Recommendations for the characterization of immunogenicity response to multiple domain biotherapeutics.

Many biotherapeutics currently in development have complex mechanisms of action and contain more than one domain, each with a specific role or function. Examples include antibody-drug conjugates (ADC), PEGylated, fusion proteins and bi-specific antibodies. As with any biotherapeutic molecule, a multi-domain biotherapeutic (MDB) can elicit immune responses resulting in the production of specific anti-drug antibodies (ADA) when administered to patients. As it is beneficial to align industry standards for evaluating immunogenicity of MDBs, this paper highlights pertinent immunogenicity risk factors and describes steps involved in the design of a testing strategy to detect and characterize binding (non-neutralizing and neutralizing, NAb) ADAs. In a common tier based approach, samples identified as ADA screen positive are confirmed for the binding specificity of the antibodies to the drug molecule via a confirmatory assay. The confirmation of specificity is generally considered as a critical step of the tier based approach in overall ADA response evaluation. Further characterization of domain specificity of polyclonal anti-MDB ADA response may be required based on the analysis of molecule specific risk factors. A risk based approach in evaluating the presence of NAbs for MDB is discussed in this article. Analysis of domain-specific neutralizing antibody reactivity should be based on the risk assessment as well as the information learned during binding ADA evaluation. Situations where additional characterization of NAb specificity is possible and justified are discussed. Case studies demonstrating applicability of the risk factor based approach are presented. In general, the presence of a domain with high immunogenicity risk or presence of a domain with high endogenous protein homology may result in an overall high immunogenicity risk level for the entire MDB and can benefit from domain specificity characterization of immune response. For low immunogenicity risk MDBs, domain specificity characterization could be re-considered at later clinical phases based on the need to explain specific clinical observations. Inclusion of domain specificity characterization in early phase clinical studies for MDBs with limited clinical immunogenicity experience may be considered to help understand its value in later clinical development. It is beneficial and is recommended to have a well-defined plan for the characterization of ADA domain specificity and data analysis prior to the initiation of sample testing. Overall, best practices for immunogenicity evaluation of complex MDBs are discussed.

[1]  Marian Kelley,et al.  Recommendations for the design, optimization, and qualification of cell-based assays used for the detection of neutralizing antibody responses elicited to biological therapeutics. , 2007, Journal of immunological methods.

[2]  A. Janssens Romiplostim for the treatment of primary immune thrombocytopenia , 2012, Expert review of hematology.

[3]  S. Zeuzem,et al.  Peginterferon alfa-2a (40 kDa) monotherapy: a novel agent for chronic hepatitis C therapy , 2001, Expert opinion on investigational drugs.

[4]  Daniela Verthelyi,et al.  Managing uncertainty: a perspective on risk pertaining to product quality attributes as they bear on immunogenicity of therapeutic proteins. , 2012, Journal of pharmaceutical sciences.

[5]  Viswanath Devanarayan,et al.  Recommendations for the validation of immunoassays used for detection of host antibodies against biotechnology products. , 2008, Journal of pharmaceutical and biomedical analysis.

[6]  P. Ruf,et al.  Bi20 (fBTA05), a novel trifunctional bispecific antibody (anti‐CD20 × anti‐CD3), mediates efficient killing of B‐cell lymphoma cells even with very low CD20 expression levels , 2008, International journal of cancer.

[7]  Gopi Shankar,et al.  Scientific and regulatory considerations on the immunogenicity of biologics. , 2006, Trends in biotechnology.

[8]  H. Okudaira,et al.  Reaginic antibody formation in the mouse. II. Enhancement and suppression of anti-hapten antibody formation by priming with carrier. , 1973 .

[9]  R Seitz,et al.  Taking immunogenicity assessment of therapeutic proteins to the next level. , 2011, Biologicals : journal of the International Association of Biological Standardization.

[10]  J. M. Harris,et al.  Pegylation: a novel process for modifying pharmacokinetics. , 2001, Clinical pharmacokinetics.

[11]  M. Hershfield,et al.  Control of hyperuricemia in subjects with refractory gout, and induction of antibody against poly(ethylene glycol) (PEG), in a phase I trial of subcutaneous PEGylated urate oxidase , 2005, Arthritis research & therapy.

[12]  Robin Marsden,et al.  Summary of Confirmation Cut Point Discussions , 2011, The AAPS Journal.

[13]  M. Raff,et al.  The carrier effect in the secondary response to hapten‐protein conjugates. II. Cellular cooperation , 1971, European journal of immunology.

[14]  D. Dimmock,et al.  Successful immune tolerance induction to enzyme replacement therapy in CRIM-negative infantile Pompe disease , 2012, Genetics in Medicine.

[15]  Emma D. Deeks Certolizumab Pegol , 2013, Drugs.

[16]  H. Hartung,et al.  Atacicept: targeting B cells in multiple sclerosis , 2010, Therapeutic advances in neurological disorders.

[17]  L L Miller,et al.  Abrogation of the hematological and biological activities of the interleukin-3/granulocyte-macrophage colony-stimulating factor fusion protein PIXY321 by neutralizing anti-PIXY321 antibodies in cancer patients receiving high-dose carboplatin. , 1999, Blood.

[18]  D. Mason,et al.  Deaggregated homologous immunoglobulin‐peptide conjugates induce peptide‐specific T cell nonresponsiveness in vivo , 1998, European journal of immunology.

[19]  D. Stanworth,et al.  Factors influencing the immunogenicity of the haptenic drug chlorhexidine in mice. II. The role of the carrier and adjuvants in the induction of IgE and IgG anti-hapten responses. , 1986, Immunology.

[20]  E. Vitetta,et al.  The use of haptenated immunoglobulins to induce B cell tolerance in vitro. The roles of hapten density and the Fc portion of the immunoglobulin carrier. , 1983, Journal of immunology.

[21]  Rosenberg As Immunogenicity of biological therapeutics: a hierarchy of concerns. , 2003, Developments in biologicals.

[22]  P. V. Schouwenburg,et al.  Immunogenicity of anti-TNF biologic therapies for rheumatoid arthritis , 2013, Nature Reviews Rheumatology.

[23]  Guy Georges,et al.  The intriguing options of multispecific antibody formats for treatment of cancer. , 2013, Cancer genomics & proteomics.

[24]  Viswanath Devanarayan,et al.  Recommendations on risk-based strategies for detection and characterization of antibodies against biotechnology products. , 2008, Journal of immunological methods.

[25]  Dan Lu,et al.  Clinical pharmacology of trastuzumab emtansine (T-DM1): an antibody–drug conjugate in development for the treatment of HER2-positive cancer , 2012, Cancer Chemotherapy and Pharmacology.

[26]  Y. Inada,et al.  Biomedical and biotechnological applications of PEG- and PM-modified proteins. , 1995, Trends in biotechnology.

[27]  T. Boone,et al.  Clinical validation of the "in silico" prediction of immunogenicity of a human recombinant therapeutic protein. , 2007, Clinical immunology.

[28]  J. Bluestone,et al.  Pathologic Role and Temporal Appearance of Newly Emerging Autoepitopes in Relapsing Experimental Autoimmune Encephalomyelitis1 , 2000, The Journal of Immunology.

[29]  D. Scott,et al.  Epitope-specific tolerance induction with an engineered immunoglobulin. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[30]  D. Seimetz,et al.  Catumaxomab: clinical development and future directions. , 2010, mAbs.

[31]  A. Compston,et al.  A Novel Strategy To Reduce the Immunogenicity of Biological Therapies , 2010, The Journal of Immunology.

[32]  T. Ishida,et al.  Anti-PEG IgM elicited by injection of liposomes is involved in the enhanced blood clearance of a subsequent dose of PEGylated liposomes. , 2007, Journal of controlled release : official journal of the Controlled Release Society.

[33]  P. Lampertico,et al.  PegIFN-α2a for the treatment of chronic hepatitis B and C: a 10-year history , 2013, Expert review of anti-infective therapy.

[34]  L. Lundquist Abatacept: a novel treatment for rheumatoid arthritis , 2007, Expert opinion on pharmacotherapy.

[35]  Pascal Richette,et al.  Antibodies against polyethylene glycol in healthy subjects and in patients treated with PEG-conjugated agents , 2012, Expert opinion on drug delivery.

[36]  I. Nestorov,et al.  A Novel PEGylated Interferon Beta‐1a for Multiple Sclerosis: Safety, Pharmacology, and Biology , 2012, Journal of clinical pharmacology.

[37]  V. Tuohy,et al.  A predictable sequential determinant spreading cascade invariably accompanies progression of experimental autoimmune encephalomyelitis: a basis for peptide-specific therapy after onset of clinical disease , 1996, The Journal of experimental medicine.

[38]  Daniel Baltrukonis,et al.  Comparison of competitive ligand-binding assay and bioassay formats for the measurement of neutralizing antibodies to protein therapeutics. , 2011, Journal of pharmaceutical and biomedical analysis.

[39]  R. Kinkel,et al.  Diversity and plasticity of self recognition during the development of multiple sclerosis. , 1997, The Journal of clinical investigation.

[40]  P. Senter,et al.  The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma , 2012, Nature Biotechnology.

[41]  L. Garrison,et al.  Etanercept: therapeutic use in patients with rheumatoid arthritis , 1999, Annals of the rheumatic diseases.

[42]  P. Fournier,et al.  Bispecific Antibodies and Trispecific Immunocytokines for Targeting the Immune System Against Cancer , 2012, BioDrugs.

[43]  George Scott,et al.  Recommendations for the design and optimization of immunoassays used in the detection of host antibodies against biotechnology products. , 2004, Journal of immunological methods.

[44]  C. Won,et al.  PEG-modified biopharmaceuticals , 2009 .

[45]  A. Ohtsu,et al.  Phase 1 study of trebananib (AMG 386), an angiogenesis targeting angiopoietin-1/2 antagonist, in Japanese patients with advanced solid tumors , 2012, Cancer Chemotherapy and Pharmacology.

[46]  Huub Schellekens,et al.  The Immunogenicity of Polyethylene Glycol: Facts and Fiction , 2013, Pharmaceutical Research.

[47]  A. Wahl,et al.  In vivo drug-linker stability of an anti-CD30 dipeptide-linked auristatin immunoconjugate. , 2005, Clinical cancer research : an official journal of the American Association for Cancer Research.

[48]  Arunan Kaliyaperumal,et al.  Immunogenicity testing strategy and bioanalytical assays for antibody-drug conjugates. , 2013, Bioanalysis.

[49]  S. Mouritsen,et al.  Induction of cross-reactive antibodies against a self protein by immunization with a modified self protein containing a foreign T helper epitope. , 1997, Molecular immunology.

[50]  J. W. Lindsey Use of reinduced experimental autoimmune encephalomyelitis to evaluate the importance of epitope spread. , 1998, International immunology.

[51]  A. Thakur,et al.  Activated T cells from umbilical cord blood armed with anti‐CD3 × anti‐CD20 bispecific antibody mediate specific cytotoxicity against CD20+ targets with minimal allogeneic reactivity: a strategy for providing antitumor effects after cord blood transplants , 2012, Transfusion.

[52]  Ray Yin,et al.  A double antigen bridging immunogenicity ELISA for the detection of antibodies to polyethylene glycol polymers. , 2011, Journal of pharmacological and toxicological methods.

[53]  Gopi Shankar,et al.  A risk-based bioanalytical strategy for the assessment of antibody immune responses against biological drugs , 2007, Nature Biotechnology.