scispace - formally typeset
Search or ask a question
Author

Antonio Omuro

Other affiliations: University of Paris, Yale Cancer Center, Kettering University  ...read more
Bio: Antonio Omuro is an academic researcher from Yale University. The author has contributed to research in topics: Temozolomide & Bevacizumab. The author has an hindex of 48, co-authored 174 publications receiving 11151 citations. Previous affiliations of Antonio Omuro include University of Paris & Yale Cancer Center.


Papers
More filters
Journal ArticleDOI
Robert M. Samstein1, Chung-Han Lee1, Chung-Han Lee2, Alexander N. Shoushtari2, Alexander N. Shoushtari1, Matthew D. Hellmann2, Matthew D. Hellmann1, Ronglai Shen1, Yelena Y. Janjigian1, Yelena Y. Janjigian2, David Barron1, Ahmet Zehir1, Emmet Jordan1, Antonio Omuro1, Thomas Kaley1, Sviatoslav M. Kendall1, Robert J. Motzer1, Robert J. Motzer2, A. Ari Hakimi1, Martin H. Voss2, Martin H. Voss1, Paul Russo1, Jonathan E. Rosenberg2, Jonathan E. Rosenberg1, Gopa Iyer1, Gopa Iyer2, Bernard H. Bochner1, Dean F. Bajorin2, Dean F. Bajorin1, Hikmat Al-Ahmadie1, Jamie E. Chaft2, Jamie E. Chaft1, Charles M. Rudin2, Charles M. Rudin1, Gregory J. Riely1, Gregory J. Riely2, Shrujal S. Baxi1, Shrujal S. Baxi2, Alan L. Ho2, Alan L. Ho1, Richard J. Wong1, David G. Pfister2, David G. Pfister1, Jedd D. Wolchok1, Jedd D. Wolchok2, Christopher A. Barker1, Philip H. Gutin1, Cameron Brennan1, Viviane Tabar1, Ingo K. Mellinghoff1, Lisa M. DeAngelis1, Charlotte E. Ariyan1, Nancy Y. Lee1, William D. Tap1, William D. Tap2, Mrinal M. Gounder2, Mrinal M. Gounder1, Sandra P. D'Angelo2, Sandra P. D'Angelo1, Leonard B. Saltz2, Leonard B. Saltz1, Zsofia K. Stadler1, Zsofia K. Stadler2, Howard I. Scher2, Howard I. Scher1, José Baselga1, José Baselga2, Pedram Razavi2, Pedram Razavi1, Christopher A. Klebanoff1, Christopher A. Klebanoff2, Rona Yaeger1, Rona Yaeger2, Neil H. Segal1, Neil H. Segal2, Geoffrey Y. Ku1, Geoffrey Y. Ku2, Ronald P. DeMatteo1, Marc Ladanyi1, Naiyer A. Rizvi3, Michael F. Berger1, Nadeem Riaz1, David B. Solit1, Timothy A. Chan1, Luc G. T. Morris1 
TL;DR: Analysis of advanced cancer patients treated with immune-checkpoint inhibitors shows that tumor mutational burden, as assessed by targeted next-generation sequencing, predicts survival after immunotherapy across multiple cancer types.
Abstract: Immune checkpoint inhibitor (ICI) treatments benefit some patients with metastatic cancers, but predictive biomarkers are needed. Findings in selected cancer types suggest that tumor mutational burden (TMB) may predict clinical response to ICI. To examine this association more broadly, we analyzed the clinical and genomic data of 1,662 advanced cancer patients treated with ICI, and 5,371 non-ICI-treated patients, whose tumors underwent targeted next-generation sequencing (MSK-IMPACT). Among all patients, higher somatic TMB (highest 20% in each histology) was associated with better overall survival. For most cancer histologies, an association between higher TMB and improved survival was observed. The TMB cutpoints associated with improved survival varied markedly between cancer types. These data indicate that TMB is associated with improved survival in patients receiving ICI across a wide variety of cancer types, but that there may not be one universal definition of high TMB.

2,343 citations

Journal ArticleDOI
Ahmet Zehir1, Ryma Benayed1, Ronak Shah1, Aijazuddin Syed1, Sumit Middha1, Hyunjae R. Kim1, Preethi Srinivasan1, Jianjiong Gao1, Debyani Chakravarty1, Sean M. Devlin1, Matthew D. Hellmann1, David Barron1, Alison M. Schram1, Meera Hameed1, Snjezana Dogan1, Dara S. Ross1, Jaclyn F. Hechtman1, Deborah DeLair1, Jinjuan Yao1, Diana Mandelker1, Donavan T. Cheng1, Raghu Chandramohan1, Abhinita Mohanty1, Ryan Ptashkin1, Gowtham Jayakumaran1, Meera Prasad1, Mustafa H Syed1, Anoop Balakrishnan Rema1, Zhen Y Liu1, Khedoudja Nafa1, Laetitia Borsu1, Justyna Sadowska1, Jacklyn Casanova1, Ruben Bacares1, Iwona Kiecka1, Anna Razumova1, Julie B Son1, Lisa Stewart1, Tessara Baldi1, Kerry Mullaney1, Hikmat Al-Ahmadie1, Efsevia Vakiani1, Adam Abeshouse1, Alexander V Penson1, Philip Jonsson1, Niedzica Camacho1, Matthew T. Chang1, Helen Won1, Benjamin Gross1, Ritika Kundra1, Zachary J. Heins1, Hsiao-Wei Chen1, Sarah Phillips1, Hongxin Zhang1, Jiaojiao Wang1, Angelica Ochoa1, Jonathan Wills1, Michael H. Eubank1, Stacy B. Thomas1, Stuart Gardos1, Dalicia N. Reales1, Jesse Galle1, Robert Durany1, Roy Cambria1, Wassim Abida1, Andrea Cercek1, Darren R. Feldman1, Mrinal M. Gounder1, A. Ari Hakimi1, James J. Harding1, Gopa Iyer1, Yelena Y. Janjigian1, Emmet Jordan1, Ciara Marie Kelly1, Maeve A. Lowery1, Luc G. T. Morris1, Antonio Omuro1, Nitya Raj1, Pedram Razavi1, Alexander N. Shoushtari1, Neerav Shukla1, Tara Soumerai1, Anna M. Varghese1, Rona Yaeger1, Jonathan A. Coleman1, Bernard H. Bochner1, Gregory J. Riely1, Leonard B. Saltz1, Howard I. Scher1, Paul Sabbatini1, Mark E. Robson1, David S. Klimstra1, Barry S. Taylor1, José Baselga1, Nikolaus Schultz1, David M. Hyman1, Maria E. Arcila1, David B. Solit1, Marc Ladanyi1, Michael F. Berger1 
TL;DR: A large-scale, prospective clinical sequencing initiative using a comprehensive assay, MSK-IMPACT, through which tumor and matched normal sequence data from a unique cohort of more than 10,000 patients with advanced cancer are compiled and identified clinically relevant somatic mutations, novel noncoding alterations, and mutational signatures that were shared by common and rare tumor types.
Abstract: 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.

2,330 citations

Journal ArticleDOI
06 Nov 2013-JAMA
TL;DR: The clinical management of malignant gliomas is reviewed, including genetic and environmental risk factors such as cell phones, diagnostic pitfalls, symptom management, specific antitumor therapy, and common complications.
Abstract: Importance Glioblastomas and malignant gliomas are the most common primary malignant brain tumors, with an annual incidence of 5.26 per 100 000 population or 17 000 new diagnoses per year. These tumors are typically associated with a dismal prognosis and poor quality of life. Objective To review the clinical management of malignant gliomas, including genetic and environmental risk factors such as cell phones, diagnostic pitfalls, symptom management, specific antitumor therapy, and common complications. Evidence Review Search of PubMed references from January 2000 to May 2013 using the terms glioblastoma , glioma , malignant glioma , anaplastic astrocytoma , anaplastic oligodendroglioma , anaplastic oligoastrocytoma , and brain neoplasm . Articles were also identified through searches of the authors’ own files. Evidence was graded using the American Heart Association classification system. Findings Only radiation exposure and certain genetic syndromes are well-defined risk factors for malignant glioma. The treatment of newly diagnosed glioblastoma is based on radiotherapy combined with temozolomide. This approach doubles the 2-year survival rate to 27%, but overall prognosis remains poor. Bevacizumab is an emerging treatment alternative that deserves further study. Grade III tumors have been less well studied, and clinical trials to establish standards of care are ongoing. Patients with malignant gliomas experience frequent clinical complications, including thromboembolic events, seizures, fluctuations in neurologic symptoms, and adverse effects from corticosteroids and chemotherapies that require proper management and prophylaxis. Conclusions and Relevance Glioblastoma remains a difficult cancer to treat, although therapeutic options have been improving. Optimal management requires a multidisciplinary approach and knowledge of potential complications from both the disease and its treatment.

1,756 citations

Journal ArticleDOI
TL;DR: PLX3397 was well tolerated and readily crossed the blood-tumor barrier but showed no efficacy, and additional studies are ongoing, testing combination strategies and potential biomarkers to identify patients with greater likelihood of response.
Abstract: Background The colony stimulating factor 1 receptor (CSF1R) ligands, CSF1 and interleukin-34, and the KIT ligand, stem cell factor, are expressed in glioblastoma (GB). Microglia, macrophages, blood vessels, and tumor cells also express CSF1R, and depletion of the microglia reduces tumor burden and invasive capacity. PLX3397 is an oral, small molecule that selectively inhibits CSF1R and KIT, penetrates the blood-brain barrier in model systems, and represents a novel approach for clinical development. Methods We conducted a phase II study in patients with recurrent GB. The primary endpoint was 6-month progression-free survival (PFS6). Secondary endpoints included overall survival response rate, safety, and plasma/tumor tissue pharmacokinetics. Exploratory endpoints included pharmacodynamic measures of drug effect in blood and tumor tissue. Results A total of 37 patients were enrolled, with 13 treated prior to a planned surgical resection (Cohort 1) and 24 treated without surgery (Cohort 2). PLX3397 was given at an oral dose of 1000 mg daily and was well tolerated. The primary efficacy endpoint of PFS6 was only 8.6%, with no objective responses. Pharmacokinetic endpoints revealed a median maximal concentration (Cmax) of 8090 ng/mL, with a time to attain Cmax of 2 hour in plasma. Tumor tissue obtained after 7 days of drug exposure revealed a median drug level of 5500 ng/g. Pharmacodynamic changes included an increase in colony stimulating factor 1 and reduced CD14(dim)/CD16+ monocytes in plasma compared with pretreatment baseline values. Conclusion PLX3397 was well tolerated and readily crossed the blood-tumor barrier but showed no efficacy. Additional studies are ongoing, testing combination strategies and potential biomarkers to identify patients with greater likelihood of response.

393 citations

Journal ArticleDOI
TL;DR: R-MPV combined with consolidation rdWBRT and cytarabine is associated with high response rates, long-term disease control, and minimal neurotoxicity in primary CNS lymphoma.
Abstract: Purpose A multicenter phase II study was conducted to assess the efficacy of rituximab, methotrexate, procarbazine, and vincristine (R-MPV) followed by consolidation reduced-dose whole-brain radiotherapy (rdWBRT) and cytarabine in primary CNS lymphoma. Patients and Methods Patients received induction chemotherapy with R-MPV (five to seven cycles); those achieving a complete response (CR) received rdWBRT (23.4 Gy), and otherwise, standard WBRT was offered (45 Gy). Consolidation cytarabine was given after the radiotherapy. The primary end point was 2-year progression-free survival (PFS) in patients receiving rdWBRT. Exploratory end points included prospective neuropsychological evaluation, analysis of magnetic resonance imaging (MRI) white matter changes using the Fazekas scale, and evaluation of the apparent diffusion coefficient (ADC) as a prognostic factor. Results Fifty-two patients were enrolled, with median age of 60 years (range, 30 to 79 years) and median Karnofsky performance score of 70 (range, 50...

351 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: Benefits of adjuvant temozolomide with radiotherapy lasted throughout 5 years of follow-up, and a benefit of combined therapy was recorded in all clinical prognostic subgroups, including patients aged 60-70 years.
Abstract: BACKGROUND: In 2004, a randomised phase III trial by the European Organisation for Research and Treatment of Cancer (EORTC) and National Cancer Institute of Canada Clinical Trials Group (NCIC) reported improved median and 2-year survival for patients with glioblastoma treated with concomitant and adjuvant temozolomide and radiotherapy. We report the final results with a median follow-up of more than 5 years. METHODS: Adult patients with newly diagnosed glioblastoma were randomly assigned to receive either standard radiotherapy or identical radiotherapy with concomitant temozolomide followed by up to six cycles of adjuvant temozolomide. The methylation status of the methyl-guanine methyl transferase gene, MGMT, was determined retrospectively from the tumour tissue of 206 patients. The primary endpoint was overall survival. Analyses were by intention to treat. This trial is registered with Clinicaltrials.gov, number NCT00006353. FINDINGS: Between Aug 17, 2000, and March 22, 2002, 573 patients were assigned to treatment. 278 (97%) of 286 patients in the radiotherapy alone group and 254 (89%) of 287 in the combined-treatment group died during 5 years of follow-up. Overall survival was 27.2% (95% CI 22.2-32.5) at 2 years, 16.0% (12.0-20.6) at 3 years, 12.1% (8.5-16.4) at 4 years, and 9.8% (6.4-14.0) at 5 years with temozolomide, versus 10.9% (7.6-14.8), 4.4% (2.4-7.2), 3.0% (1.4-5.7), and 1.9% (0.6-4.4) with radiotherapy alone (hazard ratio 0.6, 95% CI 0.5-0.7; p<0.0001). A benefit of combined therapy was recorded in all clinical prognostic subgroups, including patients aged 60-70 years. Methylation of the MGMT promoter was the strongest predictor for outcome and benefit from temozolomide chemotherapy. INTERPRETATION: Benefits of adjuvant temozolomide with radiotherapy lasted throughout 5 years of follow-up. A few patients in favourable prognostic categories survive longer than 5 years. MGMT methylation status identifies patients most likely to benefit from the addition of temozolomide. FUNDING: EORTC, NCIC, Nelia and Amadeo Barletta Foundation, Schering-Plough.

6,161 citations

Journal ArticleDOI
TL;DR: This Perspective has organized known cancer-associated metabolic changes into six hallmarks: deregulated uptake of glucose and amino acids, use of opportunistic modes of nutrient acquisition, useof glycolysis/TCA cycle intermediates for biosynthesis and NADPH production, increased demand for nitrogen, alterations in metabolite-driven gene regulation, and metabolic interactions with the microenvironment.

3,565 citations

Journal ArticleDOI
TL;DR: The fifth edition of the WHO Classification of Tumors of the Central Nervous System (CNS), published in 2021, is the sixth version of the international standard for the classification of brain and spinal cord tumors as mentioned in this paper.
Abstract: The fifth edition of the WHO Classification of Tumors of the Central Nervous System (CNS), published in 2021, is the sixth version of the international standard for the classification of brain and spinal cord tumors. Building on the 2016 updated fourth edition and the work of the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy, the 2021 fifth edition introduces major changes that advance the role of molecular diagnostics in CNS tumor classification. At the same time, it remains wedded to other established approaches to tumor diagnosis such as histology and immunohistochemistry. In doing so, the fifth edition establishes some different approaches to both CNS tumor nomenclature and grading and it emphasizes the importance of integrated diagnoses and layered reports. New tumor types and subtypes are introduced, some based on novel diagnostic technologies such as DNA methylome profiling. The present review summarizes the major general changes in the 2021 fifth edition classification and the specific changes in each taxonomic category. It is hoped that this summary provides an overview to facilitate more in-depth exploration of the entire fifth edition of the WHO Classification of Tumors of the Central Nervous System.

2,908 citations

01 Jan 2013
TL;DR: In this article, the landscape of somatic genomic alterations based on multidimensional and comprehensive characterization of more than 500 glioblastoma tumors (GBMs) was described, including several novel mutated genes as well as complex rearrangements of signature receptors, including EGFR and PDGFRA.
Abstract: We describe the landscape of somatic genomic alterations based on multidimensional and comprehensive characterization of more than 500 glioblastoma tumors (GBMs). We identify several novel mutated genes as well as complex rearrangements of signature receptors, including EGFR and PDGFRA. TERT promoter mutations are shown to correlate with elevated mRNA expression, supporting a role in telomerase reactivation. Correlative analyses confirm that the survival advantage of the proneural subtype is conferred by the G-CIMP phenotype, and MGMT DNA methylation may be a predictive biomarker for treatment response only in classical subtype GBM. Integrative analysis of genomic and proteomic profiles challenges the notion of therapeutic inhibition of a pathway as an alternative to inhibition of the target itself. These data will facilitate the discovery of therapeutic and diagnostic target candidates, the validation of research and clinical observations and the generation of unanticipated hypotheses that can advance our molecular understanding of this lethal cancer.

2,616 citations

Journal ArticleDOI
Robert M. Samstein1, Chung-Han Lee1, Chung-Han Lee2, Alexander N. Shoushtari2, Alexander N. Shoushtari1, Matthew D. Hellmann2, Matthew D. Hellmann1, Ronglai Shen1, Yelena Y. Janjigian2, Yelena Y. Janjigian1, David Barron1, Ahmet Zehir1, Emmet Jordan1, Antonio Omuro1, Thomas Kaley1, Sviatoslav M. Kendall1, Robert J. Motzer2, Robert J. Motzer1, A. Ari Hakimi1, Martin H. Voss2, Martin H. Voss1, Paul Russo1, Jonathan E. Rosenberg2, Jonathan E. Rosenberg1, Gopa Iyer2, Gopa Iyer1, Bernard H. Bochner1, Dean F. Bajorin2, Dean F. Bajorin1, Hikmat Al-Ahmadie1, Jamie E. Chaft1, Jamie E. Chaft2, Charles M. Rudin1, Charles M. Rudin2, Gregory J. Riely1, Gregory J. Riely2, Shrujal S. Baxi1, Shrujal S. Baxi2, Alan L. Ho1, Alan L. Ho2, Richard J. Wong1, David G. Pfister2, David G. Pfister1, Jedd D. Wolchok2, Jedd D. Wolchok1, Christopher A. Barker1, Philip H. Gutin1, Cameron Brennan1, Viviane Tabar1, Ingo K. Mellinghoff1, Lisa M. DeAngelis1, Charlotte E. Ariyan1, Nancy Y. Lee1, William D. Tap1, William D. Tap2, Mrinal M. Gounder2, Mrinal M. Gounder1, Sandra P. D'Angelo2, Sandra P. D'Angelo1, Leonard B. Saltz1, Leonard B. Saltz2, Zsofia K. Stadler1, Zsofia K. Stadler2, Howard I. Scher1, Howard I. Scher2, José Baselga1, José Baselga2, Pedram Razavi1, Pedram Razavi2, Christopher A. Klebanoff1, Christopher A. Klebanoff2, Rona Yaeger1, Rona Yaeger2, Neil H. Segal2, Neil H. Segal1, Geoffrey Y. Ku1, Geoffrey Y. Ku2, Ronald P. DeMatteo1, Marc Ladanyi1, Naiyer A. Rizvi3, Michael F. Berger1, Nadeem Riaz1, David B. Solit1, Timothy A. Chan1, Luc G. T. Morris1 
TL;DR: Analysis of advanced cancer patients treated with immune-checkpoint inhibitors shows that tumor mutational burden, as assessed by targeted next-generation sequencing, predicts survival after immunotherapy across multiple cancer types.
Abstract: Immune checkpoint inhibitor (ICI) treatments benefit some patients with metastatic cancers, but predictive biomarkers are needed. Findings in selected cancer types suggest that tumor mutational burden (TMB) may predict clinical response to ICI. To examine this association more broadly, we analyzed the clinical and genomic data of 1,662 advanced cancer patients treated with ICI, and 5,371 non-ICI-treated patients, whose tumors underwent targeted next-generation sequencing (MSK-IMPACT). Among all patients, higher somatic TMB (highest 20% in each histology) was associated with better overall survival. For most cancer histologies, an association between higher TMB and improved survival was observed. The TMB cutpoints associated with improved survival varied markedly between cancer types. These data indicate that TMB is associated with improved survival in patients receiving ICI across a wide variety of cancer types, but that there may not be one universal definition of high TMB.

2,343 citations