). In addition, the point of light chain attachment to the heavy chain also differ among the IgG subclasses. For example, IgG1 light chain and heavy chain are bound through a disulfide bond near the midpoint of heavy chain (i.e., near the end of CH1).  In contrast, IgG2, IgG3 and IgG4 are joined at one quarter the distance from the heavy chain amino termini (i.e., near the end of VH). 

 

Different human IgG subclasses have varying abilities to activate immune effector functions. For example, the Fc portion of human IgG1 and IgG3 (but nor IgG2 or IgG4) is capable of binding to C1q, leading to the activation of the classical complement (CDC) cascade. IgG1 and IgG3 can also bind Fcg receptors (FcgRs) on immune effector cells, such as natural killer (NK) cells and macrophages, and recruit them to induce strong ADCC.  Over the last decade, many technologies have emerged to improve antibody-directed immune effector functions. They include altering the glycosylation pattern or sequence of the Fc region with the aim to enhance its binding to C1q or FcgRs, and constructing bispecific antibodies or fragments (e.g., BiTE) that engage target cells and immune effector components simultaneously (see the text and graph below).

 

 

Mouse Antibody Classes and Subclasses

 

Ther are five Ig classes or isotypes (IgA, IgD, IgE, IgG, and IgM) from mice, same as humans. Each isotype has a different heavy chain. The mouse IgG subclasses include IgG1, IgG2a, IgG2b, IgG2c, and IgG3. For IgG2a and IgG2c, however, inbred mouse strains with the Igh1-b allele have IgG2c instead of IgG2a. The murine heavy chain locus has only one of these two subclass genes in addition to the others. Like human, mouse IgG subclasses are very important in immune effector function. For example, mouse IgGs display remarkable differences in anti-bacterial responses (IgG3 >> IgG2b > IgG2a >> IgG1) and opsonophagocytic activities (IgG3 > IgG2b = IgG2a >> IgG1).

 

 

Antibody Applications

 

Conventional antibodies have been utilized in research for protein detection through Western blot, immunohistochemistry (IHC) and enzyme-linked immunosorbent assays (ELISA) for decades. Antibodies have also been developed for diagnostic applications such as pregnancy tests and detection of the viruses in the blood, such as an ELISA that detects HIV. Moreover, antibodies are used commonly in therapeutic applications. For example, Infliximab (Humira) is a human antibody that recognizes tumor necrosis factor alpha (TNFα) and is used in the treatment of Crohn's disease and rheumatoid arthritis. Trastuzumab, or Herceptin, is an antibody used in the treatment of metastatic breast cancer that binds to the epidermal growth factor receptor 2 (EGFR2 or Her2).

 

Typical antibody applications include:

 

• Immunodetection
− Immunoblotting or Western Blot (WB)
− Immunohistochemistry (IHC)
− Immunofluorescent Microscopy (IFM)
− Flow Cytometry (FC, FACS)
− Immunoprecipitation (IP, Chromatin-IP)
− Immunoassay (ELISA, RIA, EIA, ELISPOT)
− Functional assays (activation, blocking, neutralization)
 
Immunophenotyping (cancer diagnosis, prognosis)
 

• Therapeutic use (cancers, infectious diseases & inflammation)

 

 

Recombinant Antibody Production

 

Antibodies are unique in their high affinity and specificity for recognizing a target antigen, a quality that has made them one of the most useful macromolecules in Life Sciences, Biotechnology and Biomedical applications. Modern biotechnology has facilitated the large-scale production of recombinant antibodies. To date, almost all therapeutic antibodies in the clinic and on the market are expressed recombinantly in mammalian cells.

 

Recombinant antibodies have the highest standards of quality and purity in terms of the composition and specificity of antigen-binding. They are able to target specific epitopes, recruit the immune system where appropriate, maintain long serum half-lives, and deliver clinical benefits in patients. The generation of antibody-producing stable cell lines is an important component of the therapeutic antibody development process. CHO has become the industry “workhorse” for the production of therapeutic antibodies. However, the industry relies on several proprietary expression systems and selection methods (e.g., DHFR and GS) for antibody cell line generation, which is a time and resource consuming process (typically 6 to 18 months).

 

 

Recombinant Antibodies for Research Use

 

Antibodies are highly sensitive and specific for particular epitopes, which makes them ideal reagents for research, in particular in antigen detection and quantification. Currently, most research antibodies are produced in animals as monoclonal (with homogenous isotype and antigen specificity) and polyclonal (heterogeneous isotype and antigen specificity) antibodies. A polyclonal antibody supply is dependent on the source animal, and thus no two batches against a particular antigen will be identical. In contrast, monoclonal antibodies are grown from hybridomas, which can produce a continuous supply of homogenous antibody and are the current standard for research antibody production.

 

There are growing interests in using recombinant antibodies for research due to the homogeneity and reproducibility for a recombinant product with defined sequence and composition. It also allows a continuous supply of homogenous antibody (homogeneous composition and antigen specificity) and will probably replace hybridoma to become the future standard for research antibody production. In addition genetic engineering enable the quick switching of isotype, species, and/or subclass of a specific antibody, thus making it possible to generate a complete set of monoclonal antibody from all classes and subclasses with an identical antigen-binding specificity. This is particularly useful for the applications that classes/subclasses matter, e..g., immunostaining and flow cytometry analysis involving the use of secondary antibody as well as in vivo functional studies.

 

 

Antibody Fragments and Derivatives

 

Antibody fragments can be produced through chemical or genetic mechanisms. Chemical fragmentation utilizes reducing agents to break the disulfide bonds and digests the antibody with proteases such as pepsin and papain. For example, chemical and protease digestion of full size antibodies yield antigen binding fragments (Fab) from the variable regions of IgGor IgM. Although biochemical methods are able to generate antibody fragments, it is quite laborious and requires a large quantity of purified antibody starting material. In contrast, genetic engineering and construction of fragments offers the ability to create a multitude of fragment containing molecules, each with unique binding and functional characteristics.

 

 

The abbreviations in the graph above are as follows:

• Fab: fragment, antigen-binding (one arm)
• F(ab')2: fragment, antigen-binding, including hinge region (both arms)
• Fab': fragment, antigen-binding, including hinge region (one arm)
• scFv: single-chain variable fragment
• di-scFv: dimeric single-chain variable fragment
• BiTE: bi-specific T-cell engager

 

Genetic engineering allows the production of a single chain variable fragment (scFv) , which is Fv fragment (VH and VL) linked by a flexible peptide. Manipulation of the orientation of V-domains and the linker length creates different forms of Fv molecules. For example, when the linker is at least 12 amino acids long, the scFv fragment is primarily a monomer. Linkers of 3-11 amino acid long yield a dimeric scFv, which thus creates a bivalent “diabody”. If the linker length is less than three amino acids, scFv molecules associate into “triabody” or “tetrabody”, a multivalent form of scFv with greater binding avidity to the target antigen than a monmeric form. scFv fragments can be generated with two different variable domains, yielding a bispecific molecule to bind to two different epitopes. “Minibodies” are scFv-CH3 fusion proteins that assemble into bivalent dimers. Genetic engineering can also be used to create bispecific (Fab’)2 and trifunctional antibody (see the graph above). 

 

Disadvantages of full size antibodies include their inability to penetrate into certain tissues due to their relatively large size. The Fc region will frequently elicit an immune response, which may be detrimental in certain patients. For research, the Fc domain often causes nonspecific binding, which may impair detection specificity. Fragments offer advantages over a full size antibody for some applications. For example, antibody fragments are small enough to infiltrate into some tissues that full size antibodies are unable, which may help in both therapeutic and immunostaining procedures. However, these fragments lacking Fc are degraded in the body much more rapidly than the full length antibodies.

 

 

G&P Biosciences Antibody Production Capability

 

G&P Biosciences has developed unique mammalian expression systems for recombinant antibody production in a high throughput and time effective manner, especially suitable for research laboratory needs. We have exploited a large panel of expression vectors and selection methods for stable antibody cell line generation. Our expression vectors are designed to allow high throughput cloning of immunoglobulin genes and subsequent expression as whole antibodies or fragments. We can express a variety of different class and subclasses of IgG (e.g., human IgG1-4 including some allotypic variants) from many species, including human, mouse and rabbit. We can also produce many Fv and Fab fragment-derived molecules, such as Fab, Fab’, F(ab)’2, “minibody”, scFv-Fc and bispecific antibodies (visit our "Antidoy Products" and “Antibody Services” to learn more and order).

 

 

» Go to the next topic: Therapeutic Antibody Discovery & Development

 

« Return to Technical Support main page

 

Special Offers & Rewards

Promotion

Earn discounts, credits or rewards with your purchases.

Purchase or Clone My Genes

My Gene Clones

Get the sequence-verified, expression-ready gene clones.

Recombinant Proteins

Recombinant Proteins

Oder your high-quality, recombinant proteins of interest.

Recombinant Antibodies

Recombinant Antibodies

Try our products & services for your antibody R&D.

Virus-Based Gene Expression

Virus Gene Delivery

Acquire high titer, ready-to-use viral particles.

Get in Touch with Us

Send your question or feedback on our products & services