Your browser doesn't support javascript.
loading
Intracellular Released Payload Influences Potency and Bystander-Killing Effects of Antibody-Drug Conjugates in Preclinical Models.
Li, Fu; Emmerton, Kim K; Jonas, Mechthild; Zhang, Xinqun; Miyamoto, Jamie B; Setter, Jocelyn R; Nicholas, Nicole D; Okeley, Nicole M; Lyon, Robert P; Benjamin, Dennis R; Law, Che-Leung.
Affiliation
  • Li F; Preclinical Science, Seattle Genetics, Inc., Bothell, Washington. fli@seagen.com.
  • Emmerton KK; Preclinical Science, Seattle Genetics, Inc., Bothell, Washington.
  • Jonas M; Translational Research, Seattle Genetics, Inc., Bothell, Washington.
  • Zhang X; Chemistry, Seattle Genetics, Inc., Bothell, Washington.
  • Miyamoto JB; Preclinical Science, Seattle Genetics, Inc., Bothell, Washington.
  • Setter JR; Chemistry, Seattle Genetics, Inc., Bothell, Washington.
  • Nicholas ND; Experimental Pharmacology, Seattle Genetics, Inc., Bothell, Washington.
  • Okeley NM; Chemistry, Seattle Genetics, Inc., Bothell, Washington.
  • Lyon RP; Chemistry, Seattle Genetics, Inc., Bothell, Washington.
  • Benjamin DR; Translational Research, Seattle Genetics, Inc., Bothell, Washington.
  • Law CL; Preclinical Science, Seattle Genetics, Inc., Bothell, Washington.
Cancer Res ; 76(9): 2710-9, 2016 05 01.
Article in En | MEDLINE | ID: mdl-26921341
ABSTRACT
Antibody-drug conjugates (ADC) comprise targeting antibodies armed with potent small-molecule payloads. ADCs demonstrate specific cell killing in clinic, but the basis of their antitumor activity is not fully understood. In this study, we investigated the degree to which payload release predicts ADC activity in vitro and in vivo ADCs were generated to target different receptors on the anaplastic large cell lymphoma line L-82, but delivered the same cytotoxic payload (monomethyl auristatin E, MMAE), and we found that the intracellular concentration of released MMAE correlated with in vitro ADC-mediated cytotoxicity independent of target expression or drugantibody ratios. Intratumoral MMAE concentrations consistently correlated with the extent of tumor growth inhibition in tumor xenograft models. In addition, we developed a robust admixed tumor model consisting of CD30(+) and CD30(-) cancer cells to study how heterogeneity of target antigen expression, a phenomenon often observed in cancer specimens, affects the treatment response. CD30-targeting ADC delivering membrane permeable MMAE or pyrrolobenzodiazepine dimers demonstrated potent bystander killing of neighboring CD30(-) cells. In contrast, a less membrane permeable payload, MMAF, failed to mediate bystander killing in vivo, suggesting local diffusion and distribution of released payloads represents a potential mechanism of ADC-mediated bystander killing. Collectively, our findings establish that the biophysical properties and amount of released payloads are chief factors determining the overall ADC potency and bystander killing. Cancer Res; 76(9); 2710-9. ©2016 AACR.
Subject(s)

Full text: 1 Database: MEDLINE Main subject: Oligopeptides / Immunoconjugates / Antibodies, Monoclonal / Antineoplastic Agents Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Cancer Res Year: 2016 Type: Article

Full text: 1 Database: MEDLINE Main subject: Oligopeptides / Immunoconjugates / Antibodies, Monoclonal / Antineoplastic Agents Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Cancer Res Year: 2016 Type: Article