Orthogonal Cysteine Protection Enables Homogeneous Multi‐Drug Antibody–Drug Conjugates

Abstract A strategy for the preparation of homogeneous antibody–drug conjugates (ADCs) containing multiple payloads has been developed. This approach utilizes sequential unmasking of cysteine residues with orthogonal protection to enable site‐specific conjugation of each drug. In addition, because the approach utilizes conjugation to native antibody cysteine residues, it is widely applicable and enables high drug loading for improved ADC potency. To highlight the benefits of ADC dual drug delivery, this strategy was applied to the preparation of ADCs containing two classes of auristatin drug‐linkers that have differing physiochemical properties and exert complementary anti‐cancer activities. Dual‐auristatin ADCs imparted activity in cell line and xenograft models that are refractory to ADCs comprised of the individual auristatin components. This work presents a facile method for construction of potent dual‐drug ADCs and demonstrates how delivery of multiple cytotoxic warheads can lead to improved ADC activities. Lastly, we anticipate that the conditions utilized herein for orthogonal cysteine unmasking are not restricted to ADCs and can be broadly utilized for site‐specific protein modification.

[1]  Corbin E. Meacham,et al.  Tumour heterogeneity and cancer cell plasticity , 2013, Nature.

[2]  R. Chari,et al.  Antibody-drug conjugates: an emerging concept in cancer therapy. , 2014, Angewandte Chemie.

[3]  Xiaoxiao He,et al.  Labeling Thiols on Proteins, Living Cells, and Tissues with Enhanced Emission Induced by FRET , 2013, Scientific Reports.

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

[5]  R. Chari,et al.  Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. , 2014, Journal of medicinal chemistry.

[6]  Guy Cavet,et al.  Functional genomics identifies ABCC3 as a mediator of taxane resistance in HER2-amplified breast cancer. , 2008, Cancer research.

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

[8]  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.

[9]  M. Follettie,et al.  Tumor Cells Chronically Treated with a Trastuzumab–Maytansinoid Antibody–Drug Conjugate Develop Varied Resistance Mechanisms but Respond to Alternate Treatments , 2015, Molecular Cancer Therapeutics.

[10]  E. K. Maloney,et al.  Antibody-maytansinoid conjugates designed to bypass multidrug resistance. , 2010, Cancer Research.

[11]  F. Loganzo,et al.  Development of Solid-Phase Site-Specific Conjugation and Its Application toward Generation of Dual Labeled Antibody and Fab Drug Conjugates. , 2016, Bioconjugate chemistry.

[12]  Damon L. Meyer,et al.  Enhanced activity of monomethylauristatin F through monoclonal antibody delivery: effects of linker technology on efficacy and toxicity. , 2006, Bioconjugate chemistry.

[13]  R. Soriano,et al.  A Novel Anti-CD22 Anthracycline-Based Antibody–Drug Conjugate (ADC) That Overcomes Resistance to Auristatin-Based ADCs , 2015, Clinical Cancer Research.

[14]  Antoine Maruani,et al.  Recent advances in the construction of antibody-drug conjugates. , 2016, Nature chemistry.

[15]  Edward Chu,et al.  A history of cancer chemotherapy. , 2008, Cancer research.

[16]  Daniel A. Richards,et al.  Dual modification of biomolecules. , 2016, Organic & biomolecular chemistry.

[17]  Enrique Miranda,et al.  A plug-and-play approach to antibody-based therapeutics via a chemoselective dual click strategy , 2015, Nature Communications.

[18]  P. Senter,et al.  Self-hydrolyzing maleimides improve the stability and pharmacological properties of antibody-drug conjugates , 2014, Nature Biotechnology.

[19]  J. Lambert,et al.  New developments for antibody-drug conjugate-based therapeutic approaches. , 2016, Current opinion in immunology.

[20]  Kim K. Emmerton,et al.  Intracellular Released Payload Influences Potency and Bystander-Killing Effects of Antibody-Drug Conjugates in Preclinical Models. , 2016, Cancer research.

[21]  J. Connors,et al.  Brentuximab vedotin combined with ABVD or AVD for patients with newly diagnosed Hodgkin's lymphoma: a phase 1, open-label, dose-escalation study. , 2013, The Lancet. Oncology.

[22]  S. Kane,et al.  CD30 Downregulation, MMAE Resistance, and MDR1 Upregulation Are All Associated with Resistance to Brentuximab Vedotin , 2015, Molecular Cancer Therapeutics.

[23]  N. McGranahan,et al.  Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. , 2015, Cancer cell.

[24]  Ravi V. J. Chari,et al.  Antikörper‐Wirkstoff‐Konjugate: ein neues Konzept in der Krebstherapie , 2014 .

[25]  R. Advani,et al.  Brentuximab vedotin in the front-line treatment of patients with CD30+ peripheral T-cell lymphomas: results of a phase I study. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

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

[27]  Antoine Maruani,et al.  Corrigendum: Recent advances in the construction of antibody-drug conjugates. , 2016, Nature chemistry.

[28]  C. Bertozzi,et al.  Site-Specific Antibody–Drug Conjugates: The Nexus of Bioorthogonal Chemistry, Protein Engineering, and Drug Development , 2014, Bioconjugate chemistry.

[29]  M. Ferrer,et al.  Formation of disulfide bonds in synthetic peptides and proteins. , 1994, Methods in molecular biology.

[30]  A. Brik,et al.  Efficient Palladium-Assisted One-Pot Deprotection of (Acetamidomethyl)Cysteine Following Native Chemical Ligation and/or Desulfurization To Expedite Chemical Protein Synthesis. , 2016, Angewandte Chemie.