scispace - formally typeset
Search or ask a question
Institution

Foundation Medicine

About: Foundation Medicine is a based out in . It is known for research contribution in the topics: Cancer & Targeted therapy. The organization has 430 authors who have published 1085 publications receiving 41789 citations.

Papers published on a yearly basis

Papers
More filters
Journal ArticleDOI
TL;DR: In this article, the authors performed molecular analyses on the residual disease (RD) of 74 clinically-defined triple-negative breast cancers after NAC including next-generation sequencing (NGS) on 20 matched pre-treatment biopsies and combined NGS and digital RNA expression analysis identified diverse molecular lesions and pathway activation in drugresistant tumor cells.
Abstract: Neoadjuvant chemotherapy (NAC) induces a pathologic complete response (pCR) in approximately 30% of patients with triple-negative breast cancers (TNBC). In patients lacking a pCR, NAC selects a subpopulation of chemotherapy-resistant tumor cells. To understand the molecular underpinnings driving treatment-resistant TNBCs, we performed comprehensive molecular analyses on the residual disease (RD) of 74 clinically-defined TNBCs after NAC including next-generation sequencing (NGS) on 20 matched pre-treatment biopsies. Combined NGS and digital RNA expression analysis identified diverse molecular lesions and pathway activation in drug-resistant tumor cells. Ninety percent of the tumors contained a genetic alteration potentially treatable with a currently available targeted therapy. Thus, profiling residual TNBCs after NAC identifies targetable molecular lesions in the chemotherapy-resistant component of the tumor which may mirror micro-metastases destined to recur clinically. These data can guide biomarker-driven adjuvant studies targeting these micro-metastases to improve the outcome of patients with TNBC who do not respond completely to NAC.

412 citations

Journal ArticleDOI
TL;DR: The possibility that Ras–MAPK pathway activation promotes immune-evasion in TNBC is suggested, and clinical trials combining MEK- and PD-L1–targeted therapies are support, and Ras/MAPK activation and MHC expression may be predictive biomarkers of response to immune checkpoint inhibitors.
Abstract: Purpose: Tumor-infiltrating lymphocytes (TILs) in the residual disease (RD) of triple-negative breast cancers (TNBCs) after neoadjuvant chemotherapy (NAC) are associated with improved survival, but insight into tumor cell-autonomous molecular pathways affecting these features are lacking. Experimental Design: We analyzed TILs in the RD of clinically and molecularly characterized TNBCs after NAC and explored therapeutic strategies targeting combinations of MEK inhibitors with PD-1/PD-L1-targeted immunotherapy in mouse models of breast cancer. Results: Presence of TILs in the RD was significantly associated with improved prognosis. Genetic or transcriptomic alterations in Ras/MAPK signaling were significantly correlated with lower TILs. MEK inhibition up-regulated cell-surface major histocompatibility complex (MHC) expression and PD-L1 in TNBC cells both in vivo and in vitro. Moreover, combined MEK and PDL-1/PD-1 inhibition enhanced anti-tumor immune responses in mouse models of breast cancer. Conclusions: These data suggest the possibility that Ras/MAPK pathway activation promotes immune-evasion in TNBC, and support clinical trials combining MEK- and PD-L1-targeted therapies. Furthermore, Ras/MAPK activation and MHC expression may be predictive biomarkers of response to immune checkpoint inhibitors.

410 citations

Journal ArticleDOI
TL;DR: Targeting of a larger fraction of identified molecular alterations, yielding a higher ‘matching score’, was correlated with significantly improved disease control rates, as well as longer progression-free and overall survival rates, compared to targeting of fewer somatic alterations.
Abstract: Cancer treatments have evolved from indiscriminate cytotoxic agents to selective genome- and immune-targeted drugs that have transformed the outcomes of some malignancies1. Tumor complexity and heterogeneity suggest that the 'precision medicine' paradigm of cancer therapy requires treatment to be personalized to the individual patient2-6. To date, precision oncology trials have been based on molecular matching with predetermined monotherapies7-14. Several of these trials have been hindered by very low matching rates, often in the 5-10% range15, and low response rates. Low matching rates may be due to the use of limited gene panels, restrictive molecular matching algorithms, lack of drug availability, or the deterioration and death of end-stage patients before therapy can be implemented. We hypothesized that personalized treatment with combination therapies would improve outcomes in patients with refractory malignancies. As a first test of this concept, we implemented a cross-institutional prospective study (I-PREDICT, NCT02534675 ) that used tumor DNA sequencing and timely recommendations for individualized treatment with combination therapies. We found that administration of customized multidrug regimens was feasible, with 49% of consented patients receiving personalized treatment. Targeting of a larger fraction of identified molecular alterations, yielding a higher 'matching score', was correlated with significantly improved disease control rates, as well as longer progression-free and overall survival rates, compared to targeting of fewer somatic alterations. Our findings suggest that the current clinical trial paradigm for precision oncology, which pairs one driver mutation with one drug, may be optimized by treating molecularly complex and heterogeneous cancers with combinations of customized agents.

391 citations

Journal ArticleDOI
TL;DR: TMB appears to be an important independent biomarker within MSI-H mCRC to stratify patients for likelihood of response to ICPIs if validated in prospective studies, and may play an important role in guiding the sequencing and/or combinations of ICPI in MSI-h mC RC.

390 citations

Journal ArticleDOI
TL;DR: Melanomas with NF1 mutations harbored high mutational loads and high response rates, whereas BRAF/NRAS/NF1 wild-type melanomas had a lower mutational load, and TCR clonality did not predict response.
Abstract: Therapeutic antibodies blocking programmed death-1 and its ligand (PD-1/PD-L1) induce durable responses in a substantial fraction of melanoma patients. We sought to determine whether the number and/or type of mutations identified using a next-generation sequencing (NGS) panel available in the clinic was correlated with response to anti-PD-1 in melanoma. Using archival melanoma samples from anti-PD-1/PD-L1-treated patients, we performed hybrid capture-based NGS on 236-315 genes and T-cell receptor (TCR) sequencing on initial and validation cohorts from two centers. Patients who responded to anti-PD-1/PD-L1 had higher mutational loads in an initial cohort (median, 45.6 vs. 3.9 mutations/MB; P = 0.003) and a validation cohort (37.1 vs. 12.8 mutations/MB; P = 0.002) compared with nonresponders. Response rate, progression-free survival, and overall survival were superior in the high, compared with intermediate and low, mutation load groups. Melanomas with NF1 mutations harbored high mutational loads (median, 62.7 mutations/MB) and high response rates (74%), whereas BRAF/NRAS/NF1 wild-type melanomas had a lower mutational load. In these archival samples, TCR clonality did not predict response. Mutation numbers in the 315 genes in the NGS platform strongly correlated with those detected by whole-exome sequencing in The Cancer Genome Atlas samples, but was not associated with survival. In conclusion, mutational load, as determined by an NGS platform available in the clinic, effectively stratified patients by likelihood of response. This approach may provide a clinically feasible predictor of response to anti-PD-1/PD-L1. Cancer Immunol Res; 4(11); 959-67. ©2016 AACR.

388 citations


Authors

Showing all 430 results

NameH-indexPapersCitations
Bartolome R. Celli11865063423
Vincent A. Miller10564765822
Vera J. Suman7629435533
Adam Brufsky7446027570
Jeffrey S. Ross7034721334
Philip J. Stephens6923629559
Kai Wang69109521841
Siraj M. Ali6546325639
Jeffrey S. Ross5934320600
Roman Yelensky5716415073
Doron Lipson5718017014
Garrett M. Frampton5521522373
Geoffrey R. Oxnard5424514325
Brian M. Alexander502649200
Jeffrey P. Gregg461074882
Network Information
Related Institutions (5)
University of Texas MD Anderson Cancer Center
92.5K papers, 4.7M citations

94% related

Memorial Sloan Kettering Cancer Center
65.3K papers, 4.4M citations

92% related

Roswell Park Cancer Institute
19.5K papers, 944.8K citations

91% related

City of Hope National Medical Center
16.4K papers, 755.4K citations

91% related

The Royal Marsden NHS Foundation Trust
13.4K papers, 668.8K citations

90% related

Performance
Metrics
No. of papers from the Institution in previous years
YearPapers
2021137
2020144
2019138
2018142
2017193
2016112