학술논문

Activating NOTCH1 Mutations Define a Distinct Subgroup of Patients With Adenoid Cystic Carcinoma Who Have Poor Prognosis, Propensity to Bone and Liver Metastasis, and Potential Responsiveness to Notch1 Inhibitors
Document Type
article
Source
Journal of Clinical Oncology. 35(3)
Subject
Biomedical and Clinical Sciences
Clinical Sciences
Oncology and Carcinogenesis
Cancer
Pediatric Cancer
Digestive Diseases
Rare Diseases
Clinical Research
Pediatric
Dental/Oral and Craniofacial Disease
Genetics
Aetiology
2.1 Biological and endogenous factors
Adult
Aged
Animals
Antineoplastic Agents
Biomarkers
Tumor
Bone Neoplasms
Carcinoma
Adenoid Cystic
DNA Mutational Analysis
Disease-Free Survival
Female
Genetic Predisposition to Disease
HEK293 Cells
Humans
Immunohistochemistry
Kaplan-Meier Estimate
Liver Neoplasms
Male
Mice
Nude
Middle Aged
Molecular Targeted Therapy
Mutation
Phenotype
Predictive Value of Tests
Proportional Hazards Models
Receptor
Notch1
Retrospective Studies
Risk Factors
Salivary Gland Neoplasms
Time Factors
Transfection
Treatment Outcome
Xenograft Model Antitumor Assays
Oncology & Carcinogenesis
Oncology and carcinogenesis
Language
Abstract
Purpose Adenoid cystic carcinomas (ACCs) represent a heterogeneous group of chemotherapy refractory tumors, with a subset demonstrating an aggressive phenotype. We investigated the molecular underpinnings of this phenotype and assessed the Notch1 pathway as a potential therapeutic target. Methods We genotyped 102 ACCs that had available pathologic and clinical data. Notch1 activation was assessed by immunohistochemistry for Notch1 intracellular domain. Luciferase reporter assays were used to confirm Notch1 target gene expression in vitro. The Notch1 inhibitor brontictuzumab was tested in patient-derived xenografts from patients with ACC and in a patient with ACC who was enrolled in a phase I study. Results NOTCH1 mutations occurred predominantly (14 of 15 patients) in the negative regulatory region and Pro-Glu-Ser-Thr-rich domains, the same two hotspots seen in T-cell acute lymphoblastic leukemias, and led to pathway activation in vitro. NOTCH1-mutant tumors demonstrated significantly higher levels of Notch1 pathway activation than wild-type tumors on the basis of Notch1 intracellular domain staining ( P = .004). NOTCH1 mutations define a distinct aggressive ACC subgroup with a significantly higher likelihood of solid subtype ( P < .001), advanced-stage disease at diagnosis ( P = .02), higher rate of liver and bone metastasis ( P ≤ .02), shorter relapse-free survival (median, 13 v 34 months; P = .01), and shorter overall survival (median 30 v 122 months; P = .001) when compared with NOTCH1 wild-type tumors. Significant tumor growth inhibition with brontictuzumab was observed exclusively in the ACC patient-derived xenograft model that harbored a NOTCH1 activating mutation. Furthermore, an index patient with NOTCH1-mutant ACC had a partial response to brontictuzumab. Conclusion NOTCH1 mutations define a distinct disease phenotype characterized by solid histology, liver and bone metastasis, poor prognosis, and potential responsiveness to Notch1 inhibitors. Clinical studies targeting Notch1 in a genotype-defined ACC subgroup are warranted.