Tim Brennan
Foundation Medicine
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Tim Brennan.
Cancer Discovery | 2015
Garrett Michael Frampton; Siraj M. Ali; Mark Rosenzweig; Juliann Chmielecki; Xinyuan Lu; Todd Michael Bauer; Mikhail Akimov; Jose A. Bufill; Carrie B. Lee; David Jentz; Rick Hoover; Sai-Hong Ignatius Ou; Ravi Salgia; Tim Brennan; Zachary R. Chalmers; Savina Jaeger; Alan Huang; Julia A. Elvin; Rachel L. Erlich; Alex Fichtenholtz; Kyle Gowen; Joel Greenbowe; Adrienne Johnson; Depinder Khaira; Caitlin McMahon; Eric M. Sanford; Steven Roels; Jared White; Joel Greshock; Robert Schlegel
UNLABELLED Focal amplification and activating point mutation of the MET gene are well-characterized oncogenic drivers that confer susceptibility to targeted MET inhibitors. Recurrent somatic splice site alterations at MET exon 14 (METex14) that result in exon skipping and MET activation have been characterized, but their full diversity and prevalence across tumor types are unknown. Here, we report analysis of tumor genomic profiles from 38,028 patients to identify 221 cases with METex14 mutations (0.6%), including 126 distinct sequence variants. METex14 mutations are detected most frequently in lung adenocarcinoma (3%), but also frequently in other lung neoplasms (2.3%), brain glioma (0.4%), and tumors of unknown primary origin (0.4%). Further in vitro studies demonstrate sensitivity to MET inhibitors in cells harboring METex14 alterations. We also report three new patient cases with METex14 alterations in lung or histiocytic sarcoma tumors that showed durable response to two different MET-targeted therapies. The diversity of METex14 mutations indicates that diagnostic testing via comprehensive genomic profiling is necessary for detection in a clinical setting. SIGNIFICANCE Here we report the identification of diverse exon 14 splice site alterations in MET that result in constitutive activity of this receptor and oncogenic transformation in vitro. Patients whose tumors harbored these alterations derived meaningful clinical benefit from MET inhibitors. Collectively, these data support the role of METex14 alterations as drivers of tumorigenesis, and identify a unique subset of patients likely to derive benefit from MET inhibitors.
Genome Medicine | 2013
Matthias Lechner; Garrett Michael Frampton; Tim Fenton; Andrew Feber; Gary A. Palmer; Amrita Jay; Nischalan Pillay; Martin Forster; Maureen T. Cronin; Doron Lipson; Vincent A. Miller; Tim Brennan; Stephen Henderson; Francis Vaz; Paul O'Flynn; Nicholas Kalavrezos; Roman Yelensky; Stephan Beck; Philip J. Stephens; Chris Boshoff
BackgroundHuman papillomavirus positive (HPV+) head and neck squamous cell carcinoma (HNSCC) is an emerging disease, representing a distinct clinical and epidemiological entity. Understanding the genetic basis of this specific subtype of cancer could allow therapeutic targeting of affected pathways for a stratified medicine approach.MethodsTwenty HPV+ and 20 HPV- laser-capture microdissected oropharyngeal carcinomas were used for paired-end sequencing of hybrid-captured DNA, targeting 3,230 exons in 182 genes often mutated in cancer. Copy number alteration (CNA) profiling, Sequenom MassArray sequencing and immunohistochemistry were used to further validate findings.ResultsHPV+ and HPV- oropharyngeal carcinomas cluster into two distinct subgroups. TP53 mutations are detected in 100% of HPV negative cases and abrogation of the G1/S checkpoint by CDKN2A/B deletion and/or CCND1 amplification occurs in the majority of HPV- tumors.ConclusionThese findings strongly support a causal role for HPV, acting via p53 and RB pathway inhibition, in the pathogenesis of a subset of oropharyngeal cancers and suggest that studies of CDK inhibitors in HPV- disease may be warranted. Mutation and copy number alteration of PI3 kinase (PI3K) pathway components appears particularly prevalent in HPV+ tumors and assessment of these alterations may aid in the interpretation of current clinical trials of PI3K, AKT, and mTOR inhibitors in HNSCC.
European Urology | 2015
Elizabeth R. Plimack; Roland L. Dunbrack; Tim Brennan; Mark Andrake; Yan Zhou; Ilya G. Serebriiskii; Michael Slifker; Katherine Alpaugh; Essel Dulaimi; Norma Alonzo Palma; Jean H. Hoffman-Censits; Marijo Bilusic; Yu Ning Wong; Alexander Kutikov; Rosalia Viterbo; Richard E. Greenberg; David Y.T. Chen; Edouard J. Trabulsi; Roman Yelensky; David J. McConkey; Vincent A. Miller; Erica A. Golemis; Eric A. Ross
BACKGROUND Cisplatin-based neoadjuvant chemotherapy (NAC) before cystectomy is the standard of care for muscle-invasive bladder cancer (MIBC), with 25-50% of patients expected to achieve a pathologic response. Validated biomarkers predictive of response are currently lacking. OBJECTIVE To discover and validate biomarkers predictive of response to NAC for MIBC. DESIGN, SETTING, AND PARTICIPANTS Pretreatment MIBC samples prospectively collected from patients treated in two separate clinical trials of cisplatin-based NAC provided the discovery and validation sets. DNA from pretreatment tumor tissue was sequenced for all coding exons of 287 cancer-related genes and was analyzed for base substitutions, indels, copy number alterations, and selected rearrangements in a Clinical Laboratory Improvements Amendments-certified laboratory. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The mean number of variants and variant status for each gene were correlated with response. Variant data from the discovery cohort were used to create a classification tree to discriminate responders from nonresponders. The resulting decision rule was then tested in the independent validation set. RESULTS AND LIMITATIONS Patients with a pathologic complete response had more alterations than those with residual tumor in both the discovery (p=0.024) and validation (p=0.018) sets. In the discovery set, alteration in one or more of the three DNA repair genes ATM, RB1, and FANCC predicted pathologic response (p<0.001; 87% sensitivity, 100% specificity) and better overall survival (p=0.007). This test remained predictive for pathologic response in the validation set (p=0.033), with a trend towards better overall survival (p=0.055). These results require further validation in additional sample sets. CONCLUSIONS Genomic alterations in the DNA repair-associated genes ATM, RB1, and FANCC predict response and clinical benefit after cisplatin-based chemotherapy for MIBC. The results suggest that defective DNA repair renders tumors sensitive to cisplatin. PATIENT SUMMARY Chemotherapy given before bladder removal (cystectomy) improves the chance of cure for some but not all patients with muscle-invasive bladder cancer. We found a set of genetic mutations that when present in tumor tissue predict benefit from neoadjuvant chemotherapy, suggesting that testing before chemotherapy may help in selecting patients for whom this approach is recommended.
Journal of Clinical Oncology | 2014
Elizabeth R. Plimack; Jean H. Hoffman-Censits; Rosalia Viterbo; Edouard J. Trabulsi; Eric A. Ross; Richard E. Greenberg; David Y.T. Chen; Yu Ning Wong; Jianqing Lin; Alexander Kutikov; Efrat Dotan; Tim Brennan; Norma Alonzo Palma; Essel Dulaimi; Reza Mehrazin; Stephen A. Boorjian; William Kevin Kelly; Robert G. Uzzo; Gary R. Hudes
PURPOSE Neoadjuvant cisplatin-based chemotherapy is standard of care for muscle-invasive bladder cancer (MIBC); however, it is infrequently adopted in practice because of concerns regarding toxicity and delay to cystectomy. We hypothesized that three cycles of neoadjuvant accelerated methotrexate, vinblastine, doxorubicin, and cisplatin (AMVAC) would be safe, shorten the time to surgery, and yield similar pathologic complete response (pT0) rates compared with historical controls. PATIENTS AND METHODS Patients with cT2-T4a and N0-N1 MIBC were eligible and received three cycles of AMVAC with pegfilgrastim followed by radical cystectomy with lymph node dissection. The primary end point was pT0 rate. Telomere length (TL) and p53 mutation status were correlated with response and toxicity. RESULTS Forty-four patients were accrued; 60% had stage III to IV disease; median age was 64 years. Forty patients were evaluable for response, with 15 (38%; 95% CI, 23% to 53%) showing pT0 at cystectomy, meeting the primary end point of the study. Another six patients (14%) were downstaged to non-muscle invasive disease. Most (82%) experienced only grade 1 to 2 treatment-related toxicities. There were no grade 3 or 4 renal toxicities and no treatment-related deaths. One patient developed metastases and thus did not undergo cystectomy; all others (n = 43) proceeded to cystectomy within 8 weeks after last chemotherapy administration. Median time from start of chemotherapy to cystectomy was 9.7 weeks. TL and p53 mutation did not predict response or toxicity. CONCLUSION AMVAC is well tolerated and results in similar pT0 rates with 6 weeks of treatment compared with standard 12-week regimens. Further analysis is ongoing to ascertain whether molecular alterations in tumor samples can predict response to chemotherapy.
Blood | 2016
Jie He; Omar Abdel-Wahab; Michelle Nahas; Kai Wang; Raajit Rampal; Andrew M. Intlekofer; Jay Patel; Andrei V. Krivstov; Garrett Michael Frampton; Lauren E. Young; Shan Zhong; Mark Bailey; Jared White; Steven Roels; Jason Deffenbaugh; Alex Fichtenholtz; Tim Brennan; Mark Rosenzweig; Kimberly Pelak; Kristina M. Knapp; Kristina Brennan; Amy Donahue; Geneva Young; Lazaro Garcia; Selmira T. Beckstrom; Mandy Zhao; Emily White; Vera Banning; Jamie Buell; Kiel Iwanik
The spectrum of somatic alterations in hematologic malignancies includes substitutions, insertions/deletions (indels), copy number alterations (CNAs), and a wide range of gene fusions; no current clinically available single assay captures the different types of alterations. We developed a novel next-generation sequencing-based assay to identify all classes of genomic alterations using archived formalin-fixed paraffin-embedded blood and bone marrow samples with high accuracy in a clinically relevant time frame, which is performed in our Clinical Laboratory Improvement Amendments-certified College of American Pathologists-accredited laboratory. Targeted capture of DNA/RNA and next-generation sequencing reliably identifies substitutions, indels, CNAs, and gene fusions, with similar accuracy to lower-throughput assays that focus on specific genes and types of genomic alterations. Profiling of 3696 samples identified recurrent somatic alterations that impact diagnosis, prognosis, and therapy selection. This comprehensive genomic profiling approach has proved effective in detecting all types of genomic alterations, including fusion transcripts, which increases the ability to identify clinically relevant genomic alterations with therapeutic relevance.
Journal of Hematology & Oncology | 2015
Vivek Subbiah; Caitlin McMahon; Shreyaskumar Patel; Ralph Zinner; Elvio G. Silva; Julia A. Elvin; Ishwaria M. Subbiah; Chimela Ohaji; Dhakshina Moorthy Ganeshan; Deepa Anand; Charles Levenback; Jenny Berry; Tim Brennan; Juliann Chmielecki; Zachary R. Chalmers; John Mayfield; Vincent A. Miller; Philip J. Stephens; Jeffrey S. Ross; Siraj M. Ali
BackgroundRecurrent, metastatic mesenchymal myxoid tumors of the gynecologic tract present a management challenge as there is minimal evidence to guide systemic therapy. Such tumors also present a diagnostic dilemma, as myxoid features are observed in leiomyosarcomas, inflammatory myofibroblastic tumors (IMT), and mesenchymal myxoid tumors. Comprehensive genomic profiling was performed in the course of clinical care on a case of a recurrent, metastatic myxoid uterine malignancy (initially diagnosed as smooth muscle tumor of uncertain malignant potential (STUMP)), to guide identify targeted therapeutic options. To our knowledge, this case represents the first report of clinical response to targeted therapy in a tumor harboring a DCTN1-ALK fusion protein.MethodsHybridization capture of 315 cancer-related genes plus introns from 28 genes often rearranged or altered in cancer was applied to >50 ng of DNA extracted from this sample and sequenced to high, uniform coverage. Therapy was given in the context of a phase I clinical trial ClinicalTrials.gov Identifier: (NCT01548144).ResultsImmunostains showed diffuse positivity for ALK1 expression and comprehensive genomic profiling identified an in frame DCTN1-ALK gene fusion. The diagnosis of STUMP was revised to that of an IMT with myxoid features. The patient was enrolled in a clinical trial and treated with an anaplastic lymphoma kinase (ALK) inhibitor (crizotinib/Xalkori®) and a multikinase VEGF inhibitor (pazopanib/Votrient®). The patient experienced an ongoing partial response (6+ months) by response evaluation criteria in solid tumors (RECIST) 1.1 criteria.ConclusionsFor myxoid tumors of the gynecologic tract, comprehensive genomic profiling can identify clinical relevant genomic alterations that both direct treatment targeted therapy and help discriminate between similar diagnostic entities.
Leukemia | 2017
Kai Wang; M Sanchez-Martin; Xujun Wang; Kristina M. Knapp; Richard Koche; Ly P. Vu; Michelle Nahas; Jie He; Michael Hadler; Eytan M. Stein; Martin S. Tallman; Amy Donahue; Garrett Michael Frampton; Doron Lipson; Steven Roels; Phil Stephens; E M Sanford; Tim Brennan; Geoff Otto; Roman Yelensky; V.A. Miller; Michael G. Kharas; Ross L. Levine; Adolfo A. Ferrando; Scott A. Armstrong; Andrei V. Krivtsov
Genomic studies have identified recurrent somatic mutations in acute leukemias. However, current murine models do not sufficiently encompass the genomic complexity of human leukemias. To develop preclinical models, we transplanted 160 samples from patients with acute leukemia (acute myeloid leukemia, mixed lineage leukemia, B-cell acute lymphoblastic leukemia, T-cell ALL) into immunodeficient mice. Of these, 119 engrafted with expected immunophenotype. Targeted sequencing of 374 genes and 265 frequently rearranged RNAs detected recurrent and novel genetic lesions in 48 paired primary tumor (PT) and patient-derived xenotransplant (PDX) samples. Overall, the frequencies of 274 somatic variant alleles correlated between PT and PDX samples, although the data were highly variable for variant alleles present at 0–10%. Seventeen percent of variant alleles were detected in either PT or PDX samples only. Based on variant allele frequency changes, 24 PT-PDX pairs were classified as concordant while the other 24 pairs showed various degree of clonal discordance. There was no correlation of clonal concordance with clinical parameters of diseases. Significantly more bone marrow samples than peripheral blood samples engrafted discordantly. These data demonstrate the utility of developing PDX banks for modeling human leukemia, and emphasize the importance of genomic profiling of PDX and patient samples to ensure concordance before performing mechanistic or therapeutic studies.
Cancer Research | 2015
Elizabeth R. Plimack; Roland L. Dunbrack; Tim Brennan; Mark Andrake; Yan Zhou; Ilya G. Serebriiskii; Essel Dulaimi Al-Saleem; Jean H. Hoffman-Censits; Marijo Bilusic; Yu-Ning Wong; Alexander Kutikov; Rosalia Viterbo; Richard E. Greenberg; David Y.T. Chen; Edouard J. Trabulsi; Roman Yelensky; Vincent A. Miller; Erica A. Golemis; Eric A. Ross
Background: Cisplatin based NAC prior to cystectomy is standard of care for MIBC, with 40-50% expected to respond with ≤pT1N0M0. Biomarkers predictive of response are lacking. Methods: MIBC pts who received 3 cycles of cisplatin based NAC on 1 of 2 prospective multicenter clinical trials were included. Pts treated with accelerated methotrexate, vinblastine, doxorubicin + cisplatin (AMVAC) provided the discovery set [n = 34, 15/34 (44%) ≤pT1N0M0]. Pts treated with dose dense gemcitabine + cisplatin (DDGC) provided the validation set [n = 24, 11/24 (46%) ≤pT1N0M0]. DNA from pre-treatment tumor tissue underwent sequencing for all coding exons of 287 cancer related genes and was analyzed for presence of base substitutions, indels, copy number alterations, and selected re-arrangements. The mean number of variants and variant status for each gene were correlated with response using two-sample t-test and Fisher9s exact tests. Variant data were used to create a classification tree to discriminate responders vs. non-responders in the AMVAC discovery cohort. The resulting decision rule was then tested in the independent DDGC validation set. Overall survival analysis was performed using Kaplan-Meier. Results: Pts with pT0 had significantly more alterations than those with residual tumor in both the AMVAC discovery (p = .024) and DDGC validation (p = 0.018) set. In the AMVAC discovery set, alteration in ≥1 of the three DNA repair genes ATM, RB1 or FANCC predicted for ≤pT1N0M0 (p Conclusions: Alterations in ≥1 of ATM, RB1 and FANCC predict response to cisplatin based chemotherapy defined as ≤pT1N0M0 in both our AMVAC discovery and DDGC validation sets. We hypothesize that defects in these genes, which are important for maintenance of chromatin structure and DNA repair, confer sensitivity to DNA damaging chemotherapy and explain the accumulation of alterations seen among pts with pT0. External validation in collaboration with the cooperative groups is planned. Citation Format: Elizabeth R. Plimack, Roland L. Dunbrack, Timothy A. Brennan, Mark D. Andrake, Yan Zhou, Ilya Serebriiskii, Essel Dulaimi Al-Saleem, Jean Hoffman-Censits, Marijo Bilusic, Yu-Ning Wong, Alexander Kutikov, Rosalia Viterbo, Richard Greenberg, David Chen, Costas D. Lallas, Edouard J. Trabulsi, Roman Yelensky, Vincent A. Miller, Erica Golemis, Eric Ross. Defects in DNA repair genes and sensitivity to cisplatin based neoadjuvant chemotherapy (NAC) for bladder cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4298. doi:10.1158/1538-7445.AM2015-4298
The Journal of Molecular Diagnostics | 2018
Travis A. Clark; Jon Chung; Mark Kennedy; Jason D. Hughes; Niru Chennagiri; Daniel S. Lieber; Bernard Fendler; Lauren Young; Mandy Zhao; Michael Coyne; Virginia Breese; Geneva Young; Amy Donahue; Dean Pavlick; Alyssa Tsiros; Tim Brennan; Shan Zhong; Tariq I Mughal; Mark Bailey; Jie He; Steven Roels; Garrett Michael Frampton; Jill M. Spoerke; Steven Gendreau; Mark R. Lackner; Erica Schleifman; Eric Peters; Jeffrey S. Ross; Siraj M. Ali; Vincent A. Miller
Genomic profiling of circulating tumor DNA derived from cell-free DNA (cfDNA) in blood can provide a noninvasive method for detecting genomic biomarkers to guide clinical decision making for cancer patients. We developed a hybrid capture–based next-generation sequencing assay for genomic profiling of circulating tumor DNA from blood (FoundationACT). High-sequencing coverage and molecular barcode–based error detection enabled accurate detection of genomic alterations, including short variants (base substitutions, short insertions/deletions) and genomic re-arrangements at low allele frequencies (AFs), and copy number amplifications. Analytical validation was performed on 2666 reference alterations. The assay achieved >99% overall sensitivity (95% CI, 99.1%–99.4%) for short variants at AF >0.5%, >95% sensitivity (95% CI, 94.2%–95.7%) for AF 0.25% to 0.5%, and 70% sensitivity (95% CI, 68.2%–71.5%) for AF 0.125% to 0.25%. No false positives were detected in 62 samples from healthy volunteers. Genomic alterations detected by FoundationACT demonstrated high concordance with orthogonal assays run on the same clinical cfDNA samples. In 860 routine clinical FoundationACT cases, genomic alterations were detected in cfDNA at comparable frequencies to tissue; for the subset of cases with temporally matched tissue and blood samples, 75% of genomic alterations and 83% of short variant mutations detected in tissue were also detected in cfDNA. On the basis of analytical validation results, FoundationACT has been approved for use in our Clinical Laboratory Improvement Amendments–certified/College of American Pathologists–accredited/New York State–approved laboratory.
Cancer Research | 2016
Dean Pavlick; Siraj M. Ali; Julia A. Elvin; Phil Stephens; Vincent A. Miller; Jeffrey S. Ross; James Suh; Jo-Anne Vergilio; Juliann Chmielecki; Tim Brennan; John R. Crawford; Denise M. Malicki; Hyunah Ahn; Victor Wong
Purpose: NTRK1-3 fusions are oncogenic drivers in lung and other carcinomas in adults, suggest benefit from targeted therapy. We examined a large series of pediatric/adolescent advanced cancer cases to identify those with NTRK fusions. Background: The NTRK, neurotrophic tyrosine kinase receptor, genes encode proteins essential for normal neuronal growth and development. Fusions of NTRK family members have recently been linked to oncogenesis as well as identified as potential targets of therapy. In this study, a hybrid capture based comprehensive genomic profiling (CGP) assay was used to study a large series of pediatric solid tumors to search for NTRK gene fusions. Methods: CGP using hybridization capture of at least 3,320 exons from 182 cancer-related genes and 37 introns of 14 genes commonly rearranged in cancer (previous version of the test) was applied to ≥ 50ng of DNA extracted from 1351 pediatric/adolescent/young adult tumors ( Results:, From 1351 pediatric/adolescent young adult advanced cancer patients, 7 (0.52%) harbored NTRK family member fusions. Of these, 5 were pediatric ( Conclusions: 0.5% pediatric/adolescent/young advanced cancers harbor NTRK1 and NTRK3 fusions, and all such tumors are mesenchymal in origin. Of these, an index case benefitted from crizotinib treatment, suggesting pathways to clinical benefit for such cases. Citation Format: Dean Pavlick, Siraj Mahamed Ali, Julia A. Elvin, Phil J. Stephens, Vincent A. Miller, Jeffrey S. Ross, James H. Suh, Jo-Anne Vergilio, Juliann Chmielecki, Tim Brennan, John R. Crawford, Denise M. Malicki, Hyunah Ahn, Victor N. Wong. Identification of NTRK fusions in pediatric tumors via comprehensive genomic profiling. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Pediatric Cancer Research: From Mechanisms and Models to Treatment and Survivorship; 2015 Nov 9-12; Fort Lauderdale, FL. Philadelphia (PA): AACR; Cancer Res 2016;76(5 Suppl):Abstract nr A46.