학술논문

Neutralization of BCL-2/XL Enhances the Cytotoxicity of T-DM1 In Vivo
Document Type
article
Source
Molecular Cancer Therapeutics. 18(6)
Subject
Biomedical and Clinical Sciences
Oncology and Carcinogenesis
Breast Cancer
Cancer
Ado-Trastuzumab Emtansine
Aniline Compounds
Animals
Antineoplastic Agents
Immunological
Apoptosis
Breast Neoplasms
Cytotoxicity
Immunologic
Drug Resistance
Neoplasm
Drug Therapy
Combination
Female
Humans
Immunoconjugates
Mice
Mice
Inbred NOD
Mice
SCID
Proto-Oncogene Proteins c-bcl-2
Receptor
ErbB-2
Sulfonamides
Xenograft Model Antitumor Assays
bcl-X Protein
Receptor
erbB-2
Pharmacology and Pharmaceutical Sciences
Oncology & Carcinogenesis
Biochemistry and cell biology
Oncology and carcinogenesis
Language
Abstract
One of the most recent advances in the treatment of HER2+ breast cancer is the development of the antibody-drug conjugate, T-DM1. T-DM1 has proven clinical benefits for patients with advanced and/or metastatic breast cancer who have progressed on prior HER2-targeted therapies. However, T-DM1 resistance ultimately occurs and represents a major obstacle in the effective treatment of this disease. Because anti-apoptotic BCL-2 family proteins can affect the threshold for induction of apoptosis and thus limit the effectiveness of the chemotherapeutic payload, we examined whether inhibition of BCL-2/XL would enhance the efficacy of T-DM1 in five HER2-expressing patient-derived breast cancer xenograft models. Inhibition of BCL-2/XL via navitoclax/ABT-263 significantly enhanced the cytotoxicity of T-DM1 in two of three models derived from advanced and treatment-exposed metastatic breast tumors. No additive effects of combined treatment were observed in the third metastatic tumor model, which was highly sensitive to T-DM1, as well as a primary treatment-exposed tumor, which was refractory to T-DM1. A fifth model, derived from a treatment naïve primary breast tumor, was sensitive to T-DM1 but markedly benefited from combination treatment. Notably, both PDXs that were highly responsive to the combination therapy expressed low HER2 protein levels and lacked ERBB2 amplification, suggesting that BCL-2/XL inhibition can enhance sensitivity of tumors with low HER2 expression. Toxicities associated with combined treatments were significantly ameliorated with intermittent ABT-263 dosing. Taken together, these studies provide evidence that T-DM1 cytotoxicity could be significantly enhanced via BCL-2/XL blockade and support clinical investigation of this combination beyond ERBB2-amplified and/or HER2-overexpressed tumors.