Monoclonal antibody therapeutics and immunogenicity


Abstract

Background

The introduction of monoclonal antibody drugs has had major impact in medicine, leading to extraordinary progress in the treatment of a wide variety of diseases, particularly malignancies and chronic inflammatory and autoimmune diseases.

Content

This chapter provides a general overview of monoclonal antibody drugs approved for clinical use, including the basic biology of antibodies, production, pharmacologic characteristics, nomenclature, and various clinical indications. Because of the widespread use of tumor necrosis factor (TNF) antagonists in clinical medicine, we focus on the clinical use of this class of monoclonal antibody drugs to explain the mechanisms that elicit immunogenicity and to describe methodologies for drug monitoring and immunogenicity testing in the clinical laboratory. Finally, interference of monoclonal antibodies with clinical laboratory testing is reviewed since this is an emerging issue increasingly faced by laboratories as the number of patients treated with these drugs is on the rise.

Monoclonal antibody therapeutics (MAT) were initially introduced in transplantation medicine , following the approval of the first monoclonal antibody drug, muromonab-CD3, by the United States Food and Drug Administration (FDA) in 1985 as an antirejection agent in renal transplant patients. Subsequently in the 1990s several other monoclonal antibodies were approved for clinical use such as rituximab in 1997, followed by trastuzumab and infliximab a year later. All three of these MAT achieved remarkable success, ranking among the top best-selling drugs ever since. The implementation of MAT led to unprecedented progress in the treatment of a wide variety of diseases, including malignancies and chronic inflammatory and autoimmune diseases. MAT are used in oncology, rheumatology, gastroenterology, neurology, and many other areas of medicine. In recent years there have been dramatic advancements in cancer immunotherapy with the introduction of antibody drugs targeting checkpoint inhibitors cytotoxic T lymphocyte-associated 4 (CTLA-4) and programmed cell death-1 (PD-1).

As of the end of 2019, there were 100 unique monoclonal antibody, antibody fragment, or Fc fusion-based drugs, and 20 biosimilars approved by the FDA for the treatment of various diseases. These numbers are expected to increase dramatically in coming years since many more are currently in development.

Biology of antibodies

Antibodies, also known as immunoglobulins (Ig), are naturally occurring large glycoprotein molecules that can be found membrane-bound on the surface of B cells, or in secreted form produced by B cell-derived plasma cells. In addition to recognizing and binding specific antigens, they can induce effector functions such as complement binding and activation of the complement cascade, or induce antibody-dependent cellular cytotoxicity (ADCC). Both of these mechanisms are aimed at eliminating the antigen source by inducing cell killing. Diversity and specificity are essential features of antibodies: the immune system is genetically capable of producing antibodies to virtually any antigen that it encounters, and the antibodies produced are highly specific to that antigen. Antibody-mediated responses constitute crucial mechanisms of the adaptive immune system to provide protection against viruses, bacteria, parasites, and other pathogens.

Ig are large Y-shaped molecules. Each Ig consists of two identical heavy chains (H, 55 kD) and two light chains (L, 22 kD) ( Fig. 98.1 A). Light chains can be of either lambda (λ) or kappa (κ) isotype, which are functionally identical (for additional information, refer to Chapter 31 ). Ig heavy chains belong to five different isotypes, which are functionally and structurally different and define the five major Ig classes in humans: IgM, IgD, IgA, IgE, and IgG. IgG is the major type of Ig in normal human serum. It functions predominantly in the secondary phase of the immune response, since it takes at least 2 to 3 weeks for it to appear in the serum following antigenic encounter. IgG can be further subdivided into IgG1, IgG2, IgG3, and IgG4 subclasses in humans, which vary in abundance, the type of antigen they bind to, and in their ability to induce effector functions. Antibody subclass isotype determines the type of effector function induced; IgG3 and IgG1 are the most potent activators of the complement pathway by interacting with C1q and induce complement-dependent cytotoxicity (CDC). IgG3 and IgG1 also bind to Fc receptors on effector cells activating cellular responses such as ADCC. IgG1 is most abundant in serum, followed by IgG2, IgG3, and IgG4. Except for IgG3, which has a serum half-life of about 7 days, the rest of the IgG subclasses have half-lives of ~21 days. When effector functions such as ADCC are desired, IgG1 is the isotype of choice for MAT development (such as in oncology). When blockade rather than engaging immune effector functions is required, IgG2 and IgG4 have been used. Overall, IgG1 is the most frequently used isotype in MAT.

FIGURE 98.1, Structure of the IgG Immunoglobulin Molecule.

Structurally the IgG heavy and light chains are held together by interchain disulfide bonds in the hinge region (see Fig. 98.1 A). Although the structures are diverse, they contain well-defined domains such as the Ig variable (V) and constant (C) domains. These domains are composed of ~100 amino acid residues that share a common two-layered structure composed of two antiparallel β-sheets. The L chain folds into two domains, VL and CL, while the larger H chain consists of four domains, VH, CH1, CH2, and CH3. The antigen binding sites are located at the amino terminal variable region of both the H and the L chain, also called hypervariable regions or complementarity determining regions (CDR), which form a unique antigen binding site known as idiotype. Each variable domain contains three CDR regions, CDR1, CDR2, and CDR3, which show very high degree of sequence variability, and determine antibody specificity. Antibody diversity is generated by complex genetic mechanisms such as somatic hypermutation and affinity maturation. Despite sequence diversity, all Ig domains have highly similar three-dimensional structures. The available highest-resolution crystal structure of a typical full-length human IgG1 determined by x-ray crystallography is shown on Fig. 98.1 B. The structure indicates a highly asymmetric conformation ranging from Y shape to T shape, due to high molecular flexibility in the hinge region, which makes structure determination of complete IgG molecules difficult. However, mild enzymatic cleavage with proteolytic enzymes papain or pepsin results in fragments that are easier to crystallize ( Fig. 98.1 C). Cleavage of an intact antibody molecule with papain results in two monovalent Fab fragments and one dimeric crystallizable fragment (Fc). Cleavage with pepsin results in a bivalent, single F(ab’) fragment. In addition to intact Ig, these well-defined Ig fragments have medical and therapeutic applications themselves. The Ig domain structure is conserved in nature; its variants are commonly found as part of other proteins, including Fc receptors, adhesion molecules, costimulatory receptors and ligands, and many others.

Therapeutic antibodies

The concept of using serum therapy for treatment of disease dates back to the discovery of diphtheria antitoxin by Emil Adolf von Behring (1890), who was awarded a shared Nobel prize in 1901 for the development of serum therapies against diphtheria and tetanus. Serum polyclonal antibody preparations and pooled IgG from healthy individuals have been used for prevention of several infectious diseases (e.g., hepatitis A), or for replacement therapy in patients with Ig deficiencies. Intravenous Ig, approved in 1980, and subcutaneous Ig, approved in 2006, are widely used as replacement therapy, or for the treatment of various autoimmune diseases or as part of desensitization protocols or treatment of rejection in transplantation.

Polyclonal antibodies used for therapeutic interventions include Rh (D) Ig, an enriched fraction of antibodies directed to the D blood group antigen for the prevention of rhesus D alloimmunization in pregnancy. Anti-thymocyte globulin (ATG) is another example of a widely used polyclonal antibody therapeutic prepared by immunizing mammals (commonly rabbit or horse) with human thymic lymphocytes. ATG administered to patients binds to lymphocytes and depletes them, leading to a profound suppression of the cellular immune response. ATG is used for the treatment of aplastic anemia and organ rejection.

Generation of monoclonal antibody therapeutics

Development of the hybridoma technology in 1975 allowed for production of monoclonal antibodies in vitro, using a hybridoma cell obtained by fusing an immortal myeloma cell (plasma cell-derived tumor cell) and a splenic normal B-cell derived plasma cell. This technology allowed for in vitro production of large amounts of MAT against a wide range of targets, including soluble and cell surface proteins. Early MAT products contained large amounts of nonhuman (i.e., mouse) proteins due to the use of mouse spleen derived plasma cells, which was responsible for the development of adverse effects due to the generation of human anti-mouse antibodies. Clinical experience with the first MAT, murine anti-CD3 muromonab, indicated poor pharmacokinetics and high immunogenicity, making treatment ineffective, which eventually resulted in withdrawal of the drug.

Using recombinant DNA technologies and better understanding of antibody structure-function correlation allowed for the development of chimeric (>65% human on average), humanized (>80% human), and fully human (>95% human) antibodies. Chimeric antibodies contain murine sequences in the variable region since they are initially developed as mouse antibodies. The rest of the molecule is replaced with human sequences, which reduces, but does not eliminate immunogenicity, and improves effector functions due to the presence of the human Fc part. The first chimeric antibody, abciximab, a human-mouse Fab fragment, was approved in the US in 1994, followed by rituximab in 1997, the first chimeric full-length antibody. The next phase of technology development allowed for the exchange of rodent sequences almost entirely to human sequences throughout the molecule, by grafting of the rodent CDR onto human IgG. The first humanized antibody, daclizumab, an antibody to IL-2 receptor α subunit, was approved in the United States in 1997 for the treatment of transplant rejection (withdrawn in 2009). Generation of fully human antibodies became possible with the development of phage-display technology, and transgenic mouse platforms. Phage display platform has become a commodity since the technology patents expired recently. It allows for designing and manipulating the repertoire of antibody genes used as antibody sources, followed by in vitro selection. The first antibody to reach the US market developed by this technology was the tumor necrosis factor (TNF) antagonist adalimumab, approved in 2002. In contrast to phage display platform, transgenic animal technology allows for in vivo selection process and requires less optimization and shorter timelines to reach clinical development. Antibody-producing plasma cells isolated from the spleen of immunized, genetically modified mice that express fully human monoclonal antibodies but are unable to produce mouse antibodies, are used to create hybridomas, which are used for subsequent large-scale production. MAT produced by transgenic technologies include panitumumab, approved in 2006 for oncology, and golimumab, a TNF antagonist approved in 2009.

As of the end of 2019, there were 100 unique monoclonal antibody-based drugs approved in the United States ( Table 98.1 ). Eighty-six of these are monoclonal antibodies or antibody fragments, 12 are Fc fusion proteins or peptides, and 2 are engineered chimeric antigen receptor T-cell (CAR T) therapies. Of the 86 monoclonal antibody drugs 4 are murine, 10 are chimeric, 40 humanized (human-murine, human-rat, human-camelid), and 32 are fully human. Most of these drugs are full-length antibodies, the most common isotype being IgG1, which occurs in 61 (71%) of approved antibodies; 8 are IgG2 (9%), 2 are IgG2/IgG4 hybrid (2%), and 13 are IgG4 (15%). Additional structures include camelid, and single chain Fv fragments. 11 out of the 12 approved Fc fusion drugs contain IgG1, and one is fused to IgG4 Fc. Engineering the Fc part is becoming more common in order to modulate effector functions, such as using glycoengineering to enhance effector function. As an example, afucosylation of IgG1 Fc in benralizumab facilitates binding to FcγRIII on natural killer (NK) cells, enhancing ADCC-mediated apoptosis of eosinophils and basophils, which is beneficial for the treatment of severe eosinophilic asthma. The Fc part can also be modified to silence effector functions such as in abatacept or eculizumab. Another approach for improving pharmacokinetics is to incorporate mutations to increase neonatal Fc receptor (FcRn) binding, such as the YTE mutation in some antibodies in development. Fusing IgG Fc with other proteins or peptides improves their stability and half-life. This approach is used in 12 currently approved MAT including TNF receptor, enzymes (phosphatase), peptides (glucagon-like), and clotting factors (VIII, IX). In addition to the full-length antibodies, five of the MAT are Fab antibody fragments. Some Fabs are fused to stabilizer molecules to improve pharmacokinetics in the absence of the Fc part, such as certolizumab pegol, which is PEGylated. Another innovative approach takes advantage of bispecific monoclonal antibodies designed as single-chain variable fragments of two different antibodies combined, such as in blinatumomab (approved in 2014 for acute leukemia), which is a bi-specific T-cell engager, connecting T cells via CD3 with tumor cells expressing CD19, facilitating immunologic synapse formation and tumor cell killing. Most of the approved MAT are nonconjugated intact IgG. However, ten of the MAT used in oncology are conjugated with drug molecules such as antimitotic agents, enzymes, or radioisotopes, which provide additional therapeutic benefits aimed at killing the tumor cell.

TABLE 98.1
Monoclonal Antibody Therapeutics Approved by the US Food and Drug Administration as of December 2019
Data obtained from FDA Center for Drug Evaluation and Research (CDER) list of licensed biological products; from FDA news releases; and from FDA-approved package inserts.
Product (Proper) Name Proprietary Name Date of Licensure (month/day/year) Molecular Target Protein Format Source Major Indication
Oncology
Capromab pendetide ProstaScint 10/28/1996 Prostate specific membrane antigen (PSMA), or glutamate carboxypep-tidase 2 Murine IgG1, conjugated to Indium-111-pendetide Mouse hybridoma Imaging of prostate cancer
Rituximab Rituxan 11/26/1997 CD20 Chimeric human-murine IgG1κ CHO cells NHL, CLL, RA
Trastuzumab Herceptin 09/25/1998 EGFR (HER2) Humanized IgG1κ CHO cells HER2-positive breast cancer
Alemtuzumab Campath, Lemtrada 05/07/2001 CD52 Humanized rat IgG1κ CHO cells B-cell CLL
Ibritumomab tiuxetan Zevalin 02/19/2002 CD20 Murine IgG1 conjugated to Indium or yttrium CHO cells NH lymphoma
Cetuximab Erbitux 02/12/2004 EGFR Chimeric murine-human mAb Fab SP2/0 murine myeloma Metastatic colorectal carcinoma (EGFR+)
Bevacizumab Avastin 02/26/2004 VEGF Humanized IgG1 CHO Metastatic colorectal cancer, Her2 negative metastatic breast cancer
Panitumumab Vectibix 09/27/2006 EGFR Human IgG2 Transgenic technology/Hybridoma cells Metastatic colorectal carcinoma
Ofatumumab Arzerra 10/26/2009 CD20 Human IgG1κ Mouse hybridoma CLL
Ipilimumab Yervoy 03/25/2011 CTLA-4 Human IgG1κ CHO cells Metastatic melanoma
Brentuximab vedotin Adcetris 08/19/2011 CD30 Chimeric human-murine IgG1 coupled with monomethyl auristatin E (MMAE), a microtubule disrupting agent, through a protease-cleavable linker CHO cells for the mAb HL, anaplastic LCL
Pertuzumab Perjeta 06/08/2012 EGFR (HER2) Humanized IgG1 CHO cells HER2-positive breast cancer
Ziv-aflibercept Zaltrap 08/03/2012 VEGF VEGFR fused to human IgG1 Fc CHO cells Metastatic colorectal cancer
Ado-trastuzumab emtansine Kadcyla 02/22/2013 EGFR (HER2) Humanized IgG1k linked to anti-microtubule agent DM1 CHO cells for the mAb HER2-positive breast cancer
Obinutuzumab Gazyva 11/01/2013 CD20 Humanized IgG1 CHO cells CLL
Ramucirumab Cyramza 04/21/2014 VEGFR2 Human IgG1 Hybridoma cells Gastric adenocarcinoma
Pembrolizumab Keytruda 09/04/2014 PD-1 (CD279) Humanized IgG4κ Hybridoma Melanoma, NSCLC, Head and Neck cancer
Blinatumomab Blincyto 12/03/2014 CD19, CD3d Murine antibody, constructed BiTE (bi-specific T-cell engager) CHO cells B-cell ALL
Nivolumab Opdivo 12/22/2014 PD-1 (CD279) Human IgG4 Transgenic mouse technology Metastatic melanoma
Dinutuximab Unituxin 03/10/2015 GD2, disialogan-glioside Chimeric IgG1κ Murine hybridoma Neuroblastoma
Daratumumab Darzalex 11/16/2015 CD38 Human IgG1κ CHO cells Multiple myeloma
Necitumumab Portrazza 11/24/2015 EGFR Human IgG1κ NSO Myeloma cells NSCLC
Elotuzumab Empliciti 11/30/2015 SLAMF7 Humanized IgG1κ NSO Myeloma cells Multiple myeloma
Atezolizumab Tecentriq 05/18/2016 PD-L1 (CD274) Humanized IgG1, Fc engineered (nonglycosylated) Mouse hybridoma Urothelial carcinoma
Olaratumab Lartruvo 10/19/2016 PDGFRa Human IgG1 NSO Myeloma cells Soft tissue sarcoma (STS)
Avelumab Bavencio 03/23/2017 PD-L1 (CD274) Human IgG1λ CHO cells Metastatic Merkel cell carcinoma
Durvalumab Imfinzi 05/01/2017 PD-L1 (CD274) Human IgG1κ CHO cells Urothelial carcinoma
Rituximab and Hyaluronidase human Rituxan Hycela 06/22/2017 CD20 Chimeric human-murine IgG1κ CHO cells Follicular lymphoma, diffuse large B-cell lymphoma, CLL
Inotuzumab Ozogamicin Besponsa 08/17/2017 CD22 Humanized IgG4κ attached to N-acetyl-γ-calicheamicin dimethylhydrazide CHO cells ALL
Tisagenle-cleucel Kymriah 08/30/2017 CD19 Chimeric antigen receptor (CAR) Expressed by patient’s own cells by in vitro molecular engineering ALL
Gemtuzumab ozogamicin Mylotarg 09/01/2017 CD33 Humanized IgG4κ attached to calicheamicin NSO Myeloma cells CD33 positive AML
Axicabtagene ciloleucel Yescarta 10/18/2017 CD19 Chimeric antigen receptor (CAR) Expressed by patient’s own cells by in vitro molecular engineering Large B-cell lymphoma
Mogamulizumab-kpkc Poteligeo 08/08/2018 CCR4 Humanized IgG1κ CHO cells T-cell malignancies: mycosis fungoides or Sézary syndrome
Moxetumomab pasudotox-tdfk Lumoxiti 09/13/2018 CD22 Murine IgV fused to Pseudomonas exotoxin PE38 Escherichia coli Hairy cell leukemia
Cemiplimab-rwlc Libtayo 09/28/2018 PD-1 (CD279) Human IgG4 CHO cells Cutaneous squamous cell carcinoma (CSCC)
Trastuzumab and Hyaluronidase-oysk Herceptin Hylecta 02/28/2019 EGFR (HER2) Humanized IgG1κ CHO cells HER2-positive breast cancer
Polatuzumab vedotin-piiq Polivy 06/10/2019 CD79b Humanized IgG1 fused to anti-mitotic agent MMAE through protease-cleavable linker CHO cells Diffuse large B-cell lymphoma
Enfortumab vedotin-ejfv Padcev 12/18/2019 Nectin 4 Human IgG1κ conjugated to ani-mitotic agent monomethyl auristatin E (MMAE) through protease-cleavable linker CHO cells Advanced or metastatic urothelial cancer
Fam-trastuzumab deruxtecan-nxki Enhertu 12/20/2019 Her2 Humanized IgG1 linked to topoisomerase inhibitor DXd through protease cleavable linker CHO cells Metastatic breast cancer
Rheumatology
Infliximab Remicade 08/24/1998 TNF Chimeric human-murine IgG1κ Mouse hybridoma RA, PsA, AS, Crohn disease, PsO
Etanercept Enbrel 11/02/1998 TNF p75 TNFR2—huIgG1 Fc fusion CHO cells RA, PsO, AS, JIA, plaque psoriasis
Adalimumab Humira 12/31/2002 TNF Human IgG1κ CHO cells RA, PsA, AS, JIA, Crohn disease, plaque psoriasis, hidradenitis suppurativa, uveitis
Abatacept Orencia 12/23/2005 CTLA-4 ligands CD80 (B7-1), CD86 (B7-2) CTLA-4 fused to IgG1 Fc CHO cells RA, PsA, JIA
Certolizumab pegol Cimzia 04/22/2008 TNF Humanized IgG4 Fab E. coli RA, PsA, AS, Crohn disease
Golimumab Simponi 04/24/2009 TNF Human IgG1κ Mouse SP2/0 hybridoma RA, PsA, AS, Spondyloarthritis, UC
Tocilizumab Actemra 01/08/2010
10/21/2013
IL-6R (CD126) Humanized IgG1κ Hybridoma RA, JIA, Giant cell arteritis, Cytokine release syndrome
Golimumab Simponi Aria 07/18/2013 TNF Human IgG1κ Transgenic mouse technology RA, PsA, AS
Sarilumab Kevzara 05/22/2017 IL-6R (CD126) Human IgG1 CHO cells RA
Secukinumab Cosentyx 01/21/2015 IL-17A Human IgG1κ CHO cells Psoriasis, AS
Ustekinumab Stelara 09/25/2009
09/23/2016
p40 subunit of IL-12 and Il-23 Human IgG1κ SP2/0 murine myeloma Plaque psoriasis, PsA, Crohn disease
Canakinumab Ilaris 06/17/2009 IL-1β Human IgG1κ Mouse Sp2/0-Ag14 cell line Cryopyrin-associated periodic syndromes: Familial cold autoinflammatory syndrome and Muckle-Wells syndrome; JIA
Dermatology
Guselkumab Tremfya 07/13/2017 IL-23 p19 Human IgG1λ Mammalian cell line Plaque psoriasis
Tildrakizumab-asmn Ilumya 03/20/2018 IL-23 p19 Humanized IgG1κ CHO cells Plaque psoriasis
Risankizumab-rzaa Skyrizi 04/23/2019 IL-23 p19 Humanized IgG1 Mammalian cell line Plaque psoriasis
Ustekinumab Stelara 09/25/2009
9/23/2016
p40 subunit of IL-12 and Il-23 Human IgG1κ SP2/0 murine myeloma Plaque psoriasis, PsA, Crohn disease
Secukinumab Cosentyx 01/21/2015 IL-17A Human IgG1κ CHO cells Psoriasis, AS
Ixekizumab Taltz 03/22/2016 IL-17A Humanized IgG4κ Mouse hybridoma Plaque psoriasis
Brodalumab Siliq 02/15/2017 IL-17RA Human IgG2 CHO cells Plaque psoriasis
Dupilumab Dupixent 03/28/2017 IL-4Ra, IL-4 and IL-13 receptor Human IgG4 CHO cells Eczema, atopic dermatitis
Blood Disease
Emicizumab-kxwh Hemlibra 11/16/2017 Factor Ixa and Factor X Humanized IgG4, bispecific CHO cells Hemophilia
Caplacizumab-yhdp Cablivi 02/06/2019 von Willebrand factor Humanized bivalent antibody fragment (Nanobody, heavy chain fragment from Camelidae) E. coli Acquired thrombotic thrombocytopenic purpura (aTTP)
Efmoroctocog alfa Eloctate 06/06/2014 Factor VIII substitute Factor VII fused to human IgG1 Fc HEK cell line Hemophilia A
Luspatercept-aamt Reblozyl 11/08/2019 TGF-β superfamily ligands Activin receptor type IIB linked to human IgG1 Fc domain CHO cells Erythroid maturation agent
Crizanlizumab-tmca Adakveo 11/15/2019 P-selectin Humanized IgG2κ CHO cells Sickle cell disease
Romiplostim Nplate 08/22/2008 Thrombopoietin receptor c-Mpl Peptibody, fused to IgG1 Fc E. coli chronic immune thrombocytopenic purpura
Eftrenonacog alfa Alprolix 03/28/2014 Factor IX substitute Factor IX fused to human IgG1 Fc HEK293H cell line Hemophilia B
Idarucizumab Praxbind 10/16/2015 Dabigatran, thrombin inhibitor drug Humanized IgG1 Fab CHO cells Anticoagulant reversal
Gastroenterology
Infliximab Remicade 08/24/1998 TNF Chimeric human-murine IgG1κ Mouse hybridoma RA, PsA, AS, Crohn disease, plaque psoriasis
Adalimumab Humira 12/31/2002 TNF Human IgG1κ CHO cells RA, PsA, AS, JIA, Crohn disease, plaque psoriasis, hidradenitis suppurativa, uveitis
Certolizumab pegol Cimzia 04/22/2008 TNF Humanized IgG4 Fab E. coli RA, PsA, AS, Crohn disease
Golimumab Simponi 04/24/2009 TNF Human IgG1κ Mouse SP2/0 hybridoma RA, PsA, AS, Spondyloarthritis, UC
Ustekinumab Stelara 09/25/2009
09/23/2016
p40 subunit of IL-12 and Il-23 Human IgG1κ SP2/0 murine myeloma Plaque psoriasis, PsA, Crohn disease
Natalizumab Tysabri 11/23/2004 α4-integrin Humanized IgG4κ Murine myeloma MS, Crohn disease
Vedolizumab Entyvio 05/20/2014 a4b7 integrin Humanized IgG1 CHO cells UC, Crohn disease
Autoinflammatory and Other Immunologic Disease
Lanadelumab-flyo Takhzyro 08/23/2018 Plasma kallikrein Human IgG1κ CHO cells Hereditary angioedema (HAE)
Emapalumab-lzsg Gamifant 11/20/2018 Interferon-γ Human IgG1 CHO cells Hemophagocytic lymphohistiocytosis (HLH)
Ravulizumab-cwvz Ultomiris 12/21/2018 Complement C5 Humanized IgG2/4κ CHO cells Paroxysmal nocturnal hemoglobinuria (PNH)
Eculizumab Soliris 03/16/2007 C5 Humanized IgG2/4κ Murine hybridoma PNH
Canakinumab Ilaris 06/17/2009 IL-1β Human IgG1κ Mouse Sp2/0-Ag14 cell line Cryopyrin-associated periodic syndromes: Familial cold autoinflammatory syndrome and Muckle-Wells syndrome; JIA
Siltuximab Sylvant 04/23/2014 IL-6 Chimeric IgG1 CHO cells multicentric Castleman’s disease
Rilonacept Arcalyst 02/27/2008 IL-1 IL-1R and IL-1R accessory protein linked to hu IgG1 Fc CHO cells Cryopyrin-associated periodic syndromes: Familial cold autoinflammatory syndrome and Muckle-Wells syndrome
Neurologic Disease
Ocrelizumab Ocrevus 03/28/2017 CD20 Humanized IgG1 CHO cells Progressive MS
Natalizumab Tysabri 11/23/2004 α4-integrin Humanized IgG4κ Murine myeloma MS, Crohn disease
Erenumab-aooe Aimovig 05/17/2018 Calcitonin gene-related peptide receptor (CGRP) Human IgG2λ CHO cells Migraine
Fremanezumab-vfrm Ajovy 09/14/2018 Calcitonin gene-related peptide (CGRP) ligand Humanized IgG2Δa/κ CHO cells Migraine
Galcanezumab-gnlm Emgality 09/27/2018 Calcitonin gene-related peptide (CGRP) ligand Humanized IgG4κ CHO cells Migraine
Allergy and Asthma
Reslizumab Cinqair 03/23/2016 IL-5 Humanized IgG4κ NSO Myeloma cells Severe eosinophilic asthma
Omalizumab Xolair 06/20/2003 IgE Humanized IgG1κ CHO cells Asthma caused by allergies
Benralizumab Fasenra 11/14/2017 IL-5Ra Humanized IgG1κ, Fc engineered (afucosylated) CHO cells Severe eosinophilic asthma
Mepolizumab Nucala 11/04/2015 IL-5 Humanized IgG1κ CHO cells Severe eosinophilic asthma; Eosinophilic granulomatosis with polyangiitis (EGPA), appr 6/6/19
Dupilumab Dupixent 03/28/2017 IL-4Ra, IL-4 and IL-13 receptor Human IgG4 CHO cells Eczema, atopic dermatitis
Infectious Disease
Palivizumab Synagis 06/19/1998 RSV F protein, A antigenic site Humanized IgG1κ Mouse hybridoma RSV infection
Raxibacumab Raxibacumab 12/14/2012 Bacillus anthracis PA toxin Human IgG1λ Murine hybridoma Anthrax
Ibalizumab-uiyk Trogarzo 03/06/2018 CD4 Humanized IgG4 NSO Myeloma cells HIV infection
Obiltoxaximab Anthim 03/18/2016 B. anthracis PA toxin Chimeric IgG1κ, affinity enhanced Mouse hybridoma Anthrax—Biodefense
Bezlotoxumab Zinplava 10/21/2016 Clostridium difficile toxin B Human IgG1 CHO cells C. difficile infection
Bone Disease
Burosumab-twza Crysvita 04/17/2018 Fibroblast Growth Factor (FGF23) Human IgG1κ CHO cells X-linked hypophosphatemia (XLH), a form of rickets
Romosozumab-aqqg Evenity 04/09/2019 Sclerostin Humanized IgG2 CHO cells Osteoporosis in postmenopausal women
Denosumab Prolia, Xgeva 06/01/2010 RANKL, NF-κB ligand Human IgG2 CHO cells Osteoporosis, bone metastasis from solid tumors
Asfotase alfa Strensiq 10/23/2015 TNSALP enzyme replacement Catalytic domain of human tissue nonspecific alkaline phosphatase (TNSALP), fused to human IgG1 Fc, and a deca-aspartate peptide as bone targeting domain CHO cells Hypophosphatasia
Cardiovascular Disease
Abciximab ReoPro 12/22/1994 GP IIb/IIIa platelet receptor Chimeric human-murine mAb Fab Mouse hybridoma CVD
Alirocumab Praluent 07/24/2015 PCSK9, paraprotein convertase subtilisin/kexin type 9 Human IgG1 CHO cells Familial hypercholesterolemia, CVD
Evolocumab Repatha 08/27/2015 PCSK9, paraprotein convertase subtilisin/kexin type 10 Human IgG2 CHO cells Familial hypercholesterolemia, CVD
Ophthalmology
Brolucizumab-dbll Beovu 10/07/2019 VEGF Humanized single-chain Fv (scFv) antibody fragment E. coli Neovascular (Wet) age-related macular degeneration (AMD)
Ranibizumab Lucentis 06/30/2006 VEGF-A Humanized IgG1κ mAb fragment E. coli Macular edema, age-related macular degeneration
Aflibercept Eylea 11/18/2011 VEGF VEGFR fused to human IgG1 Fc CHO cells Macular edema
Transplant
Basiliximab Simulect 05/12/1998 CD25, IL-2Rα Chimeric human-murine IgG1κ Mouse hybridoma Transplant rejection
Belatacept Nulojix 06/15/2011 CTLA-4 ligands CD80 (B7-1), CD86 (B7-2) CTLA-4 (2 amino acid mutant) fused to IgG1 Fc CHO cells Transplant rejection
Systemic Autoimmune
Belimumab Benlysta 03/09/2011 iv
07/20/2017 sc
B lymphocyte stimulator protein (BLyS)/TNFSF13B/BAFF Human IgG1λ NSO Myeloma cells SLE
Mepolizumab Nucala 11/04/2015 IL-5 Humanized IgG1κ CHO cells Severe eosinophilic asthma; Eosinophilic granulomatosis with polyangiitis (EGPA), appr 6/6/19
Metabolic Disease
Dulaglutide Trulicity 09/18/2014 GLP-1R Glucagon-like peptide-1 agonist (GLP-1) fused to human IgG4 Fc HEK293H cell line Type II diabetes
ALL, Acute lymphocytic leukemia; AML, acute myeloid leukemia; AS, ankylosing spondylitis; CHO, Chinese hamster ovary; CLL, chronic lymphocytic leukemia; CTLA4, cytotoxic T lymphocyte-associated 4; CVD, cardiovascular disease; EGFR, epidermal growth factor receptor; GD2, disialoganglioside-2; GLP-1, glucagon-like peptide-1; GP, glycoprotein; HEK, human embryonic kidney; HL, Hodgkin’s lymphoma; hu, human; Ig, immunoglobulin; IL, interleukin; JIA, juvenile idiopathic arthritis; LCL, large cell lymphoma; Mpl, myeloproliferative leukemia virus oncogene; MS, multiple sclerosis; NHL, non-Hodgkin’s lymphoma; NSCLC, non–small cell lung cancer; PCSK9, Proprotein convertase subtilisin/kexin type 9; PD-1, programmed cell death-1; PDGFR, platelet-derived growth factor receptor; PD-L1, programmed cell death-1 ligand-1; PNH, paroxysmal nocturnal hemoglobinuria; PsA, psoriatic arthritis; PsO, plaque psoriasis; RA, rheumatoid arthritis; RANKL, receptor activator of nuclear factor κ-B ligand; RSV, respiratory syncytial virus; SLAMF7, signaling lymphocytic activation molecule family member 7; SLE, systemic lupus erythematosus; TNF, tumor necrosis factor; TNFR, tumor necrosis factor receptor; TNFSF, tumor necrosis factor ligand superfamily; TNSALP, tissue nonspecific alkaline phosphatase; VEGF, vascular endothelial growth factor.

As a major breakthrough for cancer immunotherapy, two CAR T-cell therapy agents were approved in 2017: tisagenlecleucel for advanced leukemia and axicabtagene ciloleucel for lymphoma. Chimeric antigen receptor (CAR) is engineered as a single-chain variable fragment molecule fused to signaling domains, which is then expressed on host T cells, grafting monoclonal antibody specificity to the T cells. When injected back to the body, engineered host T cells expressing CAR receptors will recognize the target antigen on the cancer cells, such as CD19 on leukemic or lymphoma cells, and will induce tumor cell killing.

Pharmacokinetic/pharmacodynamic properties

MAT are administered parenterally by intravenous, subcutaneous, or intramuscular routes. The intravenous route has the advantage of offering systemic delivery, the ability to deliver high volumes, and most importantly, assures complete bioavailability. However, limitations include inconvenience and adverse effects due to infusion reactions. Subcutaneous and intramuscular delivery offers lower bioavailability (24 to 95%) but allows self-administration and decreased rate of infusion-related adverse events. Although MAT distribute to most tissues to varying degrees, due to their large molecular weight (typically ~150 kDa) they are not able to cross the blood brain barrier and cannot enter the central nervous system.

MAT are eliminated from circulation through various mechanisms: antigen-specific target-mediated disposition and elimination through endothelial cells of the reticuloendothelial system. Target-mediated disposition involves antigen recognition and binding, typically to a membrane bound target, and subsequent clearance by endocytosis and lysosomal degradation. Since the number of antigenic targets is limited, this process is saturable and nonlinear. Elimination through the reticuloendothelial system, however, is non-saturable and linear, and it involves both free and bound drug. These two processes typically occur in parallel, for MAT targeting a membrane-bound antigen. In contrast to small molecule drugs, the liver and kidneys are not considered essential in the elimination of MAT under normal physiologic conditions, but clearance may be affected in the context of kidney injury leading to disruption of endothelial lining of the glomeruli (such as glomerulonephritis). Other routes affecting MAT clearance include loss of the drug through the gastrointestinal tract in protein losing enteropathy in patients with inflammatory bowel disease (IBD).

Since MAT are typically IgG molecules, their expected half-lives are quite long. This is explained by FcRn mediated uptake and endosomal recycling, a process that substantially extends the half-life of IgG ( Fig. 98.2 ). The binding affinity of FcRn for IgG is negligible at physiologic pH, but when IgG is internalized and enters the endosomes, in the acidic environment of endocytic vacuoles (pH 6.5) it binds to FcRn with high affinity, which rescues IgG from degradation, recycling it through transcytosis. , The poor pharmacokinetics of mouse antibody-based drugs observed in earlier trials can be explained by their low affinity to human FcRn, which makes the recycling process less efficient.

FIGURE 98.2, IgG Plasma Half-Life is Extended by Neonatal Fc Receptor (FcRn) Mediated Cellular Transcytosis and Recycling.

Of interest, high interindividual variability has been observed in pharmacokinetics of several MAT drugs. Based on multiple clinical trials, the most commonly identified covariates on MAT pharmacokinetics include body weight/surface area, gender, antidrug antibodies (ADA), creatinine clearance, age, disease severity, and inflammation markers such as C-reactive protein. Other factors influencing inter-individual variability include influence of disease elements such as proteinuria and protein losing enteropathy as mentioned above, or injury to blood-brain barrier, and influence of co-administered drugs. Better understanding of pharmacokinetics and patient-specific covariates paves the way toward personalized MAT therapy, with the goal of increasing efficacy in a cost-effective way, while decreasing toxicity.

Nomenclature

Nomenclature of MAT is currently standardized by international guidelines adopted by the American Medical Association’s United States Adopted Names Council. All monoclonal antibody product names end in the suffix –mab ( Fig. 98.3 ). The nomenclature includes a distinct starting prefix to create a unique name, which is followed by an infix representing the target or disease. Infixes indicating the target class, or disease the antibody is used to treat for include tu-/t- for tumor, li-/l- for immunomodulator, vi- for viral, ba- for bacterial, etc. Since the source of the antibody has important safety consideration in that it may induce immunogenicity in patients, a second infix included indicates the source of the antibody (the species on which the Ig sequence of the antibody is based): o for mouse, a for rat, u for human, i for primate. In addition, the degree of presence and origin of nonhuman sequences is further indicated by additional letters in the source infix such as –xi- indicating chimeric (~65% human), –zu- indicating humanized (~80% human), and -u- indicating fully human (>95% human) antibody. For engineered fusion proteins, suffix –cept indicates the presence of receptor molecules as part of the recombinant Fc fusion (such as etanercept for TNF receptor Ig fusion).

FIGURE 98.3, Standardized Nomenclature for Therapeutic Monoclonal Antibodies.

Antigens and diseases targeted by monoclonal antibody therapeutics

MAT were developed against a number of unique antigen targets. Some antigens show overlap between clinical areas, for example, TNF antagonists are used in rheumatology, as well as gastroenterology for the treatment of Crohn disease (CD). Some antigens are more frequently targeted than others (100 drugs for 61 targets), with TNF and CD20 being the most popular with 6 drugs approved for each, followed by HER2 and VEGF with 5 drugs, and CD19, EGFR, IL-17RA, IL-23 p19, PD-1, and PD-L1 with three antibodies for each. The majority of approved MAT are directed against targets in oncology (35%). Other areas include rheumatology (11%), dermatology (7%), blood disorders (7%), gastroenterology (6%), autoinflammatory and other immunologic disease (6%), allergy and asthma (4%), infectious disease (4%), neurologic disease including multiple sclerosis (4%), bone disorders (4%), cardiovascular disease (3%), ophthalmology (3%), transplant and systemic autoimmune disease (2% each), and type II diabetes (1%).

Antigenic targets for MAT are frequently cell membrane bound receptors such as GPIIb/IIIa, integrins, EGFR, CD20, CD52, CTLA-4, PD-1, PD-L1, or soluble targets including growth factors (VEGF), cytokines (TNF, IL-6, IL-17, IL-12/23), complement (C5), or IgE. While most MAT are designed to recognize human targets, a few of them that are used to treat or prevent infectious diseases bind to microbial protein targets (RSV-F protein, Bacillus anthracis PA toxin, Clostridium difficile toxin).

MAT targets for oncology can be classified into three different categories. The first category includes tumor-specific antigens, adhesion molecules serving as “postal addresses” for which killing mechanisms can be targeted. Tumor cell killing can be achieved by activation of immunologic mechanisms following antibody binding to tumor cells (e.g., ADCC or CDC), or by conjugating the antibody to another molecule mediating the killing such as toxins or radioactive isotopes. The second target category overlaps with the first, since it includes tumor cell receptors, which are targeted by MAT with the goal to block ligand binding and signal transduction, for example Her2. The third category includes immunotherapy agents such as checkpoint inhibitors designed to block co-inhibitory signals and thus directly stimulate tumor-specific T-cell responses, or CAR-T cells that express single chain chimeric antigen receptors specific to tumor antigen and are used to induce tumor cell killing.

MAT targeting cytokines include antibodies to pro-inflammatory cytokines IL-1, IL-6, IL-17, and TNF, which play crucial roles in the pathogenesis of numerous chronic inflammatory and autoimmune diseases. These MAT typically block the biological effect by either neutralizing the cytokine or blocking the cytokine receptor that mediates signaling. Due to their pleiotropic biological effects, which include playing a crucial role in host defense against pathogens, systemic pro-inflammatory cytokine blockade may interfere with host defense against infections, which is an unwanted adverse effect.

Points to remember

  • Immunoassays, cell-based assays, and mass spectrometry approaches are available to measure monoclonal antibody therapeutics (MAT) concentrations in the clinical lab.

  • It is important to interpret MAT concentration and antidrug antibodies results in the context of the assay used, and timing of blood samples.

  • Therapeutic drug monitoring of MAT is relevant in the setting of loss of response to therapy (reactive monitoring) and newer evidence suggests a role for proactive continuous monitoring during different time-points in the course of treatment to adjust therapy before loss of response occurs.

  • Interference of MAT with clinical laboratory tests is an emerging concern that can trigger unnecessary additional testing and may delay important therapeutic decisions.

  • MAT are mostly of the IgG κ isotype and some of them are given in high enough doses that they show up as monoclonal bands in protein electrophoresis and IgG κ bands upon immunofixation testing. Approaches to differentiate the MAT from an endogenous monoclonal protein have been developed.

The use of tumor necrosis factor antagonists in clinical medicine

Due to the central role of TNF in inflammation and the pathogenesis of autoimmune and chronic inflammatory diseases, TNF antagonists have revolutionized the treatment of rheumatoid arthritis (RA), ankylosing spondylitis, psoriasis, and IBD including CD and ulcerative colitis. , The therapeutic benefits have been so dramatic that TNF antagonists are still among the best-selling, most prescribed pharmaceuticals. In recent decades several monoclonal antibody-based TNF antagonists have been developed ( Fig. 98.4 , Table 98.2 ). Infliximab is a chimeric mouse/human antibody in which the mouse variable region is preserved, but the rest of the molecule is replaced by human sequences. Adalimumab and golimumab are fully human IgG1 antibodies, produced by phage display or transgenic mouse technology, respectively. Despite containing predominantly human sequences, these molecules may show structural features that are different from the endogenous proteins, such as glycosylation patterns that are characteristic of the producing cells (Chinese Hamster Ovary cells [CHO] or mouse myeloma cells), not necessarily identical to the human glycosylation pattern, which may contribute to immunogenicity. Certolizumab pegol is a humanized antibody Fab fragment, designed by engrafting the mouse CDR into a human IgG4 κ Fab framework, and it has a polyethylene glycol (PEG) molecule attached that prolongs the half-life in the circulation. Etanercept is a fusion protein composed of the extracellular part of human TNF type 2 receptor (TNF-R2, p75) fused to dimeric human IgG1 Fc. Unlike the antibody drugs targeting TNF, etanercept is able to neutralize both TNF receptor ligands, TNF and Lymphotoxin-α.

FIGURE 98.4, Tumor Necrosis Factor (TNF) Antagonists Approved for Clinical Use.

TABLE 98.2
Tumor Necrosis Factor Antagonists Currently in Clinical Use and Their Structural and Pharmacologic Properties
TNF Inhibitors Etanercept Infliximab Adalimumab Certolizumab Pegol Golimumab
Molecule hTNF receptor Fc fusion protein Chimeric TNF antibody Human TNF antibody Humanized antibody Fab, PEGylated Human TNF antibody
Protein Format Dimeric hIgG1 Fc fused to hTNF-R2 (p75) hIgG1κ, murine variable region hIgG1κ, selected by phage display hIgG4 Fab, murine CDR, conjugated to 40 kDa PEG hIgG1κ, produced by transgenic mouse technology
Source CHO cells Sp2/0 hybridoma CHO cells Escherichia coli SP2/0 hybridoma
Sequence origin Human ~75% human Human ~80% human Human
Method of administration sc 50 mg once weekly iv 5 mg/kg at 0, 2, 6, and then every 8 weeks 40 mg every 2 weeks sc 400 mg at 0, 2, 4, and then every 4 weeks sc 50 mg once a month; iv 2 mg/kg at 0 and 4, then every 8 weeks
Half-life (days) 3–5 8–10 10–20 14 14
Year of first FDA approval 1998 1998 2002 2008 2009
Indications RA, JIA, PsA, AS, PsO CD, UC, RA, AS, PsA, PsO RA, JIA, PsA, AS, CD, PsO CD, RA, PsA, AS, RA, PsA, AS
AS, Ankylosing spondylitis; CD, Crohn disease; CDR, complementarity determining region; CHO, Chinese hamster ovary; FDA, Food and Drug Administration; h, human; iv, intravenous infusion; PsA, psoriatic arthritis; PsO, plaque psoriasis; RA, rheumatoid arthritis; sc, subcutaneous injection; UC, ulcerative colitis.

Biosimilars

Patent expiration of biological drugs created the opportunity for manufacturers to develop biosimilar products, which are highly similar to and have no clinically meaningful differences from existing FDA-approved reference products. An abbreviated pathway for approval was created by Congress through the Biologics Price Competition and Innovation Act of 2009, assuring a shorter and less costly development program with the goal of providing more treatment options, lowering cost through competition, and increasing patient access to treatment. Similarity is determined by extensive structural and functional analysis, comparison of purity, chemical identity, and bioactivity; minor differences are however acceptable (e.g., stabilizer, buffer). For approval of a biosimilar, the manufacturer must demonstrate that there are no clinically meaningful differences compared to the reference product in pharmacokinetics, clinical efficacy, and safety. Assessment of clinical immunogenicity is important during biosimilar development. Due to the complexity of the manufacturing processes for MAT, involving recombinant DNA technology and production by living cells, it cannot be assumed that biosimilars are completely identical to the reference drug (in contrast to small molecule generic drugs, which have chemically identical structure to the reference drug).

Since 2015, 20 biosimilars to MAT have been approved in the US, 18 of which are monoclonal antibodies, along with 2 Fc fusions ( Table 98.3 ). The majority of them are TNF antagonists: four are biosimilars for infliximab, five for adalimumab, and two for etanercept. Biosimilars are expected to show similar immunogenicity profile compared to the reference drug, which is verified during clinical trials. However, due to the variety of assay platforms used across clinical laboratories, internal validation to verify assay performance for the biosimilar compared to the reference drug should be performed before implementing testing for biosimilar drug or ADA to biosimilars.

TABLE 98.3
US Food and Drug Administration-approved Biosimilars to Monoclonal Antibody Therapeutics
Data obtained from FDA Therapeutic Biologic Applications, Biosimilar product information, and from FDA package inserts.
Product Name Proprietary Name Date of Licensure (month/day/year) Molecular Target Protein Format Source Major Indication Reference Drug Reference Drug Proprietary Name Reference Drug Date of Licensure (month/day/year)
1 Infliximab-dyyb Inflectra 04/05/2016 TNF Chimeric IgG1κ Murine hybridoma RA, PsA, AS, Crohn disease, UC, PsO Infliximab Remicade 08/24/1998
2 Etanercept-szzs Erelzi 08/30/2016 TNF Fc fusion, p75 TNFR2—huIgG1 Fc CHO cells RA, PsA, AS, JIA, PsO Etanercept Enbrel 11/02/1998
3 Adalimumab-atto Amjevita 09/23/2016 TNF Human IgG1κ CHO cells RA, PsA, AS, JIA, Crohn disease, UC, PsO Adalimumab Humira 12/31/2002
4 Infliximab-abda Renflexis 04/21/2017 TNF Chimeric IgG1κ Murine hybridoma RA, PsA, AS, Crohn disease, UC, PsO Infliximab Remicade 08/24/1998
5 Adalimumab-adbm Cyltezo 08/25/2017 TNF Human IgG1κ CHO cells RA, PsA, AS, JIA, Crohn disease, UC, PsO Adalimumab Humira 12/31/2002
6 Bevacizumab-awwb Mvasi 09/14/2017 VEGF Humanized IgG1 CHO Metastatic colorectal cancer, Her2 negative metastatic breast cancer Bevacizumab Avastin 02/26/2004
7 Trastuzumab-dkst Ogivri 12/01/2017 EGFR (HER2) Humanized IgG1κ CHO cells HER2-positive breast cancer Trastuzumab Herceptin 09/25/1998
8 Infliximab-qbtx Ixifi 12/13/2017 TNF Chimeric IgG1κ Murine hybridoma RA, PsA, AS, Crohn disease, UC, PsO Infliximab Remicade 08/24/1998
9 Adalimumab-adaz Hyrimoz 10/30/2018 TNF Human IgG1κ CHO cells RA, PsA, AS, JIA, Crohn disease, UC, plaque psoriasis Adalimumab Humira 12/31/2002
10 Rituximab-abbs Truxima 11/28/2018 CD20 Chimeric IgG1k CHO cells NHL Rituximab Rituxan 11/26/1997
11 Trastuzumab-pkrb Herzuma 12/14/2018 EGFR (HER2) Humanized IgG1κ CHO cells HER2-positive breast cancer Trastuzumab Herceptin 09/25/1998
12 Trastuzumab-dttb Ontruzant 01/18/2019 EGFR (HER2) Humanized IgG1κ CHO cells HER2-positive breast cancer, and gastric adenocarcinoma Trastuzumab Herceptin 09/25/1998
13 Trastuzumab-qyyp Trazimera 03/11/2019 EGFR (HER2) Humanized IgG1κ CHO cells HER2-positive breast cancer, and gastric adenocarcinoma Trastuzumab Herceptin 09/25/1998
14 Etanercept-ykro Eticovo 04/26/2019 TNF Fc fusion, p75 TNFR2—huIgG1 Fc CHO cells RA, PsA, AS, JIA, PsO Etanercept Enbrel 11/02/1998
15 Trastuzumab-anns Kanjinti 06/13/2019 EGFR (HER2) Humanized IgG1κ CHO cells HER2-positive breast cancer, and gastric adenocarcinoma Trastuzumab Herceptin 09/25/1998
16 Bevacizumab-bvzr Zirabev 06/28/2019 VEGF Humanized IgG1 CHO Metastatic colorectal cancer, NSCLC, recurrent glioblastoma, RCC, cervical cancer Bevacizumab Avastin 02/26/2004
17 Rituximab-pvvr Ruxience 07/23/2019 CD20 Chimeric IgG1κ CHO cells NHL, CLL, GPA, MPA Rituximab Rituxan 11/26/1997
18 Adalimumab-bwwd Hadlima 07/25/2019 TNF Human IgG1κ Mammalian cells RA, JIA, PsA, AS, Crohn disease, UC, PsO Adalimumab Humira 12/31/2002
19 Adalimumab-afzb Abrilada 11/18/2019 TNF Human IgG1κ CHO cells RA, JIA, PsA, AS, Crohn disease, UC, PsO Adalimumab Humira 12/31/2002
20 Infliximab-axxq Avsola 12/06/2019 TNF Chimeric IgG1κ CHO cells Crohn disease, UC, RA, AS, PsA, PsO Infliximab Remicade 08/24/1998
AS, Ankylosing spondylitis; CHO, Chinese hamster ovary; EGFR, epidermal growth factor receptor; GPA, granulomatosis with polyangiitis; hu, human; Ig, immunoglobulin; JIA, juvenile idiopathic arthritis; MPA, microscopic polyangiitis; NHL, Non-Hodgkin’s Lymphoma; NSCLC, non–small cell lung cancer; PsA, psoriatic arthritis; PsO, plaque psoriasis; RA, rheumatoid arthritis; RCC, renal cell carcinoma; TNF, tumor necrosis factor; TNFSF, tumor necrosis factor ligand superfamily; TNFR, tumor necrosis factor receptor; TNSALP, tissue nonspecific alkaline phosphatase; UC, ulcerative colitis; VEGF, vascular endothelial growth factor.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here