PDGFRβ is an essential therapeutic target for BRCA1-deficient mammary tumors

Autor: Xin Hai Pei, Chuying Wang, Xiong Liu, Cheng Fan, Feng Bai, Wei Guo Zhu, Daniel P. Hollern, Charles M. Perou, Li Fu, Shiqin Liu, Li-Han Zhang, Alexandria Scott
Rok vydání: 2021
Předmět:
endocrine system diseases
medicine.medical_treatment
PDGFRβ
Gene Expression
Tumor initiation
medicine.disease_cause
Targeted therapy
Mice
0302 clinical medicine
Surgical oncology
Medicine
Molecular Targeted Therapy
skin and connective tissue diseases
Mice
Knockout

0303 health sciences
Mammary tumor
BRCA1 Protein
EMT
Disease Management
lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens
Immunohistochemistry
Gene Expression Regulation
Neoplastic

030220 oncology & carcinogenesis
Female
Disease Susceptibility
Protein Binding
Signal Transduction
Research Article
Heterozygote
Programmed cell death
Epithelial-Mesenchymal Transition
Breast Neoplasms
lcsh:RC254-282
Receptor
Platelet-Derived Growth Factor beta

03 medical and health sciences
Breast cancer
Cell Line
Tumor

Biomarkers
Tumor

Animals
Cyclin-Dependent Kinase Inhibitor p18
Humans
Cyclin-Dependent Kinase Inhibitor p16
Germ-Line Mutation
030304 developmental biology
business.industry
BRCA1
medicine.disease
Disease Models
Animal

Cancer cell
Cancer research
business
Carcinogenesis
Zdroj: Breast Cancer Research, Vol 23, Iss 1, Pp 1-17 (2021)
Breast Cancer Research : BCR
ISSN: 1465-542X
Popis: Background Basal-like breast cancers (BLBCs) are a leading cause of cancer death due to their capacity to metastasize and lack of effective therapies. More than half of BLBCs have a dysfunctional BRCA1. Although most BRCA1-deficient cancers respond to DNA-damaging agents, resistance and tumor recurrence remain a challenge to survival outcomes for BLBC patients. Additional therapies targeting the pathways aberrantly activated by BRCA1 deficiency are urgently needed. Methods Most BRCA1-deficient BLBCs carry a dysfunctional INK4-RB pathway. Thus, we created genetically engineered mice with Brca1 loss and deletion of p16INK4A, or separately p18INK4C, to model the deficient INK4-RB signaling in human BLBC. By using these mutant mice and human BRCA1-deficient and proficient breast cancer tissues and cells, we tested if there exists a druggable target in BRCA1-deficient breast cancers. Results Heterozygous germline or epithelium-specific deletion of Brca1 in p18INK4C- or p16INK4A-deficient mice activated Pdgfrβ signaling, induced epithelial-to-mesenchymal transition, and led to BLBCs. Confirming this role, targeted deletion of Pdgfrβ in Brca1-deficient tumor cells promoted cell death, induced mesenchymal-to-epithelial transition, and suppressed tumorigenesis. Importantly, we also found that pharmaceutical inhibition of Pdgfrβ and its downstream target Pkcα suppressed Brca1-deficient tumor initiation and progression and effectively killed BRCA1-deficient cancer cells. Conclusions Our work offers the first genetic and biochemical evidence that PDGFRβ-PKCα signaling is repressed by BRCA1, which establishes PDGFRβ-PKCα signaling as a therapeutic target for BRCA1-deficient breast cancers.
Databáze: OpenAIRE