Network


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

Hotspot


Dive into the research topics where Pedram Razavi is active.

Publication


Featured researches published by Pedram Razavi.


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.


Cancer Cell | 2016

PDK1-SGK1 Signaling Sustains AKT-Independent mTORC1 Activation and Confers Resistance to PI3Kα Inhibition

Pau Castel; Haley Ellis; Ruzica Bago; Eneda Toska; Pedram Razavi; F. Javier Carmona; Srinivasaraghavan Kannan; Chandra Verma; Maura N. Dickler; Sarat Chandarlapaty; Edi Brogi; Dario R. Alessi; José Baselga; Maurizio Scaltriti

Summary PIK3CA, which encodes the p110α subunit of PI3K, is frequently mutated and oncogenic in breast cancer. PI3Kα inhibitors are in clinical development and despite promising early clinical activity, intrinsic resistance is frequent among patients. We have previously reported that residual downstream mTORC1 activity upon treatment with PI3Kα inhibitors drives resistance to these agents. However, the mechanism underlying this phenotype is not fully understood. Here we show that in cancer cells resistant to PI3Kα inhibition, PDK1 blockade restores sensitivity to these therapies. SGK1, which is activated by PDK1, contributes to the maintenance of residual mTORC1 activity through direct phosphorylation and inhibition of TSC2. Targeting either PDK1 or SGK1 prevents mTORC1 activation, restoring the antitumoral effects of PI3Kα inhibition in resistant cells.


Cancer Discovery | 2017

Activating ESR1 Mutations Differentially Impact the Efficacy of ER Antagonists.

Weiyi Toy; Weir H; Pedram Razavi; Lawson M; Goeppert Au; Mazzola Am; Smith A; Wilson J; Morrow C; Wong Wl; de Stanchina E; Carlson Ke; Martin Ts; Sharmeen Uddin; Li Z; Fanning S; Katzenellenbogen Ja; Greene G; Baselga J; Chandarlapaty S

Recent studies have identified somatic ESR1 mutations in patients with metastatic breast cancer and found some of them to promote estrogen-independent activation of the receptor. The degree to which all recurrent mutants can drive estrogen-independent activities and reduced sensitivity to ER antagonists like fulvestrant is not established. In this report, we characterize the spectrum of ESR1 mutations from more than 900 patients. ESR1 mutations were detected in 10%, with D538G being the most frequent (36%), followed by Y537S (14%). Several novel, activating mutations were also detected (e.g., L469V, V422del, and Y537D). Although many mutations lead to constitutive activity and reduced sensitivity to ER antagonists, only select mutants such as Y537S caused a magnitude of change associated with fulvestrant resistance in vivo Correspondingly, tumors driven by Y537S, but not D5358G, E380Q, or S463P, were less effectively inhibited by fulvestrant than more potent and bioavailable antagonists, including AZD9496. These data point to a need for antagonists with optimal pharmacokinetic properties to realize clinical efficacy against certain ESR1 mutants.Significance: A diversity of activating ESR1 mutations exist, only some of which confer resistance to existing ER antagonists that might be overcome by next-generation inhibitors such as AZD9496. Cancer Discov; 7(3); 277-87. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 235.


Cancer Discovery | 2016

Systematic functional characterization of resistance to PI3K inhibition in breast cancer

Xiuning Le; Rajee Antony; Pedram Razavi; Daniel J. Treacy; Flora Luo; Mahmoud Ghandi; Pau Castel; Maurizio Scaltriti; José Baselga; Levi A. Garraway

PIK3CA (which encodes the PI3K alpha isoform) is the most frequently mutated oncogene in breast cancer. Small-molecule PI3K inhibitors have shown promise in clinical trials; however, intrinsic and acquired resistance limits their utility. We used a systematic gain-of-function approach to identify genes whose upregulation confers resistance to the PI3K inhibitor BYL719 in breast cancer cells. Among the validated resistance genes, Proviral Insertion site in Murine leukemia virus (PIM) kinases conferred resistance by maintaining downstream PI3K effector activation in an AKT-independent manner. Concurrent pharmacologic inhibition of PIM and PI3K overcame this resistance mechanism. We also observed increased PIM expression and activity in a subset of breast cancer biopsies with clinical resistance to PI3K inhibitors. PIM1 overexpression was mutually exclusive with PIK3CA mutation in treatment-naïve breast cancers, suggesting downstream functional redundancy. Together, these results offer new insights into resistance to PI3K inhibitors and support clinical studies of combined PIM/PI3K inhibition in a subset of PIK3CA-mutant cancers. SIGNIFICANCE PIM kinase overexpression confers resistance to small-molecule PI3K inhibitors. Combined inhibition of PIM and PI3K may therefore be warranted in a subset of breast cancers. Cancer Discov; 6(10); 1134-47. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 1069.


Cancer Discovery | 2017

Accelerating Discovery of Functional Mutant Alleles in Cancer

Matthew T. Chang; Tripti Shrestha Bhattarai; Alison M. Schram; Craig M. Bielski; Mark T.A. Donoghue; Philip Jonsson; Debyani Chakravarty; Sarah Phillips; Cyriac Kandoth; Alexander Penson; Alexander N. Gorelick; Tambudzai Shamu; Swati Patel; Christopher C. Harris; Jianjiong Gao; Selcuk Onur Sumer; Ritika Kundra; Pedram Razavi; Bob T. Li; Dalicia Reales; Nicholas D. Socci; Gowtham Jayakumaran; Ahmet Zehir; Ryma Benayed; Maria E. Arcila; Sarat Chandarlapaty; Marc Ladanyi; Nikolaus Schultz; José Baselga; Michael F. Berger

Most mutations in cancer are rare, which complicates the identification of therapeutically significant mutations and thus limits the clinical impact of genomic profiling in patients with cancer. Here, we analyzed 24,592 cancers including 10,336 prospectively sequenced patients with advanced disease to identify mutant residues arising more frequently than expected in the absence of selection. We identified 1,165 statistically significant hotspot mutations of which 80% arose in 1 in 1,000 or fewer patients. Of 55 recurrent in-frame indels, we validated that novel AKT1 duplications induced pathway hyperactivation and conferred AKT inhibitor sensitivity. Cancer genes exhibit different rates of hotspot discovery with increasing sample size, with few approaching saturation. Consequently, 26% of all hotspots in therapeutically actionable oncogenes were novel. Upon matching a subset of affected patients directly to molecularly targeted therapy, we observed radiographic and clinical responses. Population-scale mutant allele discovery illustrates how the identification of driver mutations in cancer is far from complete.Significance: Our systematic computational, experimental, and clinical analysis of hotspot mutations in approximately 25,000 human cancers demonstrates that the long right tail of biologically and therapeutically significant mutant alleles is still incompletely characterized. Sharing prospective genomic data will accelerate hotspot identification, thereby expanding the reach of precision oncology in patients with cancer. Cancer Discov; 8(2); 174-83. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 127.


Nature Genetics | 2018

Genome doubling shapes the evolution and prognosis of advanced cancers

Craig M. Bielski; Ahmet Zehir; Alexander Penson; Mark T.A. Donoghue; Walid K. Chatila; Joshua Armenia; Matthew T. Chang; Alison M. Schram; Philip Jonsson; Chaitanya Bandlamudi; Pedram Razavi; Gopa Iyer; Mark E. Robson; Zsofia K. Stadler; Nikolaus Schultz; José Baselga; David B. Solit; David M. Hyman; Michael F. Berger; Barry S. Taylor

Ploidy abnormalities are a hallmark of cancer, but their impact on the evolution and outcomes of cancers is unknown. Here, we identified whole-genome doubling (WGD) in the tumors of nearly 30% of 9,692 prospectively sequenced advanced cancer patients. WGD varied by tumor lineage and molecular subtype, and arose early in carcinogenesis after an antecedent transforming driver mutation. While associated with TP53 mutations, 46% of all WGD arose in TP53-wild-type tumors and in such cases was associated with an E2F-mediated G1 arrest defect, although neither aberration was obligate in WGD tumors. The variability of WGD across cancer types can be explained in part by cancer cell proliferation rates. WGD predicted for increased morbidity across cancer types, including KRAS-mutant colorectal cancers and estrogen receptor-positive breast cancers, independently of established clinical prognostic factors. We conclude that WGD is highly common in cancer and is a macro-evolutionary event associated with poor prognosis across cancer types.The authors identify whole-genome doubling (WGD) in 30% of ~10,000 sequenced tumors from patients with advanced cancer. WGD correlates with increased risk of death across cancer types.


Journal of Oncology | 2017

Capturing Genomic Evolution of Lung Cancers through Liquid Biopsy for Circulating Tumor DNA

Michael Offin; Jacob J. Chabon; Pedram Razavi; James M. Isbell; Charles M. Rudin; Maximilian Diehn; Bob T. Li

Genetic sequencing of malignancies has become increasingly important to uncover therapeutic targets and capture the tumors dynamic changes to drug sensitivity and resistance through genomic evolution. In lung cancers, the current standard of tissue biopsy at the time of diagnosis and progression is not always feasible or practical and may underestimate intratumoral heterogeneity. Technological advances in genetic sequencing have enabled the use of circulating tumor DNA (ctDNA) analysis to obtain information on both targetable mutations and capturing real-time Darwinian evolution of tumor clones and drug resistance mechanisms under selective therapeutic pressure. The ability to analyze ctDNA from plasma, CSF, or urine enables a comprehensive view of cancers as systemic diseases and captures intratumoral heterogeneity. Here, we describe these recent advances in the setting of lung cancers and advocate for further research and the incorporation of ctDNA analysis in clinical trials of targeted therapies. By capturing genomic evolution in a noninvasive manner, liquid biopsy for ctDNA analysis could accelerate therapeutic discovery and deliver the next leap forward in precision medicine for patients with lung cancers and other solid tumors.


Oncogene | 2018

KMT2C mediates the estrogen dependence of breast cancer through regulation of ERα enhancer function

Kinisha Gala; Qing Li; Amit U. Sinha; Pedram Razavi; Madeline Dorso; Francisco Sanchez-Vega; Young Rock Chung; Ronald C. Hendrickson; James J. Hsieh; Michael F. Berger; Nikolaus Schultz; Alessandro Pastore; Omar Abdel-Wahab; Sarat Chandarlapaty

Estrogen receptor alpha (ERα) is a ligand-activated nuclear receptor that directs proliferation and differentiation in selected cancer cell types including mammary-derived carcinomas. These master-regulatory functions of ERα require trans-acting elements such as the pioneer factor FOXA1 to establish a genomic landscape conducive to ERα control. Here, we identify the H3K4 methyltransferase KMT2C as necessary for hormone-driven ERα activity and breast cancer proliferation. KMT2C knockdown suppresses estrogen-dependent gene expression and causes H3K4me1 and H3K27ac loss selectively at ERα enhancers. Correspondingly, KMT2C loss impairs estrogen-driven breast cancer proliferation but has no effect on ER- breast cells. Whereas KMT2C loss disrupts estrogen-driven proliferation, it conversely promotes tumor outgrowth under hormone-depleted conditions. In accordance, KMT2C is one of the most frequently mutated genes in ER-positive breast cancer with KMT2C deletion correlating with significantly shorter progression-free survival on anti-estrogen therapy. From a therapeutic standpoint, KMT2C-depleted cells that develop hormone-independence retain their dependence on ERα, displaying ongoing sensitivity to ERα antagonists. We conclude that KMT2C is a key regulator of ERα activity whose loss uncouples breast cancer proliferation from hormone abundance.


Cancer Research | 2016

Abstract P6-07-08: Androgen receptor (AR) mutations in a cohort of patients with breast cancer (BC) who have undergone tumor genomic profiling

Ayca Gucalp; Tracy Ann Proverbs-Singh; Pedram Razavi; Sarat Chandarlapaty; Sujata Patil; Dara S. Ross; Ahmet Zehir; José Baselga; C. Hudis; Tiffany A. Traina

Background: AR mutations have been described as a mechanism of resistance to AR antagonists in prostate cancer. There are limited data regarding the presence of AR mutations in breast cancer. We aim to describe the presence of tumor AR mutations in a cohort of patients (pts) with breast cancer who are candidates for targeted cancer therapy, who underwent tumor genomic profiling as part of a clinical trial at MSK (NCT01775072). Methods: MSK-IMPACT is a targeted tumor sequencing assay capable of detecting mutations and other critical genetic aberrations in 410 cancer genes; these data are available through an institutional database. Following IRB approval and using an electronic medical record, we examined the subset of pts in this database with breast cancer for the presence of AR mutations and performed a chart review for clinicopathologic features and outcomes. Statistics are descriptive. Results: As of 03JUN2015, 628 of 4,379 samples that underwent MSK-IMPACT testing since 2012 were from invasive breast cancers. 6 of 628 (1%) harbored AR mutations (Table 1), none of which contribute to a reported or predicted functional alteration. Patient/tumor characteristics are shown in Table 2. Five out of 6 patients recurred between 7.4 and 117.4 months (mo) from the date of initial diagnosis. One patient is without disease recurrence at 3.9mo from diagnosis at date of last contact, 7.9mo ago. Conclusions: AR mutations were uncommon in this dataset of 628 breast cancers. Similarly, TCGA sequencing data has revealed only 12 invasive breast cancers with AR mutations (2.2% of 973 samples). The functional significance of these mutations has not been demonstrated. As tissue from therapeutic studies using AR antagonists for the treatment of breast cancer become available, mutations or amplification in AR may be more readily identified as a potential mechanism of resistance to AR-targeted therapy. Acknowledgments: We gratefully acknowledge the members of the Molecular Diagnostics Service in the Department of Pathology and the Marie-Josee and Henry R. Kravis Center for Molecular Oncology. Citation Format: Gucalp A, Proverbs-Singh TA, Razavi P, Chandarlapaty S, Patil S, Ross DS, Zehir A, Baselga J, Hudis CA, Traina TA. Androgen receptor (AR) mutations in a cohort of patients with breast cancer (BC) who have undergone tumor genomic profiling. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P6-07-08.


Science Signaling | 2018

Neratinib is effective in breast tumors bearing both amplification and mutation of ERBB2 (HER2)

Emiliano Cocco; F. Javier Carmona; Pedram Razavi; Helen H. Won; Yanyan Cai; Valentina Rossi; Carmen Chan; James Cownie; Joanne Soong; Eneda Toska; Sophie G. Shifman; Ivana Sarotto; Peter Savas; Michael J. Wick; Kyriakos P. Papadopoulos; Alyssa Moriarty; Richard E. Cutler; Francesca Avogadri-Connors; Alshad S. Lalani; Richard P. Bryce; Sarat Chandarlapaty; David M. Hyman; David B. Solit; Valentina Boni; Sherene Loi; José Baselga; Michael F. Berger; Filippo Montemurro; Maurizio Scaltriti

Metastatic breast cancer patients with coexistent HER2 mutation and amplification respond to neratinib. Neratinib for resistant metastatic breast cancer Breast cancers with amplification or mutation in the epidermal growth factor receptor (EGFR) family member HER2 are usually treated with targeted inhibitors, but resistance is common. Amplification and mutation of HER2 are generally considered mutually exclusive occurrences in treatment-naïve patients. However, Cocco et al. discovered a small proportion of treatment-naïve and, more often, previously treated patients with metastatic breast cancer in which HER2 amplification and mutation were coincident. It is not yet clear why, but these co-amplified/mutant cells were resistant to currently approved HER2-specific and HER2/EGFR-specific inhibitors but were sensitive to the new pan-EGFR inhibitor neratinib. Neratinib, which inhibits EGFR and HER2, as well as HER3 and HER4, was more effective at blocking the activity of the EGFR pathway and other receptor tyrosine kinases, common modes of resistance in HER2-driven tumors. Patients and mice bearing their tumor cells showed improved survival and even tumor regression on neratinib, suggesting that this may be a treatment option for certain breast cancer patients. Mutations in ERBB2, the gene encoding epidermal growth factor receptor (EGFR) family member HER2, are common in and drive the growth of “HER2-negative” (not ERBB2 amplified) tumors but are rare in “HER2-positive” (ERBB2 amplified) breast cancer. We analyzed DNA-sequencing data from HER2-positive patients and used cell lines and a patient-derived xenograft model to test the consequence of HER2 mutations on the efficacy of anti-HER2 agents such as trastuzumab, lapatinib, and neratinib, an irreversible pan-EGFR inhibitor. HER2 mutations were present in ~7% of HER2-positive tumors, all of which were metastatic but not all were previously treated. Compared to HER2 amplification alone, in both patients and cultured cell lines, the co-occurrence of HER2 mutation and amplification was associated with poor response to trastuzumab and lapatinib, the standard-of-care anti-HER2 agents. In mice, xenografts established from a patient whose HER2-positive tumor acquired a D769Y mutation in HER2 after progression on trastuzumab-based therapy were resistant to trastuzumab or lapatinib but were sensitive to neratinib. Clinical data revealed that six heavily pretreated patients with tumors bearing coincident HER2 amplification and mutation subsequently exhibited a statistically significant response to neratinib monotherapy. Thus, these findings indicate that coincident HER2 mutation reduces the efficacy of therapies commonly used to treat HER2-positive breast cancer, particularly in metastatic and previously HER2 inhibitor–treated patients, as well as potentially in patients scheduled for first-line treatment. Therefore, we propose that clinical studies testing the efficacy of neratinib are warranted selectively in breast cancer patients whose tumors carry both amplification and mutation of ERBB2/HER2.

Collaboration


Dive into the Pedram Razavi's collaboration.

Top Co-Authors

Avatar

Michael F. Berger

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

José Baselga

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ahmet Zehir

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sarat Chandarlapaty

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Bob T. Li

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David B. Solit

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David M. Hyman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dara S. Ross

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Maurizio Scaltriti

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lillian Mary Smyth

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge