Comparison of methods generating antibody-epitope conjugates for targeting cancer with virus-specific T cells

Therapeutic antibody-epitope conjugates (AECs) are promising new modalities to deliver immunogenic epitopes and redirect virus-specific T-cell activity to cancer cells. Nevertheless, many aspects of these antibody conjugates require optimization to increase their efficacy. Here we evaluated different strategies to conjugate an EBV epitope (YVL/A2) preceded by a protease cleavage site to the antibodies cetuximab and trastuzumab. Three approaches were taken: chemical conjugation (i.e. a thiol-maleimide reaction) to reduced cysteine side chains, heavy chain C-terminal enzymatic conjugation using sortase A, and genetic fusions, to the heavy chain (HC) C-terminus. All three conjugates were capable of T-cell activation and target-cell killing via proteolytic release of the EBV epitope and expression of the antibody target was a requirement for T-cell activation. Moreover, AECs generated with a second immunogenic epitope derived from CMV (NLV/A2) were able to deliver and redirect CMV specific T-cells, in which the amino sequence of the attached peptide appeared to influence the efficiency of epitope delivery. Therefore, screening of multiple protease cleavage sites and epitopes attached to the antibody is necessary. Taken together, our data demonstrated that multiple AECs could sensitize cancer cells to virus-specific T cells.

[1]  D. Lowy,et al.  Harnessing anti-cytomegalovirus immunity for local immunotherapy against solid tumors , 2022, Proceedings of the National Academy of Sciences of the United States of America.

[2]  R. Hagedoorn,et al.  A broad and systematic approach to identify B-cell malignancy targeting TCRs for multi-antigen based T-cell therapy. , 2021, Molecular therapy : the journal of the American Society of Gene Therapy.

[3]  A. Labrijn,et al.  Overcoming Challenges for CD3-Bispecific Antibody Therapy in Solid Tumors , 2021, Cancers.

[4]  C. Ó’Fágáin,et al.  Antibody stability: A key to performance - Analysis, influences and improvement. , 2020, Biochimie.

[5]  C. Robert A decade of immune-checkpoint inhibitors in cancer therapy , 2020, Nature Communications.

[6]  W. Meng,et al.  Protein Aggregation and Immunogenicity of Biotherapeutics. , 2020, International journal of pharmaceutics.

[7]  R. Jain,et al.  Antibody-mediated delivery of viral epitopes to tumors harnesses CMV-specific T cells for cancer therapy , 2020, Nature Biotechnology.

[8]  C. Rader Bispecific antibodies in cancer immunotherapy. , 2019, Current opinion in biotechnology.

[9]  Juan Zhang,et al.  Antibody–Drug Conjugates: A Comprehensive Review , 2019, Molecular Cancer Research.

[10]  Vladimir Ponomarev,et al.  Silencing Fc Domains in T cell–Engaging Bispecific Antibodies Improves T-cell Trafficking and Antitumor Potency , 2019, Cancer Immunology Research.

[11]  C. Hackenberger,et al.  Tag and release: strategies for the intracellular cleavage of protein conjugates. , 2019, Current opinion in chemical biology.

[12]  anonymous,et al.  Comprehensive review , 2019 .

[13]  R. Offringa,et al.  Sensitization of Tumors for Attack by Virus-Specific CD8+ T-Cells Through Antibody-Mediated Delivery of Immunogenic T-Cell Epitopes , 2019, Front. Immunol..

[14]  C. Caux,et al.  Cold Tumors: A Therapeutic Challenge for Immunotherapy , 2019, Front. Immunol..

[15]  Clark C. Chen,et al.  Virus-specific memory T cells populate tumors and can be repurposed for tumor immunotherapy , 2019, Nature Communications.

[16]  T. Schumacher,et al.  Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers , 2018, Nature Medicine.

[17]  S. M. Toor,et al.  Immune checkpoint inhibitors: recent progress and potential biomarkers , 2018, Experimental & Molecular Medicine.

[18]  P. Dreger,et al.  Antibodies conjugated with viral antigens elicit a cytotoxic T cell response against primary CLL ex vivo , 2018, Leukemia.

[19]  P. Norlén,et al.  Bispecific antibodies in cancer immunotherapy , 2018, Therapeutic advances in vaccines and immunotherapy.

[20]  Jedd D. Wolchok,et al.  Cancer immunotherapy using checkpoint blockade , 2018, Science.

[21]  E. Jaffee,et al.  Targeting neoantigens to augment antitumour immunity , 2017, Nature Reviews Cancer.

[22]  P. Burke,et al.  Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody–Drug Conjugates , 2016, Molecular Cancer Therapeutics.

[23]  G. Salvesen,et al.  Counter Selection Substrate Library Strategy for Developing Specific Protease Substrates and Probes. , 2016, Cell chemical biology.

[24]  Siqi Chen,et al.  Bispecific antibodies in cancer immunotherapy , 2016, Human vaccines & immunotherapeutics.

[25]  P. Klenerman,et al.  T cell responses to cytomegalovirus , 2016, Nature Reviews Immunology.

[26]  P. Burke,et al.  Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index , 2015, Nature Biotechnology.

[27]  W. Jiskoot,et al.  Small Amounts of Sub-Visible Aggregates Enhance the Immunogenic Potential of Monoclonal Antibody Therapeutics , 2015, Pharmaceutical Research.

[28]  R. Feederle,et al.  Antigen-armed antibodies targeting B lymphoma cells effectively activate antigen-specific CD4+ T cells. , 2015, Blood.

[29]  R. Hagedoorn,et al.  Therapeutic targeting of the BCR-associated protein CD79b in a TCR-based approach is hampered by aberrant expression of CD79b. , 2015, Blood.

[30]  J. Wolchok,et al.  Immune Checkpoint Blockade in Cancer Therapy. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[31]  Paul H. Bessette,et al.  Tumor-Specific Activation of an EGFR-Targeting Probody Enhances Therapeutic Index , 2013, Science Translational Medicine.

[32]  D. Teachey,et al.  Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood. 2015;125(26):4017-4023. , 2016, Blood.

[33]  T. Wu,et al.  Targeted coating with antigenic peptide renders tumor cells susceptible to CD8(+) T cell-mediated killing. , 2013, Molecular therapy : the journal of the American Society of Gene Therapy.

[34]  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.

[35]  T. Schumacher,et al.  Dissection of T-cell antigen specificity in human melanoma. , 2012, Cancer research.

[36]  R. J. Cohen,et al.  Promiscuous binding of extracellular peptides to cell surface class I MHC protein , 2012, Proceedings of the National Academy of Sciences.

[37]  H. Ploegh,et al.  Site‐Specific Protein Labeling via Sortase‐Mediated Transpeptidation , 2009, Current protocols in protein science.

[38]  A. Rickinson,et al.  Cellular responses to viral infection in humans: lessons from Epstein-Barr virus. , 2007, Annual review of immunology.

[39]  Damon L. Meyer,et al.  Development of potent monoclonal antibody auristatin conjugates for cancer therapy , 2003, Nature Biotechnology.

[40]  G. Dubowchik,et al.  Cathepsin B-sensitive dipeptide prodrugs. 1. A model study of structural requirements for efficient release of doxorubicin. , 1998, Bioorganic & medicinal chemistry letters.

[41]  F. Khanim,et al.  HLA A2.1-restricted cytotoxic T cells recognizing a range of Epstein-Barr virus isolates through a defined epitope in latent membrane protein LMP2 , 1993, Journal of virology.

[42]  C. Barnstable,et al.  Monoclonal Antibodies for Analysis of the HLA System , 1979, Immunological reviews.

[43]  G. Mclendon,et al.  Is protein turnover thermodynamically controlled? , 1978, The Journal of biological chemistry.

[44]  T. Vink,et al.  A simple, robust and highly efficient transient expression system for producing antibodies. , 2014, Methods.

[45]  J. Yewdell,et al.  Immunodominance in major histocompatibility complex class I-restricted T lymphocyte responses. , 1999, Annual review of immunology.