A novel anti-DR5 antibody-drug conjugate possesses a high-potential therapeutic efficacy for leukemia and solid tumors

It is well known that tumor necrosis factor-related apoptosis inducing ligand receptor 1 or 2 (DR4/DR5) is specifically expressed in various tumor cells, but less or no expression in most normal cells. Many first generations of TRAIL agonists including recombinant preparations of TRAIL, agonistic antibodies against DR4/DR5 have been developed in phase I/II clinical trials for cancer therapy. However, the outcomes of clinical trials by using DR4/DR5 agonist mono-therapy were disappointed even though the safety profile was well tolerance. In the present study, we report an anti-DR5 antibody-drug conjugate (ADC, named as Zapadcine-1) possesses a higher potential for the therapy of lymphocyte leukemia and solid cancers. Methods: Zapadcine-1 was made by a fully humanized DR5-specific monoclonal antibody (Zaptuzumab) coupled via a cleavable linker to a highly toxic inhibitor of tubulin, monomethyl auristatin D (MMAD), by using ThioBridge technology. Cytotoxicity of the ADC in various tumor cells was identified by luminescent cell viability assay and the efficacy in vivo was determined in cells derived xenografts (CDX) of Jurkat E6-1, BALL-1, Reh, and patient derived xenografts (PDX) of human acute leukemia. Preliminary safety evaluation was carried out in rat and monkey. Results: Zapadcine-1 possesses a similar binding ability to the death receptor DR5 as the naked monoclonal antibody Zaptuzumab, and can be rapidly endocytosed into the lysosome of cancer cells. Zapadcine-1 specifically kills human lymphocyte leukemia cells and solid tumor cells, but not normal cells tested. More importantly, Zapadcine-1 drastically eliminates the xenografts in both CDX and PDX models of human acute leukemia. The excellent and comparable therapeutic efficacy is also observed in lung cancer NCI-H1975 CDX mouse model. The maximum-tolerated dose (MTD) of single injected Zapadcine-1 in rat and cynomolgus monkey shows an acceptable safety profile. Conclusion: These data demonstrate a promising anti-cancer activity, meriting further exploration of its potential as a novel cancer therapeutic agent, especially for the acute lymphocyte leukemia.

[1]  T. Sauter,et al.  Systemic network analysis identifies XIAP and IκBα as potential drug targets in TRAIL resistant BRAF mutated melanoma , 2018, npj Systems Biology and Applications.

[2]  O. Micheau,et al.  Antibodies and Derivatives Targeting DR4 and DR5 for Cancer Therapy , 2017, Antibodies.

[3]  H. Gronemeyer,et al.  Dual role of DR5 in death and survival signaling leads to TRAIL resistance in cancer cells , 2017, Cell Death and Disease.

[4]  E. Szliszka,et al.  Targeting Apoptotic Activity Against Prostate Cancer Stem Cells , 2017, International journal of molecular sciences.

[5]  Jiong Cai,et al.  A novel humanized anti‐tumor necrosis factor‐related apoptosis‐inducing ligand‐R2 monoclonal antibody induces apoptotic and autophagic cell death , 2017, IUBMB life.

[6]  J. Lambert,et al.  Antibody–Drug Conjugates (ADCs) for Personalized Treatment of Solid Tumors: A Review , 2017, Advances in Therapy.

[7]  C. Dumontet,et al.  Strategies and challenges for the next generation of antibody–drug conjugates , 2017, Nature Reviews Drug Discovery.

[8]  A. Farooqi,et al.  Journey of TRAIL from Bench to Bedside and its Potential Role in Immuno-Oncology , 2017, Oncology reviews.

[9]  William A. Hammond,et al.  Emerging therapeutic agents for lung cancer , 2016, Journal of Hematology & Oncology.

[10]  黄长江,et al.  Covalent linkers in antibody-drug conjugates and methods of making and using the same , 2016 .

[11]  J. Ji,et al.  Paclitaxel enhances tumoricidal potential of TRAIL via inhibition of MAPK in resistant gastric cancer cells. , 2016, Oncology Report.

[12]  Shery Jacob,et al.  A simple practice guide for dose conversion between animals and human , 2016, Journal of basic and clinical pharmacy.

[13]  Paul Polakis,et al.  Antibody Drug Conjugates for Cancer Therapy , 2016, Pharmacological Reviews.

[14]  L. Lou,et al.  A novel humanized anti-HER2 antibody conjugated with MMAE exerts potent anti-tumor activity , 2015, Breast Cancer Research and Treatment.

[15]  M. Dorywalska,et al.  Site-Dependent Degradation of a Non-Cleavable Auristatin-Based Linker-Payload in Rodent Plasma and Its Effect on ADC Efficacy , 2015, PloS one.

[16]  B. Tomczuk,et al.  Current ADC Linker Chemistry , 2015, Pharmaceutical Research.

[17]  J. Hou,et al.  A multicenter, open‐label phase II study of recombinant CPT (Circularly Permuted TRAIL) plus thalidomide in patients with relapsed and refractory multiple myeloma , 2014, American journal of hematology.

[18]  Paul Polakis,et al.  Site-specific antibody drug conjugates for cancer therapy , 2013, mAbs.

[19]  Juan Shi,et al.  TRAIL suppresses tumor growth in mice by inducing tumor-infiltrating CD4+CD25+ Treg apoptosis , 2013, Cancer Immunology, Immunotherapy.

[20]  Juan Shi,et al.  A novel anti‐DR5 chimeric antibody and epirubicin synergistically suppress tumor growth , 2012, IUBMB life.

[21]  Juan Shi,et al.  Adeno-associated virus-mediated anti-DR5 chimeric antibody expression suppresses human tumor growth in nude mice. , 2011, Cancer letters.

[22]  L. Maestre,et al.  Immunohistochemical analysis of HLDA9 Workshop antibodies against cell-surface molecules in reactive and neoplastic lymphoid tissues. , 2011, Immunology letters.

[23]  B. Mayer,et al.  Natural Immunity Enhances the Activity of a DR5 Agonistic Antibody and Carboplatin in the Treatment of Ovarian Cancer , 2010, Molecular Cancer Therapeutics.

[24]  Juan Shi,et al.  Targeting a Novel N-terminal Epitope of Death Receptor 5 Triggers Tumor Cell Death* , 2010, The Journal of Biological Chemistry.

[25]  D. Benjamin,et al.  Intracellular Activation of SGN-35, a Potent Anti-CD30 Antibody-Drug Conjugate , 2010, Clinical Cancer Research.

[26]  Ya-xi Zhang,et al.  TRAIL receptor mediates inflammatory cytokine release in an NF-κB-dependent manner , 2009, Cell Research.

[27]  P. Senter Potent antibody drug conjugates for cancer therapy. , 2009, Current opinion in chemical biology.

[28]  Eeva-Liisa Eskelinen,et al.  Autophagy: a lysosomal degradation pathway with a central role in health and disease. , 2009, Biochimica et biophysica acta.

[29]  V. Deretic,et al.  Unveiling the roles of autophagy in innate and adaptive immunity , 2007, Nature Reviews Immunology.

[30]  Ronald Koschny,et al.  The promise of TRAIL—potential and risks of a novel anticancer therapy , 2007, Journal of Molecular Medicine.

[31]  Juan Shi,et al.  Therapeutic expression of an anti-death receptor 5 single-chain fixed-variable region prevents tumor growth in mice. , 2006, Cancer research.

[32]  Yabin Guo,et al.  A Novel Anti-human DR5 Monoclonal Antibody with Tumoricidal Activity Induces Caspase-dependent and Caspase-independent Cell Death* , 2005, Journal of Biological Chemistry.

[33]  Damon L. Meyer,et al.  Effects of Drug Loading on the Antitumor Activity of a Monoclonal Antibody Drug Conjugate , 2004, Clinical Cancer Research.

[34]  Damon L. Meyer,et al.  cAC10-vcMMAE, an anti-CD30-monomethyl auristatin E conjugate with potent and selective antitumor activity. , 2003, Blood.

[35]  G. Gores,et al.  Is TRAIL hepatotoxic? , 2001, Hepatology.

[36]  S. S. Strom,et al.  Differential hepatocyte toxicity of recombinant Apo2L/TRAIL versions , 2001, Nature Medicine.

[37]  Henning Walczak,et al.  TRAIL‐R2: a novel apoptosis‐mediating receptor for TRAIL , 1997, The EMBO journal.

[38]  R. Gentz,et al.  An antagonist decoy receptor and a death domain-containing receptor for TRAIL. , 1997, Science.

[39]  Arul M. Chinnaiyan,et al.  The Receptor for the Cytotoxic Ligand TRAIL , 1997, Science.

[40]  A. Ciechanover Intracellular protein degradation: from a vague idea through the lysosome and the ubiquitin-proteasome system and onto human diseases and drug targeting. , 2013, Bioorganic & medicinal chemistry.

[41]  Juan Shi,et al.  2A peptide-based, lentivirus-mediated anti-death receptor 5 chimeric antibody expression prevents tumor growth in nude mice. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.