Your browser doesn't support javascript.
loading
Design and Preclinical Evaluation of a Novel B7-H4-Directed Antibody-Drug Conjugate, AZD8205, Alone and in Combination with the PARP1-Selective Inhibitor AZD5305.
Kinneer, Krista; Wortmann, Philipp; Cooper, Zachary A; Dickinson, Niall J; Masterson, Luke; Cailleau, Thais; Hutchinson, Ian; Vijayakrishnan, Balakumar; McFarlane, Mary; Ball, Kathryn; Davies, Michael; Lewis, Arthur; Huang, Yue; Rosenbaum, Anton I; Yuan, Jiaqi; Chesebrough, Jon; Anderton, Judith; Monks, Noel; Novick, Steven; Wang, Jixin; Dimasi, Nazzareno; Christie, R James; Sabol, Darrin; Tosto, Frances Anne; Wallez, Yann; Leo, Elisabetta; Albertella, Mark R; Staniszewska, Anna D; Tice, David A; Howard, Philip W; Luheshi, Nadia; Sapra, Puja.
Afiliação
  • Kinneer K; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Wortmann P; Oncology R&D, AstraZeneca, Munich, Germany.
  • Cooper ZA; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Dickinson NJ; Oncology R&D, AstraZeneca, London, United Kingdom.
  • Masterson L; Oncology R&D, AstraZeneca, London, United Kingdom.
  • Cailleau T; Oncology R&D, AstraZeneca, London, United Kingdom.
  • Hutchinson I; Oncology R&D, AstraZeneca, London, United Kingdom.
  • Vijayakrishnan B; Oncology R&D, AstraZeneca, London, United Kingdom.
  • McFarlane M; BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.
  • Ball K; BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.
  • Davies M; Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
  • Lewis A; BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.
  • Huang Y; BioPharmaceuticals R&D, AstraZeneca, South San Francisco, California.
  • Rosenbaum AI; BioPharmaceuticals R&D, AstraZeneca, South San Francisco, California.
  • Yuan J; BioPharmaceuticals R&D, AstraZeneca, South San Francisco, California.
  • Chesebrough J; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Anderton J; Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
  • Monks N; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Novick S; BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland.
  • Wang J; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Dimasi N; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Christie RJ; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Sabol D; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Tosto FA; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Wallez Y; Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
  • Leo E; Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
  • Albertella MR; Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
  • Staniszewska AD; Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
  • Tice DA; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
  • Howard PW; Oncology R&D, AstraZeneca, London, United Kingdom.
  • Luheshi N; Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
  • Sapra P; Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
Clin Cancer Res ; 29(6): 1086-1101, 2023 03 14.
Article em En | MEDLINE | ID: mdl-36355054
ABSTRACT

PURPOSE:

We evaluated the activity of AZD8205, a B7-H4-directed antibody-drug conjugate (ADC) bearing a novel topoisomerase I inhibitor (TOP1i) payload, alone and in combination with the PARP1-selective inhibitor AZD5305, in preclinical models. EXPERIMENTAL

DESIGN:

IHC and deep-learning-based image analysis algorithms were used to assess prevalence and intratumoral heterogeneity of B7-H4 expression in human tumors. Several TOP1i-ADCs, prepared with Val-Ala or Gly-Gly-Phe-Gly peptide linkers, with or without a PEG8 spacer, were compared in biophysical, in vivo efficacy, and rat toxicology studies. AZD8205 mechanism of action and efficacy studies were conducted in human cancer cell line and patient-derived xenograft (PDX) models.

RESULTS:

Evaluation of IHC-staining density on a per-cell basis revealed a range of heterogeneous B7-H4 expression across patient tumors. This informed selection of bystander-capable Val-Ala-PEG8-TOP1i payload AZ14170133 and development of AZD8205, which demonstrated improved stability, efficacy, and safety compared with other linker-payload ADCs. In a study of 26 PDX tumors, single administration of 3.5 mg/kg AZD8205 provided a 69% overall response rate, according to modified RECIST criteria, which correlated with homologous recombination repair (HRR) deficiency (HRD) and elevated levels of B7-H4 in HRR-proficient models. Addition of AZD5305 sensitized very low B7-H4-expressing tumors to AZD8205 treatment, independent of HRD status and in models representing clinically relevant mechanisms of PARPi resistance.

CONCLUSIONS:

These data provide evidence for the potential utility of AZD8205 for treatment of B7-H4-expressing tumors and support the rationale for an ongoing phase 1 clinical study (NCT05123482). See related commentary by Pommier and Thomas, p. 991.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Imunoconjugados / Neoplasias Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Imunoconjugados / Neoplasias Idioma: En Ano de publicação: 2023 Tipo de documento: Article