Natural Product Splicing Inhibitors: A New Class of Antibody-Drug Conjugate (ADC) Payloads.

There is a considerable ongoing work to identify new cytotoxic payloads that are appropriate for antibody-based delivery, acting via mechanisms beyond DNA damage and microtubule disruption, highlighting their importance to the field of cancer therapeutics. New modes of action will allow a more diverse set of tumor types to be targeted and will allow for possible mechanisms to evade the drug resistance that will invariably develop to existing payloads. Spliceosome inhibitors are known to be potent antiproliferative agents capable of targeting both actively dividing and quiescent cells. A series of thailanstatin-antibody conjugates were prepared in order to evaluate their potential utility in the treatment of cancer. After exploring a variety of linkers, we found that the most potent antibody-drug conjugates (ADCs) were derived from direct conjugation of the carboxylic acid-containing payload to surface lysines of the antibody (a "linker-less" conjugate). Activity of these lysine conjugates was correlated to drug-loading, a feature not typically observed for other payload classes. The thailanstatin-conjugates were potent in high target expressing cells, including multidrug-resistant lines, and inactive in nontarget expressing cells. Moreover, these ADCs were shown to promote altered splicing products in N87 cells in vitro, consistent with their putative mechanism of action. In addition, the exposure of the ADCs was sufficient to result in excellent potency in a gastric cancer xenograft model at doses as low as 1.5 mg/kg that was superior to the clinically approved ADC T-DM1. The results presented herein therefore open the door to further exploring splicing inhibition as a potential new mode-of-action for novel ADCs.

[1]  F. Sharom,et al.  ABC efflux pump-based resistance to chemotherapy drugs. , 2009, Chemical reviews.

[2]  C. J. O’Donnell,et al.  Spliceostatin hemiketal biosynthesis in Burkholderia spp. is catalyzed by an iron/α-ketoglutarate–dependent dioxygenase , 2014, Proceedings of the National Academy of Sciences.

[3]  F. Loganzo,et al.  Cytotoxic Spliceostatins from Burkholderia sp. and Their Semisynthetic Analogues. , 2014, Journal of natural products.

[4]  M. Jurica,et al.  Spliceostatin A inhibits spliceosome assembly subsequent to prespliceosome formation , 2010, Nucleic acids research.

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

[6]  Xiangyang Liu,et al.  Genomics-guided discovery of thailanstatins A, B, and C As pre-mRNA splicing inhibitors and antiproliferative agents from Burkholderia thailandensis MSMB43. , 2013, Journal of natural products.

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

[8]  Ana T Menendez,et al.  An anti-MUC1 antibody-calicheamicin conjugate for treatment of solid tumors. Choice of linker and overcoming drug resistance. , 2005, Bioconjugate chemistry.

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

[10]  R. Ubink,et al.  Preclinical Profile of the HER2-Targeting ADC SYD983/SYD985: Introduction of a New Duocarmycin-Based Linker-Drug Platform , 2014, Molecular Cancer Therapeutics.

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

[12]  F. J. Ramos,et al.  Phase I Pharmacokinetic and Pharmacodynamic Study of the First-in-Class Spliceosome Inhibitor E7107 in Patients with Advanced Solid Tumors , 2013, Clinical Cancer Research.

[13]  Gerhard Moldenhauer,et al.  Therapeutic potential of amanitin-conjugated anti-epithelial cell adhesion molecule monoclonal antibody against pancreatic carcinoma. , 2012, Journal of the National Cancer Institute.

[14]  Frank E Koehn,et al.  Biosynthetic engineering and fermentation media development leads to gram-scale production of spliceostatin natural products in Burkholderia sp. , 2016, Metabolic engineering.

[15]  Mallika Singh,et al.  Targeting LGR5+ cells with an antibody-drug conjugate for the treatment of colon cancer , 2015, Science Translational Medicine.

[16]  D. Kroll,et al.  Design, synthesis, and biological evaluation of antibody-drug conjugates comprised of potent camptothecin analogues. , 2009, Bioconjugate chemistry.

[17]  Jennifer B. Webster,et al.  Engineered antibody-drug conjugates with defined sites and stoichiometries of drug attachment. , 2006, Protein engineering, design & selection : PEDS.

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

[19]  I. Bernstein,et al.  SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML. , 2013, Blood.

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

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

[22]  P. Trail,et al.  Site‐directed delivery of anthracyclines for treatment of cancer , 1995 .

[23]  M. Hagiwara,et al.  Spliceostatin A targets SF3b and inhibits both splicing and nuclear retention of pre-mRNA , 2007, Nature Chemical Biology.

[24]  Rajeeva Singh,et al.  Synthesis and evaluation of hydrophilic linkers for antibody-maytansinoid conjugates. , 2011, Journal of medicinal chemistry.