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

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

  • Bioconjug Chem. 2016 Aug 17;27(8):1880-8. doi: 10.1021/acs.bioconjchem.6b00291.
Sujiet Puthenveetil Frank Loganzo 1 Haiyin He Ken Dirico Michael Green Jesse Teske Sylvia Musto 1 Tracey Clark Brian Rago Frank Koehn Robert Veneziale 2 Hadi Falahaptisheh 2 Xiaogang Han Frank Barletta Judy Lucas 1 Chakrapani Subramanyam Christopher J O'Donnell L Nathan Tumey Puja Sapra 1 Hans Peter Gerber 1 Dangshe Ma 1 Edmund I Graziani
Affiliations

Affiliations

  • 1 Oncology-Rinat Research & Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10965, United States.
  • 2 Drug Safety Research and Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10977, United States.
Abstract

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.

Figures
Products