A general approach to site-specific antibody drug conjugates

Significance Here we demonstrate the ability to genetically incorporate nonnative amino acids into proteins in mammalian cells using both transient and stable platform expression systems that provide yields and fidelities compatible with commercial applications. To illustrate the utility of this methodology we have generated chemically homogeneous antibody drug conjugates (NDCs) with precise control over the site and stoichiometry of drug conjugation. In rodent xenograft models these NDCs display improved properties, including half-life, efficacy and safety, relative to conventional heterogeneous ADCs. These advances allow the generation of therapeutic antibody drug conjugates with medicinal chemistry like control over structure, which should greatly facilitate the optimization of their pharmacological activities. Using an expanded genetic code, antibodies with site-specifically incorporated nonnative amino acids were produced in stable cell lines derived from a CHO cell line with titers over 1 g/L. Using anti-5T4 and anti-Her2 antibodies as model systems, site-specific antibody drug conjugates (NDCs) were produced, via oxime bond formation between ketones on the side chain of the incorporated nonnative amino acid and hydroxylamine functionalized monomethyl auristatin D with either protease-cleavable or noncleavable linkers. When noncleavable linkers were used, these conjugates were highly stable and displayed improved in vitro efficacy as well as in vivo efficacy and pharmacokinetic stability in rodent models relative to conventional antibody drug conjugates conjugated through either engineered surface-exposed or reduced interchain disulfide bond cysteine residues. The advantages of the oxime-bonded, site-specific NDCs were even more apparent when low–antigen-expressing (2+) target cell lines were used in the comparative studies. NDCs generated with protease-cleavable linkers demonstrated that the site of conjugation had a significant impact on the stability of these rationally designed prodrug linkers. In a single-dose rat toxicology study, a site-specific anti-Her2 NDC was well tolerated at dose levels up to 90 mg/kg. These experiments support the notion that chemically defined antibody conjugates can be synthesized in commercially relevant yields and can lead to antibody drug conjugates with improved properties relative to the heterogeneous conjugates formed by nonspecific chemical modification.

[1]  M. Sliwkowski,et al.  Clinical Cancer esearch cer Therapy : Preclinical ineered Thio-Trastuzumab-DM 1 Conjugate with an roved Therapeutic Index to Target Human Epidermal R wth Factor Receptor 2 – Positive Breast Cancer , 2010 .

[2]  Kelvin H. Lee,et al.  The genomic sequence of the Chinese hamster ovary (CHO)-K1 cell line , 2011, Nature Biotechnology.

[3]  H. Rugo,et al.  Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[4]  S. Yokoyama,et al.  Adding l-lysine derivatives to the genetic code of mammalian cells with engineered pyrrolysyl-tRNA synthetases. , 2008, Biochemical and biophysical research communications.

[5]  U. RajBhandary,et al.  Complete set of orthogonal 21st aminoacyl-tRNA synthetase-amber, ochre and opal suppressor tRNA pairs: concomitant suppression of three different termination codons in an mRNA in mammalian cells. , 2004, Nucleic acids research.

[6]  B. E. Kimmel,et al.  Optimized clinical performance of growth hormone with an expanded genetic code , 2011, Proceedings of the National Academy of Sciences.

[7]  I. Wistuba,et al.  Delineation of a cellular hierarchy in lung cancer reveals an oncofetal antigen expressed on tumor-initiating cells. , 2011, Cancer research.

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

[9]  E. Hamel,et al.  Sustained intracellular retention of dolastatin 10 causes its potent antimitotic activity. , 2000, Molecular pharmacology.

[10]  Susan E. Cellitti,et al.  Site-specific protein modifications through pyrroline-carboxy-lysine residues , 2011, Proceedings of the National Academy of Sciences.

[11]  M. Sliwkowski,et al.  Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates , 2012, Nature Biotechnology.

[12]  C. J. O’Donnell,et al.  Investigational antibody drug conjugates for solid tumors , 2011, Expert opinion on investigational drugs.

[13]  Damon L. Meyer,et al.  Contribution of linker stability to the activities of anticancer immunoconjugates. , 2008, Bioconjugate chemistry.

[14]  P. Burke,et al.  A potent anti-CD70 antibody-drug conjugate combining a dimeric pyrrolobenzodiazepine drug with site-specific conjugation technology. , 2013, Bioconjugate chemistry.

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

[16]  John M Lambert,et al.  Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. , 2008, Cancer research.

[17]  G. Frantz,et al.  Antibody-drug conjugates for the treatment of non-Hodgkin's lymphoma: target and linker-drug selection. , 2009, Cancer research.

[18]  K. Aldape,et al.  A model of molecular interactions on short oligonucleotide microarrays , 2003, Nature Biotechnology.

[19]  R. Abraham,et al.  Long-term Tumor Regression Induced by an Antibody–Drug Conjugate That Targets 5T4, an Oncofetal Antigen Expressed on Tumor-Initiating Cells , 2012, Molecular Cancer Therapeutics.

[20]  N. Kedersha,et al.  Eradication of large colon tumor xenografts by targeted delivery of maytansinoids. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[21]  Paul Polakis,et al.  Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index , 2008, Nature Biotechnology.

[22]  Carolyn R Bertozzi,et al.  Cu-free click cycloaddition reactions in chemical biology. , 2010, Chemical Society reviews.

[23]  Laurent Ducry,et al.  Antibody-drug conjugates: linking cytotoxic payloads to monoclonal antibodies. , 2010, Bioconjugate chemistry.

[24]  Anna M Wu,et al.  Arming antibodies: prospects and challenges for immunoconjugates , 2005, Nature Biotechnology.

[25]  Peter G Schultz,et al.  An Expanded Eukaryotic Genetic Code , 2003, Science.

[26]  F. Wurm Production of recombinant protein therapeutics in cultivated mammalian cells , 2004, Nature Biotechnology.

[27]  Peter G Schultz,et al.  Adding amino acids to the genetic repertoire. , 2005, Current opinion in chemical biology.

[28]  M. Dorywalska,et al.  Location matters: site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. , 2013, Chemistry & biology.

[29]  P. Polakis Arming antibodies for cancer therapy. , 2005, Current opinion in pharmacology.

[30]  Peter G Schultz,et al.  Synthesis of site-specific antibody-drug conjugates using unnatural amino acids , 2012, Proceedings of the National Academy of Sciences.