Network


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

Hotspot


Dive into the research topics where Sasinya N. Scott is active.

Publication


Featured researches published by Sasinya N. Scott.


Science | 2012

Genome Sequencing Identifies a Basis for Everolimus Sensitivity

Gopa Iyer; Aphrothiti J. Hanrahan; Matthew I. Milowsky; Hikmat Al-Ahmadie; Sasinya N. Scott; Manickam Janakiraman; Mono Pirun; Chris Sander; Nicholas D. Socci; Irina Ostrovnaya; Agnes Viale; Adriana Heguy; Luke Peng; Timothy A. Chan; Bernard H. Bochner; Dean F. Bajorin; Michael F. Berger; Barry S. Taylor; David B. Solit

Tumor genome sequencing reveals the molecular basis of a patient’s unexpected and dramatic response to a cancer drug. Cancer drugs often induce dramatic responses in a small minority of patients. We used whole-genome sequencing to investigate the genetic basis of a durable remission of metastatic bladder cancer in a patient treated with everolimus, a drug that inhibits the mTOR (mammalian target of rapamycin) signaling pathway. Among the somatic mutations was a loss-of-function mutation in TSC1 (tuberous sclerosis complex 1), a regulator of mTOR pathway activation. Targeted sequencing revealed TSC1 mutations in about 8% of 109 additional bladder cancers examined, and TSC1 mutation correlated with everolimus sensitivity. These results demonstrate the feasibility of using whole-genome sequencing in the clinical setting to identify previously occult biomarkers of drug sensitivity that can aid in the identification of patients most likely to respond to targeted anticancer drugs.


The Journal of Molecular Diagnostics | 2015

Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): A Hybridization Capture-Based Next-Generation Sequencing Clinical Assay for Solid Tumor Molecular Oncology

Donavan T. Cheng; Talia Mitchell; Ahmet Zehir; Ronak Shah; Ryma Benayed; Aijazuddin Syed; Raghu Chandramohan; Zhen Yu Liu; Helen H. Won; Sasinya N. Scott; A. Rose Brannon; Catherine O'Reilly; Justyna Sadowska; Jacklyn Casanova; Angela Yannes; Jaclyn F. Hechtman; Jinjuan Yao; Wei Song; Dara S. Ross; Alifya Oultache; Snjezana Dogan; Laetitia Borsu; Meera Hameed; Khedoudja Nafa; Maria E. Arcila; Marc Ladanyi; Michael F. Berger

The identification of specific genetic alterations as key oncogenic drivers and the development of targeted therapies are together transforming clinical oncology and creating a pressing need for increased breadth and throughput of clinical genotyping. Next-generation sequencing assays allow the efficient and unbiased detection of clinically actionable mutations. To enable precision oncology in patients with solid tumors, we developed Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT), a hybridization capture-based next-generation sequencing assay for targeted deep sequencing of all exons and selected introns of 341 key cancer genes in formalin-fixed, paraffin-embedded tumors. Barcoded libraries from patient-matched tumor and normal samples were captured, sequenced, and subjected to a custom analysis pipeline to identify somatic mutations. Sensitivity, specificity, reproducibility of MSK-IMPACT were assessed through extensive analytical validation. We tested 284 tumor samples with previously known point mutations and insertions/deletions in 47 exons of 19 cancer genes. All known variants were accurately detected, and there was high reproducibility of inter- and intrarun replicates. The detection limit for low-frequency variants was approximately 2% for hotspot mutations and 5% for nonhotspot mutations. Copy number alterations and structural rearrangements were also reliably detected. MSK-IMPACT profiles oncogenic DNA alterations in clinical solid tumor samples with high accuracy and sensitivity. Paired analysis of tumors and patient-matched normal samples enables unambiguous detection of somatic mutations to guide treatment decisions.


Nature Genetics | 2014

Recurrent SMARCA4 mutations in small cell carcinoma of the ovary

Petar Jelinic; J.J. Mueller; Narciso Olvera; Fanny Dao; Sasinya N. Scott; Ronak Shah; Jianjiong Gao; Nikolaus Schultz; Mithat Gonen; Robert A. Soslow; Michael F. Berger; Douglas A. Levine

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare, highly aggressive form of ovarian cancer primarily diagnosed in young women. We identified inactivating biallelic SMARCA4 mutations in 100% of the 12 SCCOHT tumors examined. Protein studies confirmed loss of SMARCA4 expression, suggesting a key role for the SWI/SNF chromatin-remodeling complex in SCCOHT.


Clinical Cancer Research | 2014

Tumor Genetic Analyses of Patients with Metastatic Renal Cell Carcinoma and Extended Benefit from mTOR Inhibitor Therapy

Martin H. Voss; A. Ari Hakimi; Can G. Pham; A. Rose Brannon; Ying-Bei Chen; Luis F. Cunha; Oguz Akin; Han Liu; Shugaku Takeda; Sasinya N. Scott; Nicholas D. Socci; Agnes Viale; Nikolaus Schultz; Chris Sander; Victor E. Reuter; Paul Russo; Emily H. Cheng; Robert J. Motzer; Michael F. Berger; James J. Hsieh

Purpose: Rapalogs are allosteric mTOR inhibitors and approved agents for advanced kidney cancer. Reports of clonal heterogeneity in this disease challenge the concept of targeted monotherapy, yet a small subset of patients derives extended benefit. Our aim was to analyze such outliers and explore the genomic background of extreme rapalog sensitivity in the context of intratumor heterogeneity. Experimental Design: We analyzed archived tumor tissue of 5 patients with renal cell carcinoma, who previously achieved durable disease control with rapalogs (median duration, 28 months). DNA was extracted from spatially separate areas of primary tumors and metastases. Custom target capture and ultradeep sequencing was used to identify alterations across 230 target genes. Whole-exome sequence analysis was added to investigate genes beyond this original target list. Results: Five long-term responders contributed 14 specimens to explore clonal heterogeneity. Genomic alterations with activating effect on mTOR signaling were detected in 11 of 14 specimens, offering plausible explanation for exceptional treatment response through alterations in two genes (TSC1 and MTOR). In two subjects, distinct yet functionally convergent alterations activated the mTOR pathway in spatially separate sites. In 1 patient, concurrent genomic events occurred in two separate pathway components across different tumor regions. Conclusions: Analysis of outlier cases can facilitate identification of potential biomarkers for targeted agents, and we implicate two genes as candidates for further study in this class of drugs. The previously reported phenomenon of clonal convergence can occur within a targetable pathway which might have implications for biomarker development beyond this disease and this class of agents. Clin Cancer Res; 20(7); 1955–64. ©2014 AACR.


Genome Biology | 2014

Comparative sequencing analysis reveals high genomic concordance between matched primary and metastatic colorectal cancer lesions

A. Rose Brannon; Efsevia Vakiani; Brooke E. Sylvester; Sasinya N. Scott; Gregory McDermott; Ronak Shah; Krishan Kania; Agnes Viale; Dayna Oschwald; Vladimir Vacic; Anne-Katrin Emde; Andrea Cercek; Rona Yaeger; Nancy E. Kemeny; Leonard Saltz; Jinru Shia; Michael I. D’Angelica; Martin R. Weiser; David B. Solit; Michael F. Berger

BackgroundColorectal cancer is the second leading cause of cancer death in the United States, with over 50,000 deaths estimated in 2014. Molecular profiling for somatic mutations that predict absence of response to anti-EGFR therapy has become standard practice in the treatment of metastatic colorectal cancer; however, the quantity and type of tissue available for testing is frequently limited. Further, the degree to which the primary tumor is a faithful representation of metastatic disease has been questioned. As next-generation sequencing technology becomes more widely available for clinical use and additional molecularly targeted agents are considered as treatment options in colorectal cancer, it is important to characterize the extent of tumor heterogeneity between primary and metastatic tumors.ResultsWe performed deep coverage, targeted next-generation sequencing of 230 key cancer-associated genes for 69 matched primary and metastatic tumors and normal tissue. Mutation profiles were 100% concordant for KRAS, NRAS, and BRAF, and were highly concordant for recurrent alterations in colorectal cancer. Additionally, whole genome sequencing of four patient trios did not reveal any additional site-specific targetable alterations.ConclusionsColorectal cancer primary tumors and metastases exhibit high genomic concordance. As current clinical practices in colorectal cancer revolve around KRAS, NRAS, and BRAF mutation status, diagnostic sequencing of either primary or metastatic tissue as available is acceptable for most patients. Additionally, consistency between targeted sequencing and whole genome sequencing results suggests that targeted sequencing may be a suitable strategy for clinical diagnostic applications.


Clinical Cancer Research | 2013

Phase II trial of MEK inhibitor selumetinib (AZD6244, ARRY-142886) in patients with BRAFV600E/K- mutated melanoma

Federica Catalanotti; David B. Solit; Melissa Pulitzer; Michael F. Berger; Sasinya N. Scott; Tunc Iyriboz; Mario E. Lacouture; Katherine S. Panageas; Jedd D. Wolchok; Richard D. Carvajal; Gary K. Schwartz; Neal Rosen; Paul B. Chapman

Purpose: Test the hypothesis that in BRAF-mutated melanomas, clinical responses to selumetinib, a MEK inhibitor, will be restricted to tumors in which the PI3K/AKT pathway is not activated. Experimental Design: We conducted a phase II trial in patients with melanoma whose tumors harbored a BRAF mutation. Patients were stratified by phosphorylated-AKT (pAKT) expression (high vs. low) and treated with selumetinib 75 mg per os twice daily. Pretreatment tumors were also analyzed for genetic changes in 230 genes of interest using an exon-capture approach. Results: The high pAKT cohort was closed after no responses were seen in the first 10 patients. The incidence of low pAKT melanoma tumors was low (∼25% of melanomas tested) and this cohort was eventually closed because of poor accrual. However, among the five patients with melanoma accrued in the low pAKT cohort, there was one partial response (PR). Two other patients had near PRs before undergoing surgical resection of residual disease (one patient) or discontinuation of treatment due to toxicity (one patient). Among the two nonresponding, low pAKT patients with melanoma, co-mutations in MAP2K1, NF1, and/or EGFR were detected. Conclusions: Tumor regression was seen in three of five patients with BRAF-mutated, low pAKT melanomas; no responses were seen in the high pAKT cohort. These results provide rationale for co-targeting MEK and PI3K/AKT in patients with BRAF mutant melanoma whose tumors express high pAKT. However, the complexity of genetic changes in melanoma indicates that additional genetic information will be needed for optimal selection of patients likely to respond to MEK inhibitors. Clin Cancer Res; 19(8); 2257–64. ©2013 AACR.


The New England Journal of Medicine | 2015

Targeting Mutant BRAF in Relapsed or Refractory Hairy-Cell Leukemia

Enrico Tiacci; Jae H. Park; Luca De Carolis; Stephen S. Chung; Alessandro Broccoli; Sasinya N. Scott; Francesco Zaja; Sean M. Devlin; Alessandro Pulsoni; Young Rock Chung; Michele Cimminiello; Eunhee Kim; Davide Rossi; Richard Stone; Giovanna Motta; Alan Saven; Marzia Varettoni; Jessica K. Altman; Antonella Anastasia; Michael R. Grever; Achille Ambrosetti; Kanti R. Rai; Vincenzo Fraticelli; Mario E. Lacouture; Angelo Michele Carella; Ross L. Levine; Pietro Leoni; Alessandro Rambaldi; Franca Falzetti; Stefano Ascani

BACKGROUND BRAF V600E is the genetic lesion underlying hairy-cell leukemia. We assessed the safety and activity of the oral BRAF inhibitor vemurafenib in patients with hairy-cell leukemia that had relapsed after treatment with a purine analogue or who had disease that was refractory to purine analogues. METHODS We conducted two phase 2, single-group, multicenter studies of vemurafenib (at a dose of 960 mg twice daily)--one in Italy and one in the United States. The therapy was administered for a median of 16 weeks in the Italian study and 18 weeks in the U.S. study. Primary end points were the complete response rate (in the Italian trial) and the overall response rate (in the U.S. trial). Enrollment was completed (28 patients) in the Italian trial in April 2013 and is still open (26 of 36 planned patients) in the U.S. trial. RESULTS The overall response rates were 96% (25 of 26 patients who could be evaluated) after a median of 8 weeks in the Italian study and 100% (24 of 24) after a median of 12 weeks in the U.S. study. The rates of complete response were 35% (9 of 26 patients) and 42% (10 of 24) in the two trials, respectively. In the Italian trial, after a median follow-up of 23 months, the median relapse-free survival was 19 months among patients with a complete response and 6 months among those with a partial response; the median treatment-free survival was 25 months and 18 months, respectively. In the U.S. trial, at 1 year, the progression-free survival rate was 73% and the overall survival rate was 91%. Drug-related adverse events were usually of grade 1 or 2, and the events most frequently leading to dose reductions were rash and arthralgia or arthritis. Secondary cutaneous tumors (treated with simple excision) developed in 7 of 50 patients. The frequent persistence of phosphorylated ERK-positive leukemic cells in bone marrow at the end of treatment suggests bypass reactivation of MEK and ERK as a resistance mechanism. CONCLUSIONS A short oral course of vemurafenib was highly effective in patients with relapsed or refractory hairy-cell leukemia. (Funded by the Associazione Italiana per la Ricerca sul Cancro and others; EudraCT number, 2011-005487-13; ClinicalTrials.gov number NCT01711632.).


Cancer Discovery | 2014

Synthetic Lethality in ATM-Deficient RAD50-Mutant Tumors Underlies Outlier Response to Cancer Therapy

Hikmat Al-Ahmadie; Gopa Iyer; Marcel Hohl; Saurabh Asthana; Akiko Inagaki; Nikolaus Schultz; Aphrothiti J. Hanrahan; Sasinya N. Scott; A. Rose Brannon; Gregory McDermott; Mono Pirun; Irina Ostrovnaya; Philip H. Kim; Nicholas D. Socci; Agnes Viale; Gary K. Schwartz; Victor E. Reuter; Bernard H. Bochner; Jonathan E. Rosenberg; Dean F. Bajorin; Michael F. Berger; John H.J. Petrini; David B. Solit; Barry S. Taylor

UNLABELLED Metastatic solid tumors are almost invariably fatal. Patients with disseminated small-cell cancers have a particularly unfavorable prognosis, with most succumbing to their disease within two years. Here, we report on the genetic and functional analysis of an outlier curative response of a patient with metastatic small-cell cancer to combined checkpoint kinase 1 (CHK1) inhibition and DNA-damaging chemotherapy. Whole-genome sequencing revealed a clonal hemizygous mutation in the Mre11 complex gene RAD50 that attenuated ATM signaling which in the context of CHK1 inhibition contributed, via synthetic lethality, to extreme sensitivity to irinotecan. As Mre11 mutations occur in a diversity of human tumors, the results suggest a tumor-specific combination therapy strategy in which checkpoint inhibition in combination with DNA-damaging chemotherapy is synthetically lethal in tumor cells but not normal cells with somatic mutations that impair Mre11 complex function. SIGNIFICANCE Strategies to effect deep and lasting responses to cancer therapy in patients with metastatic disease have remained difficult to attain, especially in early-phase clinical trials. Here, we present an in-depth genomic and functional genetic analysis identifying RAD50 hypomorphism as a contributing factor to a curative response to systemic combination therapy in a patient with recurrent, metastatic small-cell cancer.


Nature | 2015

Alternative transcription initiation leads to expression of a novel ALK isoform in cancer

Thomas Wiesner; William R. Lee; Anna C. Obenauf; Leili Ran; Rajmohan Murali; Qi Fan Zhang; Elissa W.P. Wong; Wenhuo Hu; Sasinya N. Scott; Ronak Shah; Iñigo Landa; Julia Button; Nathalie Lailler; Andrea Sboner; Dong Gao; Devan Murphy; Zhen Cao; Shipra Shukla; Travis J. Hollmann; Lu Wang; Laetitia Borsu; Taha Merghoub; Gary K. Schwartz; Michael A. Postow; Charlotte E. Ariyan; James A. Fagin; Deyou Zheng; Marc Ladanyi; Michael F. Berger; Yu Chen

Activation of oncogenes by mechanisms other than genetic aberrations such as mutations, translocations, or amplifications is largely undefined. Here we report a novel isoform of the anaplastic lymphoma kinase (ALK) that is expressed in ∼11% of melanomas and sporadically in other human cancer types, but not in normal tissues. The novel ALK transcript initiates from a de novo alternative transcription initiation (ATI) site in ALK intron 19, and was termed ALKATI. In ALKATI-expressing tumours, the ATI site is enriched for H3K4me3 and RNA polymerase II, chromatin marks characteristic of active transcription initiation sites. ALKATI is expressed from both ALK alleles, and no recurrent genetic aberrations are found at the ALK locus, indicating that the transcriptional activation is independent of genetic aberrations at the ALK locus. The ALKATI transcript encodes three proteins with molecular weights of 61.1, 60.8 and 58.7 kilodaltons, consisting primarily of the intracellular tyrosine kinase domain. ALKATI stimulates multiple oncogenic signalling pathways, drives growth-factor-independent cell proliferation in vitro, and promotes tumorigenesis in vivo in mouse models. ALK inhibitors can suppress the kinase activity of ALKATI, suggesting that patients with ALKATI-expressing tumours may benefit from ALK inhibitors. Our findings suggest a novel mechanism of oncogene activation in cancer through de novo alternative transcription initiation.


European Urology | 2015

Genomic Predictors of Survival in Patients with High-grade Urothelial Carcinoma of the Bladder

Philip H. Kim; Eugene K. Cha; John P. Sfakianos; Gopa Iyer; Emily C. Zabor; Sasinya N. Scott; Irina Ostrovnaya; Ricardo Ramirez; Arony Sun; Ronak Shah; Alyssa Yee; Victor E. Reuter; Dean F. Bajorin; Jonathan E. Rosenberg; Nikolaus Schultz; Michael F. Berger; Hikmat Al-Ahmadie; David B. Solit; Bernard H. Bochner

UNLABELLED Urothelial carcinoma of the bladder (UCB) is genomically heterogeneous, with frequent alterations in genes regulating chromatin state, cell cycle control, and receptor kinase signaling. To identify prognostic genomic markers in high-grade UCB, we used capture-based massively parallel sequencing to analyze 109 tumors. Mutations were detected in 240 genes, with 23 genes mutated in ≥5% of cases. The presence of a recurrent phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA) mutation was associated with improved recurrence-free survival (RFS) (hazard ratio [HR]: 0.35; p=0.014) and improved cancer-specific survival (CSS) (HR: 0.35; p=0.040) in patients treated with radical cystectomy (RC). In multivariable analyses controlling for pT and pN stages, PIK3CA mutation remained associated with RFS (HR: 0.39; p=0.032). The most frequent alteration, TP53 mutation (57%), was more common in extravesical disease (69% vs 32%, p=0.005) and lymph node-positive disease (77% vs 56%, p=0.025). Patients with cyclin-dependent kinase inhibitor 2A (CDKN2A)-altered tumors experienced worse RFS (HR: 5.76; p<0.001) and worse CSS (HR: 2.94; p=0.029) in multivariable analyses. Mutations in chromatin-modifying genes were highly prevalent but not associated with outcomes. In UCB patients treated with RC, PIK3CA mutations are associated with favorable outcomes, whereas TP53 and CDKN2A alterations are associated with poor outcomes. Genomic profiling may aid in the identification of UCB patients at highest risk following RC. PATIENT SUMMARY Using next-generation sequencing, we identified genomic subsets of high-grade urothelial bladder cancer associated with favorable and unfavorable outcomes. These findings may aid in the selection of patients most likely to benefit from novel combined modality approaches.

Collaboration


Dive into the Sasinya N. Scott's collaboration.

Top Co-Authors

Avatar

Michael F. Berger

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

Bernard H. Bochner

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dean F. Bajorin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hikmat Al-Ahmadie

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gopa Iyer

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jonathan E. Rosenberg

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Eugene K. Cha

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ronak Shah

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John Sfakianos

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge