Network


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

Hotspot


Dive into the research topics where Kety Huberman is active.

Publication


Featured researches published by Kety Huberman.


The New England Journal of Medicine | 2012

Prognostic relevance of integrated genetic profiling in acute myeloid leukemia

Jay Patel; Mithat Gonen; Maria E. Figueroa; Hugo F. Fernandez; Zhuoxin Sun; Janis Racevskis; Pieter Van Vlierberghe; Igor Dolgalev; Sabrena Thomas; Olga Aminova; Kety Huberman; Janice Cheng; Agnes Viale; Nicholas D. Socci; Adriana Heguy; Athena M. Cherry; Gail H. Vance; Rodney R. Higgins; Rhett P. Ketterling; Robert E. Gallagher; Mark R. Litzow; Marcel R.M. van den Brink; Hillard M. Lazarus; Jacob M. Rowe; Selina M. Luger; Adolfo A. Ferrando; Elisabeth Paietta; Martin S. Tallman; Ari Melnick; Omar Abdel-Wahab

BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous disease with respect to presentation and clinical outcome. The prognostic value of recently identified somatic mutations has not been systematically evaluated in a phase 3 trial of treatment for AML. METHODS We performed a mutational analysis of 18 genes in 398 patients younger than 60 years of age who had AML and who were randomly assigned to receive induction therapy with high-dose or standard-dose daunorubicin. We validated our prognostic findings in an independent set of 104 patients. RESULTS We identified at least one somatic alteration in 97.3% of the patients. We found that internal tandem duplication in FLT3 (FLT3-ITD), partial tandem duplication in MLL (MLL-PTD), and mutations in ASXL1 and PHF6 were associated with reduced overall survival (P=0.001 for FLT3-ITD, P=0.009 for MLL-PTD, P=0.05 for ASXL1, and P=0.006 for PHF6); CEBPA and IDH2 mutations were associated with improved overall survival (P=0.05 for CEBPA and P=0.01 for IDH2). The favorable effect of NPM1 mutations was restricted to patients with co-occurring NPM1 and IDH1 or IDH2 mutations. We identified genetic predictors of outcome that improved risk stratification among patients with AML, independently of age, white-cell count, induction dose, and post-remission therapy, and validated the significance of these predictors in an independent cohort. High-dose daunorubicin, as compared with standard-dose daunorubicin, improved the rate of survival among patients with DNMT3A or NPM1 mutations or MLL translocations (P=0.001) but not among patients with wild-type DNMT3A, NPM1, and MLL (P=0.67). CONCLUSIONS We found that DNMT3A and NPM1 mutations and MLL translocations predicted an improved outcome with high-dose induction chemotherapy in patients with AML. These findings suggest that mutational profiling could potentially be used for risk stratification and to inform prognostic and therapeutic decisions regarding patients with AML. (Funded by the National Cancer Institute and others.).


Blood | 2009

Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies

Omar Abdel-Wahab; Ann Mullally; Cyrus V. Hedvat; Guillermo Garcia-Manero; Jay Patel; Martha Wadleigh; Sébastien Malinge; Jin Juan Yao; Outi Kilpivaara; Rukhmi Bhat; Kety Huberman; Sabrena Thomas; Igor Dolgalev; Adriana Heguy; Elisabeth Paietta; Michelle M. Le Beau; Miloslav Beran; Martin S. Tallman; Benjamin L. Ebert; Hagop M. Kantarjian; Richard Stone; D. Gary Gilliland; John D. Crispino; Ross L. Levine

Disease alleles that activate signal transduction are common in myeloid malignancies; however, there are additional unidentified mutations that contribute to myeloid transformation. Based on the recent identification of TET2 mutations, we evaluated the mutational status of TET1, TET2, and TET3 in myeloproliferative neoplasms (MPNs), chronic myelomonocytic leukemia (CMML), and acute myeloid leukemia (AML). Sequencing of TET2 in 408 paired tumor/normal samples distinguished between 68 somatic mutations and 6 novel single nucleotide polymorphisms and identified TET2 mutations in MPN (27 of 354, 7.6%), CMML (29 of 69, 42%), AML (11 of 91, 12%), and M7 AML (1 of 28, 3.6%) samples. We did not identify somatic TET1 or TET3 mutations or TET2 promoter hypermethylation in MPNs. TET2 mutations did not cluster in genetically defined MPN, CMML, or AML subsets but were associated with decreased overall survival in AML (P = .029). These data indicate that TET2 mutations are observed in different myeloid malignancies and may be important in AML prognosis.


Nature Genetics | 2013

The mutational landscape of adenoid cystic carcinoma

Allen S. Ho; Kasthuri Kannan; David M Roy; Luc G. T. Morris; Ian Ganly; Nora Katabi; Deepa Ramaswami; Logan A. Walsh; Stephanie Eng; Jason T. Huse; Jianan Zhang; Igor Dolgalev; Kety Huberman; Adriana Heguy; Agnes Viale; Marija Drobnjak; Margaret Leversha; Christine E Rice; Bhuvanesh Singh; N. Gopalakrishna Iyer; C. René Leemans; Elisabeth Bloemena; Robert L. Ferris; Raja R. Seethala; Benjamin E. Gross; Yupu Liang; Rileen Sinha; Luke Peng; Benjamin J. Raphael; Sevin Turcan

Adenoid cystic carcinomas (ACCs) are among the most enigmatic of human malignancies. These aggressive salivary gland cancers frequently recur and metastasize despite definitive treatment, with no known effective chemotherapy regimen. Here we determined the ACC mutational landscape and report the exome or whole-genome sequences of 60 ACC tumor-normal pairs. These analyses identified a low exonic somatic mutation rate (0.31 non-silent events per megabase) and wide mutational diversity. Notably, we found mutations in genes encoding chromatin-state regulators, such as SMARCA2, CREBBP and KDM6A, suggesting that there is aberrant epigenetic regulation in ACC oncogenesis. Mutations in genes central to the DNA damage response and protein kinase A signaling also implicate these processes. We observed MYB-NFIB translocations and somatic mutations in MYB-associated genes, solidifying the role of these aberrations as critical events in ACC. Lastly, we identified recurrent mutations in the FGF-IGF-PI3K pathway (30% of tumors) that might represent new avenues for therapy. Collectively, our observations establish a molecular foundation for understanding and exploring new treatments for ACC.


Cancer Research | 2010

Genomic and Biological Characterization of Exon 4 KRAS Mutations in Human Cancer

Manickam Janakiraman; Efsevia Vakiani; Zhaoshi Zeng; Christine A. Pratilas; Barry S. Taylor; Dhananjay Chitale; Ensar Halilovic; Manda Wilson; Kety Huberman; Julio Ricarte Filho; Yogindra Persaud; Douglas A. Levine; James A. Fagin; Suresh C. Jhanwar; John M. Mariadason; Alex E. Lash; Marc Ladanyi; Leonard Saltz; Adriana Heguy; Philip B. Paty; David B. Solit

Mutations in RAS proteins occur widely in human cancer. Prompted by the confirmation of KRAS mutation as a predictive biomarker of response to epidermal growth factor receptor (EGFR)-targeted therapies, limited clinical testing for RAS pathway mutations has recently been adopted. We performed a multiplatform genomic analysis to characterize, in a nonbiased manner, the biological, biochemical, and prognostic significance of Ras pathway alterations in colorectal tumors and other solid tumor malignancies. Mutations in exon 4 of KRAS were found to occur commonly and to predict for a more favorable clinical outcome in patients with colorectal cancer. Exon 4 KRAS mutations, all of which were identified at amino acid residues K117 and A146, were associated with lower levels of GTP-bound RAS in isogenic models. These same mutations were also often accompanied by conversion to homozygosity and increased gene copy number, in human tumors and tumor cell lines. Models harboring exon 4 KRAS mutations exhibited mitogen-activated protein/extracellular signal-regulated kinase kinase dependence and resistance to EGFR-targeted agents. Our findings suggest that RAS mutation is not a binary variable in tumors, and that the diversity in mutant alleles and variability in gene copy number may also contribute to the heterogeneity of clinical outcomes observed in cancer patients. These results also provide a rationale for broader KRAS testing beyond the most common hotspot alleles in exons 2 and 3.


Nature Genetics | 2014

PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors

William R. Lee; Sewit Teckie; Thomas Wiesner; Leili Ran; Carlos N. Prieto Granada; Mingyan Lin; Sinan Zhu; Zhen Cao; Yupu Liang; Andrea Sboner; William D. Tap; Jonathan A. Fletcher; Kety Huberman; Li Xuan Qin; Agnes Viale; Samuel Singer; Deyou Zheng; Michael F. Berger; Yu Chen; Cristina R. Antonescu; Ping Chi

Malignant peripheral nerve sheath tumors (MPNSTs) represent a group of highly aggressive soft-tissue sarcomas that may occur sporadically, in association with neurofibromatosis type I (NF1 associated) or after radiotherapy. Using comprehensive genomic approaches, we identified loss-of-function somatic alterations of the Polycomb repressive complex 2 (PRC2) components (EED or SUZ12) in 92% of sporadic, 70% of NF1-associated and 90% of radiotherapy-associated MPNSTs. MPNSTs with PRC2 loss showed complete loss of trimethylation at lysine 27 of histone H3 (H3K27me3) and aberrant transcriptional activation of multiple PRC2-repressed homeobox master regulators and their regulated developmental pathways. Introduction of the lost PRC2 component in a PRC2-deficient MPNST cell line restored H3K27me3 levels and decreased cell growth. Additionally, we identified frequent somatic alterations of CDKN2A (81% of all MPNSTs) and NF1 (72% of non-NF1-associated MPNSTs), both of which significantly co-occur with PRC2 alterations. The highly recurrent and specific inactivation of PRC2 components, NF1 and CDKN2A highlights their critical and potentially cooperative roles in MPNST pathogenesis.


Clinical Cancer Research | 2012

Phase II trial of temozolomide in patients with relapsed sensitive or refractory small cell lung cancer, with assessment of methylguanine-DNA methyltransferase as a potential biomarker.

Maria Catherine Pietanza; Kyuichi Kadota; Kety Huberman; Camelia S. Sima; Fiore Jj; Sumner Dk; William D. Travis; Adriana Heguy; Michelle S. Ginsberg; Holodny Ai; Timothy A. Chan; Naiyer A. Rizvi; Christopher G. Azzoli; Gregory J. Riely; Mark G. Kris; Lee M. Krug

Purpose: This phase II study was conducted to assess the efficacy of temozolomide in patients with relapsed small cell lung cancer (SCLC). Experimental Design: Patients with disease progression after one or two prior chemotherapy regimens received temozolomide at 75 mg/m2/d for 21 days of a 28-day cycle. The primary endpoint was the overall response rate [ORR; complete response (CR) plus partial response (PR)], which was evaluated separately in sensitive and refractory cohorts. In the available tissue, we assessed O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status by PCR and MGMT expression by immunohistochemistry. Results: Sixty-four patients were accrued: 48 patients in the sensitive cohort and 16 in the refractory group. One CR and 10 PRs were noted in sensitive patients [ORR, 23%; 95% confidence interval (CI), 12%–37%]. Two PRs were seen in the refractory cohort (ORR, 13%; 95% CI, 2%–38%). As second- and third-line treatment, the ORR was 22% (95% CI, 9%–40%) and 19% (95% CI, 7%–36%), respectively. Among patients with target brain lesions, 38% had a CR or PR (95% CI, 14%–68%). Grade ≥3 thrombocytopenia and neutropenia were observed in nine patients (14%). A greater number of cases with methylated MGMT had a response compared to those with unmethylated MGMT (38% vs. 7%; P = 0.08). Conclusion: Temozolomide has activity in relapsed SCLC, particularly for brain metastases. Response to temozolomide may correlate with MGMT methylation in SCLC. Clin Cancer Res; 18(4); 1138–45. ©2012 AACR.


Science Translational Medicine | 2014

Hematopoietic Stem Cell Origin of BRAFV600E Mutations in Hairy Cell Leukemia

Stephen S. Chung; Eunhee Kim; Jae H. Park; Young Rock Chung; Piro Lito; Julie Teruya-Feldstein; Wenhuo Hu; Wendy Béguelin; Sebastien Monette; Cihangir Duy; Raajit Rampal; Leon Telis; Minal Patel; Min-Kyung Kim; Kety Huberman; Nancy Bouvier; Michael F. Berger; Ari Melnick; Neal Rosen; Martin S. Tallman; Christopher Y. Park; Omar Abdel-Wahab

The cell of origin for the chronic lymphoproliferative disorder hairy cell leukemia is a long-term hematopoietic stem cell, as shown through human genetic data and murine genetic models. Finding the Origin Story for a Leukemia The cells that give rise to a cancer called hairy cell leukemia are hematopoietic stem cells, the precursors for all the types of normal blood cells, according to a new study by Chung et al. Although hairy cell leukemia is usually thought to be derived from mature B cells, it has not previously been matched with any specific stage of normal B cell development. Now, the authors performed detailed genetic analysis of human leukemia samples and also modeled this cancer in mice with different types of mutations, thus revealing the origin for hairy cell leukemia. Understanding the causes of this leukemia should help guide the design of effective treatments and may improve our understanding of similar cancers. Hairy cell leukemia (HCL) is a chronic lymphoproliferative disorder characterized by somatic BRAFV600E mutations. The malignant cell in HCL has immunophenotypic features of a mature B cell, but no normal counterpart along the continuum of developing B lymphocytes has been delineated as the cell of origin. We find that the BRAFV600E mutation is present in hematopoietic stem cells (HSCs) in HCL patients, and that these patients exhibit marked alterations in hematopoietic stem/progenitor cell (HSPC) frequencies. Quantitative sequencing analysis revealed a mean BRAFV600E-mutant allele frequency of 4.97% in HSCs from HCL patients. Moreover, transplantation of BRAFV600E-mutant HSCs from an HCL patient into immunodeficient mice resulted in stable engraftment of BRAFV600E-mutant human hematopoietic cells, revealing the functional self-renewal capacity of HCL HSCs. Consistent with the human genetic data, expression of BRafV600E in murine HSPCs resulted in a lethal hematopoietic disorder characterized by splenomegaly, anemia, thrombocytopenia, increased circulating soluble CD25, and increased clonogenic capacity of B lineage cells—all classic features of human HCL. In contrast, restricting expression of BRafV600E to the mature B cell compartment did not result in disease. Treatment of HCL patients with vemurafenib, an inhibitor of mutated BRAF, resulted in normalization of HSPC frequencies and increased myeloid and erythroid output from HSPCs. These findings link the pathogenesis of HCL to somatic mutations that arise in HSPCs and further suggest that chronic lymphoid malignancies may be initiated by aberrant HSCs.


Journal of the National Cancer Institute | 2015

Massively Parallel Sequencing-Based Clonality Analysis of Synchronous Endometrioid Endometrial and Ovarian Carcinomas

Anne M. Schultheis; Charlotte K.Y. Ng; Maria Rosaria De Filippo; Salvatore Piscuoglio; Gabriel S. Macedo; Sonia Gatius; Belen Perez Mies; Robert A. Soslow; Raymond S. Lim; Agnes Viale; Kety Huberman; Jose C. Palacios; Jorge S. Reis-Filho; Xavier Matias-Guiu; Britta Weigelt

Synchronous early-stage endometrioid endometrial carcinomas (EECs) and endometrioid ovarian carcinomas (EOCs) are associated with a favorable prognosis and have been suggested to represent independent primary tumors rather than metastatic disease. We subjected sporadic synchronous EECs/EOCs from five patients to whole-exome massively parallel sequencing, which revealed that the EEC and EOC of each case displayed strikingly similar repertoires of somatic mutations and gene copy number alterations. Despite the presence of mutations restricted to the EEC or EOC in each case, we observed that the mutational processes that shaped their respective genomes were consistent. High-depth targeted massively parallel sequencing of sporadic synchronous EECs/EOCs from 17 additional patients confirmed that these lesions are clonally related. In an additional Lynch Syndrome case, however, the EEC and EOC were found to constitute independent cancers lacking somatic mutations in common. Taken together, sporadic synchronous EECs/EOCs are clonally related and likely constitute dissemination from one site to the other.


European Urology | 2017

Genomic Biomarkers of a Randomized Trial Comparing First-line Everolimus and Sunitinib in Patients with Metastatic Renal Cell Carcinoma

James J. Hsieh; David Chen; Patricia Wang; Mahtab Marker; Almedina Redzematovic; Ying Bei Chen; S. Duygu Selcuklu; Nils Weinhold; Nancy Bouvier; Kety Huberman; Umesh Bhanot; Michael Chevinsky; Parul Patel; Patrizia Pinciroli; Helen H. Won; Daoqi You; Agnes Viale; William R. Lee; A. Ari Hakimi; Michael F. Berger; Nicholas D. Socci; Emily H. Cheng; Jennifer J. Knox; Martin H. Voss; Maurizio Voi; Robert J. Motzer

BACKGROUND Metastatic renal cell carcinoma (RCC) patients are commonly treated with vascular endothelial growth factor (VEGF) inhibitors or mammalian target of rapamycin inhibitors. Correlations between somatic mutations and first-line targeted therapy outcomes have not been reported on a randomized trial. OBJECTIVE To evaluate the relationship between tumor mutations and treatment outcomes in RECORD-3, a randomized trial comparing first-line everolimus (mTOR inhibitor) followed by sunitinib (VEGF inhibitor) at progression with the opposite sequence in 471 metastatic RCC patients. DESIGN, SETTING, AND PARTICIPANTS Targeted sequencing of 341 cancer genes at ∼540× coverage was performed on available tumor samples from 258 patients; 220 with clear cell histology (ccRCC). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Associations between somatic mutations and median first-line progression free survival (PFS1L) and overall survival were determined in metastatic ccRCC using Cox proportional hazards models and log-rank tests. RESULTS AND LIMITATIONS Prevalent mutations (≥ 10%) were VHL (75%), PBRM1 (46%), SETD2 (30%), BAP1 (19%), KDM5C (15%), and PTEN (12%). With first-line everolimus, PBRM1 and BAP1 mutations were associated with longer (median [95% confidence interval {CI}] 12.8 [8.1, 18.4] vs 5.5 [3.1, 8.4] mo) and shorter (median [95% CI] 4.9 [2.9, 8.1] vs 10.5 [7.3, 12.9] mo) PFS1L, respectively. With first-line sunitinib, KDM5C mutations were associated with longer PFS1L (median [95% CI] of 20.6 [12.4, 27.3] vs 8.3 [7.8, 11.0] mo). Molecular subgroups of metastatic ccRCC based on PBRM1, BAP1, and KDM5C mutations could have predictive values for patients treated with VEGF or mTOR inhibitors. Most tumor DNA was obtained from primary nephrectomy samples (94%), which could impact correlation statistics. CONCLUSIONS PBRM1, BAP1, and KDM5C mutations impact outcomes of targeted therapies in metastatic ccRCC patients. PATIENT SUMMARY Large-scale genomic kidney cancer studies reported novel mutations and heterogeneous features among individual tumors, which could contribute to varied clinical outcomes. We demonstrated correlations between somatic mutations and treatment outcomes in clear cell renal cell carcinoma, supporting the value of genomic classification in prospective studies.


Cancer Discovery | 2015

NF2 Loss Promotes Oncogenic RAS-Induced Thyroid Cancers via YAP-Dependent Transactivation of RAS Proteins and Sensitizes Them to MEK Inhibition

Maria E.R. Garcia-Rendueles; Julio C. Ricarte-Filho; Brian R. Untch; Iňigo Landa; Jeffrey A. Knauf; Francesca Voza; Vicki Smith; Ian Ganly; Barry S. Taylor; Yogindra Persaud; Gisele Oler; Yuqiang Fang; Suresh C. Jhanwar; Agnes Viale; Adriana Heguy; Kety Huberman; Filippo G. Giancotti; Ronald Ghossein; James A. Fagin

UNLABELLED Ch22q LOH is preferentially associated with RAS mutations in papillary and in poorly differentiated thyroid cancer (PDTC). The 22q tumor suppressor NF2, encoding merlin, is implicated in this interaction because of its frequent loss of function in human thyroid cancer cell lines. Nf2 deletion or Hras mutation is insufficient for transformation, whereas their combined disruption leads to murine PDTC with increased MAPK signaling. Merlin loss induces RAS signaling in part through inactivation of Hippo, which activates a YAP-TEAD transcriptional program. We find that the three RAS genes are themselves YAP-TEAD1 transcriptional targets, providing a novel mechanism of promotion of RAS-induced tumorigenesis. Moreover, pharmacologic disruption of YAP-TEAD with verteporfin blocks RAS transcription and signaling and inhibits cell growth. The increased MAPK output generated by NF2 loss in RAS-mutant cancers may inform therapeutic strategies, as it generates greater dependency on the MAPK pathway for viability. SIGNIFICANCE Intensification of mutant RAS signaling through copy-number imbalances is commonly associated with transformation. We show that NF2/merlin inactivation augments mutant RAS signaling by promoting YAP/TEAD-driven transcription of oncogenic and wild-type RAS, resulting in greater MAPK output and increased sensitivity to MEK inhibitors.

Collaboration


Dive into the Kety Huberman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Agnes Viale

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nicholas D. Socci

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael F. Berger

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Barry S. Taylor

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nancy Bouvier

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Camelia S. Sima

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gregory J. Riely

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Julie Teruya-Feldstein

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge