Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maeve Aine Lowery is active.

Publication


Featured researches published by Maeve Aine Lowery.


Nature Medicine | 2017

Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients

Ahmet Zehir; Ryma Benayed; Ronak Shah; Aijazuddin Syed; Sumit Middha; Hyunjae R. Kim; Preethi Srinivasan; Jianjiong Gao; Debyani Chakravarty; Sean M. Devlin; Matthew D. Hellmann; David Barron; Alison M. Schram; Meera Hameed; Snjezana Dogan; Dara S. Ross; Jaclyn F. Hechtman; Deborah DeLair; Jinjuan Yao; Diana Mandelker; Donavan T. Cheng; Raghu Chandramohan; Abhinita Mohanty; Ryan Ptashkin; Gowtham Jayakumaran; Meera Prasad; Mustafa H Syed; Anoop Balakrishnan Rema; Zhen Y Liu; Khedoudja Nafa

Tumor molecular profiling is a fundamental component of precision oncology, enabling the identification of genomic alterations in genes and pathways that can be targeted therapeutically. The existence of recurrent targetable alterations across distinct histologically defined tumor types, coupled with an expanding portfolio of molecularly targeted therapies, demands flexible and comprehensive approaches to profile clinically relevant genes across the full spectrum of cancers. We established a large-scale, prospective clinical sequencing initiative using a comprehensive assay, MSK-IMPACT, through which we have compiled tumor and matched normal sequence data from a unique cohort of more than 10,000 patients with advanced cancer and available pathological and clinical annotations. Using these data, we identified clinically relevant somatic mutations, novel noncoding alterations, and mutational signatures that were shared by common and rare tumor types. Patients were enrolled on genomically matched clinical trials at a rate of 11%. To enable discovery of novel biomarkers and deeper investigation into rare alterations and tumor types, all results are publicly accessible.


Oncologist | 2011

An Emerging Entity: Pancreatic Adenocarcinoma Associated with a Known BRCA Mutation: Clinical Descriptors, Treatment Implications, and Future Directions

Maeve Aine Lowery; David P. Kelsen; Zsofia K. Stadler; Kenneth H. Yu; Yelena Y. Janjigian; Emmy Ludwig; David R. D'Adamo; Erin E. Salo-Mullen; Mark E. Robson; Peter J. Allen; Robert C. Kurtz; Eileen Mary O'Reilly

BACKGROUND BRCA1 and BRCA2 germline mutations are associated with an elevated risk for pancreas adenocarcinoma (PAC). Other BRCA-associated cancers have been shown to have greater sensitivity to platinum and poly(ADP-ribose) polymerase (PARP) inhibitors with better clinical outcomes than in sporadic cases; however, outcomes in BRCA-associated PAC have not been reported. METHODS Patients with a known BRCA1 or BRCA2 mutation and a diagnosis of PAC were identified from the Gastrointestinal Oncology Service, Familial Pancreas Cancer Registry, and Clinical Genetics Service at Memorial Sloan-Kettering Cancer Center. RESULTS Fifteen patients, five male, with a BRCA1 (n = 4) or BRCA2 (n = 11) mutation and PAC and one patient with a BRCA1 mutation and acinar cell carcinoma of the pancreas were identified. Seven female patients (70%) had a prior history of breast cancer. Four patients received a PARP inhibitor alone or in combination with chemotherapy; three demonstrated an initial radiographic partial response by Response Evaluation Criteria in Solid Tumors whereas one patient had stable disease for 6 months. Six patients received platinum-based chemotherapy first line for metastatic disease; five of those patients had a radiographic partial response. CONCLUSION BRCA mutation-associated PAC represents an underidentified, but clinically important, subgroup of patients. This is of particular relevance given the ongoing development of therapeutic agents targeting DNA repair, which may potentially offer a significant benefit to a genetically selected population. We anticipate that further study and understanding of the clinical and biologic features of BRCA-mutant PAC will aid in the identification of tissue biomarkers indicating defective tumor DNA repair pathways in sporadic PAC.


Oncologist | 2011

Acinar Cell Carcinoma of the Pancreas: New Genetic and Treatment Insights into a Rare Malignancy

Maeve Aine Lowery; David S. Klimstra; Jinru Shia; Kenneth H. Yu; Peter J. Allen; Murray F. Brennan; Eileen Mary O'Reilly

BACKGROUND Acinar cell carcinoma (ACC) of the pancreas is a rare neoplasm, accounting for 1% of all pancreatic neoplasms. There remains a lack of data regarding the use of systemic therapy in this disease. We present a series of 40 consecutive cases of ACC of the pancreas treated at Memorial Sloan-Kettering Cancer Center, with an emphasis on evaluation of activity of new therapeutic agents. METHODS Patients reviewed at our institution from January 2000 through January 2011 were identified from an institutional database with prior institutional review board approval. Pathology was confirmed in all cases as ACC or a closely related entity. RESULTS Forty patients were identified; 29 were male (73%). The median age at diagnosis was 65 years (range, 16-87 years). The median overall survival (OS) time for patients with localized, resectable disease was 56.9 months and the OS time for patients with metastatic ACC (n = 18) was 19.6 months. Six patients with metastatic or recurrent ACC had a partial response to chemotherapy and five patients had stable disease for ≥6 months on systemic chemotherapy. Clinical observation was made of a patient with ACC and hereditary nonpolyposis colorectal cancer and a patient with ACC and a BRCA1 germline mutation. CONCLUSIONS ACC is moderately chemoresponsive to agents that have activity in pancreatic adenocarcinoma and colorectal carcinoma. A potential association between germline mutations in DNA mismatch repair genes and ACC warrants further evaluation.


JCO Precision Oncology | 2017

OncoKB: A Precision Oncology Knowledge Base

Debyani Chakravarty; Jianjiong Gao; Sarah Phillips; Ritika Kundra; Hongxin Zhang; Jiaojiao Wang; Julia E. Rudolph; Rona Yaeger; Tara Soumerai; Moriah H. Nissan; Matthew T. Chang; Sarat Chandarlapaty; Tiffany A. Traina; Paul K. Paik; Alan L. Ho; Feras M. Hantash; Andrew Grupe; Shrujal S. Baxi; Margaret K. Callahan; Alexandra Snyder; Ping Chi; Daniel C. Danila; Mrinal M. Gounder; James J. Harding; Matthew D. Hellmann; Gopa Iyer; Yelena Y. Janjigian; Thomas Kaley; Douglas A. Levine; Maeve Aine Lowery

PURPOSE With prospective clinical sequencing of tumors emerging as a mainstay in cancer care, there is an urgent need for a clinical support tool that distills the clinical implications associated with specific mutation events into a standardized and easily interpretable format. To this end, we developed OncoKB, an expert-guided precision oncology knowledge base. METHODS OncoKB annotates the biological and oncogenic effect and the prognostic and predictive significance of somatic molecular alterations. Potential treatment implications are stratified by the level of evidence that a specific molecular alteration is predictive of drug response based on US Food and Drug Administration (FDA) labeling, National Comprehensive Cancer Network (NCCN) guidelines, disease-focused expert group recommendations and the scientific literature. RESULTS To date, over 3000 unique mutations, fusions, and copy number alterations in 418 cancer-associated genes have been annotated. To test the utility of OncoKB, we annotated all genomic events in 5983 primary tumor samples in 19 cancer types. Forty-one percent of samples harbored at least one potentially actionable alteration, of which 7.5% were predictive of clinical benefit from a standard treatment. OncoKB annotations are available through a public web resource (http://oncokb.org/) and are also incorporated into the cBioPortal for Cancer Genomics to facilitate the interpretation of genomic alterations by physicians and researchers. CONCLUSION OncoKB, a comprehensive and curated precision oncology knowledge base, offers oncologists detailed, evidence-based information about individual somatic mutations and structural alterations present in patient tumors with the goal of supporting optimal treatment decisions.


Annals of Surgery | 2014

A single-arm, nonrandomized phase II trial of neoadjuvant gemcitabine and oxaliplatin in patients with resectable pancreas adenocarcinoma.

Eileen Mary O'Reilly; Anna Perelshteyn; William R. Jarnagin; Mark A. Schattner; Hans Gerdes; Marinela Capanu; Laura H. Tang; Joseph LaValle; Corinne B. Winston; Ronald P. DeMatteo; Michael I. D'Angelica; Robert C. Kurtz; Ghassan K. Abou-Alfa; David S. Klimstra; Maeve Aine Lowery; Murray F. Brennan; Daniel G. Coit; Diane Lauren Reidy; T. Peter Kingham; Peter J. Allen

Background:The role for neoadjuvant systemic therapy in resectable pancreas adenocarcinoma remains undefined. Objective:We evaluated the efficacy of gemcitabine and oxaliplatin administered as preoperative therapy in patients with resectable pancreas adenocarcinoma. Methods:Eligible patients were screened using computed tomography–pancreas angiography, laparoscopy, endoscopic ultrasonography, and fine-needle aspiration cytology to identify 38 patients who received 4 cycles of neoadjuvant gemcitabine 1000 mg/m2 intravenously over 100 minutes and oxaliplatin 80 mg/m2 intravenously over 2 hours, every 2 weeks. Patients whose tumors remained resectable at restaging proceeded to operation and subsequently received 5 cycles of adjuvant gemcitabine (1000 mg/m2 intravenously over 30 minutes days 1, 8, and 15 every 4 weeks). The primary endpoint was 18-month overall survival and secondary endpoints included radiological, tumor marker and pathological response to neoadjuvant therapy, time to recurrence, patterns of failure, and feasibility of obtaining preoperative core biopsies. Results:Thirty-five of 38 patients (92%) completed neoadjuvant therapy. Twenty-seven patients underwent tumor resection (resectability rate 71%), of which 26 initiated adjuvant therapy for a total of 23 patients (60.5%) who completed all planned therapy. The 18-month survival was 63% (24 patients alive). The median overall survival for all 38 patients was 27.2 months (95% confidence interval: 17–NA) and the median disease-specific survival was 30.6 months (95% confidence interval: 19–NA). Conclusions:This study met its endpoint and provided a signal suggesting that exploration of neoadjuvant systemic therapy is worthy of further investigation in resectable pancreas adenocarcinoma. Improved patient selection and more active systemic regimens are key. Clinical trials identification: NCT00536874.


Cancer | 2015

Identification of germline genetic mutations in patients with pancreatic cancer

Erin E. Salo-Mullen; Eileen Mary O'Reilly; David P. Kelsen; Asad Ashraf; Maeve Aine Lowery; Kenneth H. Yu; Diane Lauren Reidy; Andrew S. Epstein; Anne Lincoln; Amethyst Saldia; Lauren Jacobs; Rohini Rau-Murthy; Liying Zhang; Robert C. Kurtz; Leonard Saltz; Kenneth Offit; Mark E. Robson; Zsofia K. Stadler

Pancreatic adenocarcinoma (PAC) is part of several cancer predisposition syndromes; however, indications for genetic counseling/testing are not well‐defined. In the current study, the authors sought to determine mutation prevalence and characteristics that are predictive of an inherited predisposition for PAC.


JAMA | 2017

Mutation Detection in Patients With Advanced Cancer by Universal Sequencing of Cancer-Related Genes in Tumor and Normal DNA vs Guideline-Based Germline Testing

Diana Mandelker; Liying Zhang; Yelena Kemel; Zsofia K. Stadler; Vijai Joseph; Ahmet Zehir; Nisha Pradhan; Angela G. Arnold; Michael F. Walsh; Yirong Li; Anoop R. Balakrishnan; Aijazuddin Syed; Meera Prasad; Khedoudja Nafa; Maria I. Carlo; Karen A. Cadoo; Meg Sheehan; Megan Harlan Fleischut; Erin E. Salo-Mullen; Magan Trottier; Steven M. Lipkin; Anne Lincoln; Semanti Mukherjee; Vignesh Ravichandran; Roy Cambria; Jesse Galle; Wassim Abida; Marcia E. Arcila; Ryma Benayed; Ronak Shah

Importance Guidelines for cancer genetic testing based on family history may miss clinically actionable genetic changes with established implications for cancer screening or prevention. Objective To determine the proportion and potential clinical implications of inherited variants detected using simultaneous sequencing of the tumor and normal tissue (“tumor-normal sequencing”) compared with genetic test results based on current guidelines. Design, Setting, and Participants From January 2014 until May 2016 at Memorial Sloan Kettering Cancer Center, 10 336 patients consented to tumor DNA sequencing. Since May 2015, 1040 of these patients with advanced cancer were referred by their oncologists for germline analysis of 76 cancer predisposition genes. Patients with clinically actionable inherited mutations whose genetic test results would not have been predicted by published decision rules were identified. Follow-up for potential clinical implications of mutation detection was through May 2017. Exposure Tumor and germline sequencing compared with the predicted yield of targeted germline sequencing based on clinical guidelines. Main Outcomes and Measures Proportion of clinically actionable germline mutations detected by universal tumor-normal sequencing that would not have been detected by guideline-directed testing. Results Of 1040 patients, the median age was 58 years (interquartile range, 50.5-66 years), 65.3% were male, and 81.3% had stage IV disease at the time of genomic analysis, with prostate, renal, pancreatic, breast, and colon cancer as the most common diagnoses. Of the 1040 patients, 182 (17.5%; 95% CI, 15.3%-19.9%) had clinically actionable mutations conferring cancer susceptibility, including 149 with moderate- to high-penetrance mutations; 101 patients tested (9.7%; 95% CI, 8.1%-11.7%) would not have had these mutations detected using clinical guidelines, including 65 with moderate- to high-penetrance mutations. Frequency of inherited mutations was related to case mix, stage, and founder mutations. Germline findings led to discussion or initiation of change to targeted therapy in 38 patients tested (3.7%) and predictive testing in the families of 13 individuals (1.3%), including 6 for whom genetic evaluation would not have been initiated by guideline-based testing. Conclusions and Relevance In this referral population with selected advanced cancers, universal sequencing of a broad panel of cancer-related genes in paired germline and tumor DNA samples was associated with increased detection of individuals with potentially clinically significant heritable mutations over the predicted yield of targeted germline testing based on current clinical guidelines. Knowledge of these additional mutations can help guide therapeutic and preventive interventions, but whether all of these interventions would improve outcomes for patients with cancer or their family members requires further study. Trial Registration clinicaltrials.gov Identifier: NCT01775072


Clinical Cancer Research | 2014

Pharmacogenomic Modeling of Circulating Tumor and Invasive Cells for Prediction of Chemotherapy Response and Resistance in Pancreatic Cancer

Kenneth H. Yu; Mark Ricigliano; Manuel Hidalgo; Ghassan K. Abou-Alfa; Maeve Aine Lowery; Leonard Saltz; Joseph F. Crotty; Kristen Gary; Brandon Cooper; Rena G. Lapidus; Mariola Sadowska; Eileen Mary O'Reilly

Purpose: Despite a challenging prognosis, modern cytotoxic therapy can induce tumor responses and extend life in pancreatic adenocarcinoma (PDAC). Pharmacogenomic (PGx) modeling of tumor tissue can predict the efficacy of chemotherapeutic agents in preclinical cancer models. We hypothesized that PGx profiling of circulating tumor and invasive cells (CTIC) isolated from peripheral blood could predict tumor response, progression, and resistance. Experimental Design: A PGx model was created and validated in preclinical models. A prospective clinical trial was conducted. Fifty patients with advanced PDAC were enrolled. Before treatment, 10 mL of peripherally drawn blood was collected. CTICs isolated from this blood sample were expression profiled and the PGx model was used to predict effective and ineffective chemotherapeutic agents. The treating physicians were blinded to PGx prediction. Results: We found that CTICs could be reliably isolated, total RNA extracted and profiled from 10 mL of peripheral blood from patients with unresectable PDAC before chemotherapy treatment and at disease progression. Using previously created PGx models to predict chemotherapy sensitivity, we found that clinical benefit was seen for study participants treated with chemotherapy regimens predicted to be effective versus chemotherapy regimens predicted to be ineffective with regard to progression-free (10.4 mo vs. 3.6 mo; P < 0.0001; HR, 0.14) and overall survival (17.2 mo vs. 8.3 mo; P < 0.0249; HR, 0.29). Conclusions: These findings suggest that PGx profiling of CTICs can predict treatment response. Clin Cancer Res; 20(20); 5281–9. ©2014 AACR.


Clinical Cancer Research | 2017

Real Time Genomic Profiling of Pancreatic Ductal Adenocarcinoma: Potential Actionability and Correlation with Clinical Phenotype

Maeve Aine Lowery; Emmet Jordan; Olca Basturk; Ryan Ptashkin; Ahmet Zehir; Michael F. Berger; Tanisha Leach; Brian Herbst; Gokce Askan; Hannah Maynard; Danielle C. Glassman; Christina M. Covington; Nikolaus Schultz; Ghassan K. Abou-Alfa; James J. Harding; David S. Klimstra; Jaclyn F. Hechtman; David M. Hyman; Peter J. Allen; William R. Jarnagin; Vinod P. Balachandran; Anna M. Varghese; Mark A. Schattner; Kenneth H. Yu; Leonard Saltz; David B. Solit; Christine A. Iacobuzio-Donahue; Steven D. Leach; Eileen Mary O'Reilly

Purpose: Molecular profiling in cancer has identified potential actionable drug targets that have prompted attempts to discover clinically validated biomarkers to guide therapeutic decision-making and enrollment to clinical trials. We evaluated whether comprehensive genetic analysis of patients with pancreatic adenocarcinoma is feasible within a clinically relevant timeframe and whether such analyses provide predictive and/or prognostic information along with identification of potential targets for therapy. Experimental Design: Archival or prospectively acquired FFPE samples and matched normal DNA from N = 336 patients with pancreatic cancer were analyzed using a hybridization capture–based, next-generation sequencing assay designed to perform targeted deep sequencing of all exons and selected introns of 410 key cancer-associated genes. Demographic and treatment data were prospectively collected with the goal of correlating treatment outcomes and drug response with molecular profiles. Results: The median time from protocol consent to reporting of the genomic results was 45 days with a median time from tissue delivery of 20 days. All genetic alterations identified were stratified based upon prior evidence that the mutation is a predictive biomarker of drug response using the MSKCC OncoKB classification. Three of 225 patients (1%) received a matched therapy based upon the sequencing results. Conclusions: The practical application of molecular results to guide individual patient treatment is currently limited in patients with pancreatic adenocarcinoma. Future prospective molecular profiling efforts should seek to incorporate routine germline genetic analysis and the identification of DNA profiles that predict for clinical benefit from agents that target DNA damage repair and or immunotherapy. Clin Cancer Res; 23(20); 6094–100. ©2017 AACR.


Journal of Clinical Oncology | 2017

Phase II Study of BGJ398 in Patients With FGFR-Altered Advanced Cholangiocarcinoma

Milind Javle; Maeve Aine Lowery; Rachna T. Shroff; Karl Heinz Weiss; Christoph Springfeld; Mitesh J. Borad; Ramesh K. Ramanathan; Lipika Goyal; Saeed Sadeghi; Teresa Macarulla; Anthony B. El-Khoueiry; Ivan Borbath; Su Pin Choo; Do Youn Oh; Philip A. Philip; Li-Tzong Chen; Thanyanan Reungwetwattana; Eric Van Cutsem; Kun-Huei Yeh; Kristen K. Ciombor; Richard S. Finn; Anuradha Patel; Suman Sen; Dale Porter; Randi Isaacs; Andrew X. Zhu; Ghassan K. Abou-Alfa; Tanios Bekaii-Saab

Purpose No standard treatment exists for patients with cholangiocarcinoma for whom first-line gemcitabine-based therapy fails. Fibroblast growth factor receptor 2 ( FGFR2) fusions/translocations are present in 13% to 17% of intrahepatic cholangiocarcinomas. BGJ398, an orally bioavailable, selective pan-FGFR kinase inhibitor, has shown preliminary clinical activity against tumors with FGFR alterations. Methods A multicenter, open-label, phase II study ( ClinicalTrials.gov identifier: NCT02150967) evaluated BGJ398 antitumor activity in patients age ≥ 18 years with advanced or metastatic cholangiocarcinoma containing FGFR2 fusions or other FGFR alterations whose disease had progressed while receiving prior therapy. Patients received BGJ398 125 mg once daily for 21 days, then 7 days off (28-day cycles). The primary end point was investigator-assessed overall response rate. Results Sixty-one patients (35 women; median age, 57 years) with FGFR2 fusion (n = 48), mutation (n = 8), or amplification (n = 3) participated. At the prespecified data cutoff (June 30, 2016), 50 patients had discontinued treatment. All responsive tumors contained FGFR2 fusions. The overall response rate was 14.8% (18.8% FGFR2 fusions only), disease control rate was 75.4% (83.3% FGFR2 fusions only), and estimated median progression-free survival was 5.8 months (95% CI, 4.3 to 7.6 months). Adverse events included hyperphosphatemia (72.1% all grade), fatigue (36.1%), stomatitis (29.5%), and alopecia (26.2%). Grade 3 or 4 treatment-related adverse events occurred in 25 patients (41%) and included hyperphosphatemia (16.4%), stomatitis (6.6%), and palmar-plantar erythrodysesthesia (4.9%). Conclusion BGJ398 is a first-in-class FGFR kinase inhibitor with manageable toxicities that shows meaningful clinical activity against chemotherapy-refractory cholangiocarcinoma containing FGFR2 fusions. This promising antitumor activity supports continued development of BGJ398 in this highly selected patient population.

Collaboration


Dive into the Maeve Aine Lowery's collaboration.

Top Co-Authors

Avatar

Eileen Mary O'Reilly

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kenneth H. Yu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ghassan K. Abou-Alfa

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Marinela Capanu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David P. Kelsen

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Peter J. Allen

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David S. Klimstra

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

James J. Harding

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anna M. Varghese

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Eileen M. O’Reilly

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge