Network


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

Hotspot


Dive into the research topics where Azra H. Ligon is active.

Publication


Featured researches published by Azra H. Ligon.


Nature Genetics | 1997

Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers

Peter A. Steck; Mark A. Pershouse; Samar A. Jasser; W. K. Alfred Yung; Huai Lin; Azra H. Ligon; Lauren A. Langford; Michelle L. Baumgard; Thomas Hattier; Thaylon Davis; Cheryl Frye; Rong Hu; Bradley D. Swedlund; David H.-F. Teng; Scan V. Tavtigian

Deletions involving regions of chromosome 10 occur in the vast majority (>90%) of human glioblastoma multiformes. A region at chromosome 10q23–24 was implicated to contain a tumour suppressor gene and the identification of homozygous deletions in four glioma cell lines further refined the location. We have identified a gene, designated MMAC1, that spans these deletions and encodes a widely expressed 5.5-kb mRNA. The predicted MMAC1 protein contains sequence motifs with significant homology to the catalytic domain of protein phosphatases and to the cytoskeletal proteins, tensin and auxilin. MMAC1 coding-region mutations were observed in a number of glioma, prostate, kidney and breast carcinoma cell lines or tumour specimens. Our results identify a strong candidate tumour suppressor gene at chromosome 10q23.3, whose loss of function appears to be associated with the oncogenesis of multiple human cancers.


Nature | 2010

The landscape of somatic copy-number alteration across human cancers

Rameen Beroukhim; Craig H. Mermel; Dale Porter; Guo Wei; Soumya Raychaudhuri; Jerry Donovan; Jordi Barretina; Jesse S. Boehm; Jennifer Dobson; Mitsuyoshi Urashima; Kevin T. Mc Henry; Reid M. Pinchback; Azra H. Ligon; Yoon-Jae Cho; Leila Haery; Heidi Greulich; Michael R. Reich; Wendy Winckler; Michael S. Lawrence; Barbara A. Weir; Kumiko Tanaka; Derek Y. Chiang; Adam J. Bass; Alice Loo; Carter Hoffman; John R. Prensner; Ted Liefeld; Qing Gao; Derek Yecies; Sabina Signoretti

A powerful way to discover key genes with causal roles in oncogenesis is to identify genomic regions that undergo frequent alteration in human cancers. Here we present high-resolution analyses of somatic copy-number alterations (SCNAs) from 3,131 cancer specimens, belonging largely to 26 histological types. We identify 158 regions of focal SCNA that are altered at significant frequency across several cancer types, of which 122 cannot be explained by the presence of a known cancer target gene located within these regions. Several gene families are enriched among these regions of focal SCNA, including the BCL2 family of apoptosis regulators and the NF-κΒ pathway. We show that cancer cells containing amplifications surrounding the MCL1 and BCL2L1 anti-apoptotic genes depend on the expression of these genes for survival. Finally, we demonstrate that a large majority of SCNAs identified in individual cancer types are present in several cancer types.


The New England Journal of Medicine | 2015

PD-1 Blockade with Nivolumab in Relapsed or Refractory Hodgkin's Lymphoma

Stephen M. Ansell; Alexander M. Lesokhin; Ivan Borrello; Ahmad Halwani; Emma C. Scott; Martin Gutierrez; Stephen J. Schuster; Michael Millenson; Deepika Cattry; Gordon J. Freeman; Scott J. Rodig; Bjoern Chapuy; Azra H. Ligon; Lili Zhu; Joseph F. Grosso; Su Y oung Kim; John M. Timmerman; Margaret A. Shipp; Philippe Armand

BACKGROUND Preclinical studies suggest that Reed-Sternberg cells exploit the programmed death 1 (PD-1) pathway to evade immune detection. In classic Hodgkins lymphoma, alterations in chromosome 9p24.1 increase the abundance of the PD-1 ligands, PD-L1 and PD-L2, and promote their induction through Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling. We hypothesized that nivolumab, a PD-1-blocking antibody, could inhibit tumor immune evasion in patients with relapsed or refractory Hodgkins lymphoma. METHODS In this ongoing study, 23 patients with relapsed or refractory Hodgkins lymphoma that had already been heavily treated received nivolumab (at a dose of 3 mg per kilogram of body weight) every 2 weeks until they had a complete response, tumor progression, or excessive toxic effects. Study objectives were measurement of safety and efficacy and assessment of the PDL1 and PDL2 (also called CD274 and PDCD1LG2, respectively) loci and PD-L1 and PD-L2 protein expression. RESULTS Of the 23 study patients, 78% were enrolled in the study after a relapse following autologous stem-cell transplantation and 78% after a relapse following the receipt of brentuximab vedotin. Drug-related adverse events of any grade and of grade 3 occurred in 78% and 22% of patients, respectively. An objective response was reported in 20 patients (87%), including 17% with a complete response and 70% with a partial response; the remaining 3 patients (13%) had stable disease. The rate of progression-free survival at 24 weeks was 86%; 11 patients were continuing to participate in the study. Reasons for discontinuation included stem-cell transplantation (in 6 patients), disease progression (in 4 patients), and drug toxicity (in 2 patients). Analyses of pretreatment tumor specimens from 10 patients revealed copy-number gains in PDL1 and PDL2 and increased expression of these ligands. Reed-Sternberg cells showed nuclear positivity of phosphorylated STAT3, indicative of active JAK-STAT signaling. CONCLUSIONS Nivolumab had substantial therapeutic activity and an acceptable safety profile in patients with previously heavily treated relapsed or refractory Hodgkins lymphoma. (Funded by Bristol-Myers Squibb and others; ClinicalTrials.gov number, NCT01592370.).


Nature Genetics | 2011

Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis

Jason W. Locasale; Alexandra R. Grassian; Tamar Melman; Costas A. Lyssiotis; Katherine R. Mattaini; Adam J. Bass; Gregory J. Heffron; Christian M. Metallo; Taru A. Muranen; Hadar Sharfi; Atsuo T. Sasaki; Dimitrios Anastasiou; Edouard Mullarky; Natalie I. Vokes; Mika Sasaki; Rameen Beroukhim; Gregory Stephanopoulos; Azra H. Ligon; Matthew Meyerson; Andrea L. Richardson; Lynda Chin; Gerhard Wagner; John M. Asara; Joan S. Brugge; Lewis C. Cantley; Matthew G. Vander Heiden

Most tumors exhibit increased glucose metabolism to lactate, however, the extent to which glucose-derived metabolic fluxes are used for alternative processes is poorly understood. Using a metabolomics approach with isotope labeling, we found that in some cancer cells a relatively large amount of glycolytic carbon is diverted into serine and glycine metabolism through phosphoglycerate dehydrogenase (PHGDH). An analysis of human cancers showed that PHGDH is recurrently amplified in a genomic region of focal copy number gain most commonly found in melanoma. Decreasing PHGDH expression impaired proliferation in amplified cell lines. Increased expression was also associated with breast cancer subtypes, and ectopic expression of PHGDH in mammary epithelial cells disrupted acinar morphogenesis and induced other phenotypic alterations that may predispose cells to transformation. Our findings show that the diversion of glycolytic flux into a specific alternate pathway can be selected during tumor development and may contribute to the pathogenesis of human cancer.


Blood | 2010

Recurrent BRAF mutations in Langerhans cell histiocytosis

Gayane Badalian-Very; Jo-Anne Vergilio; Barbara A. Degar; Laura E. MacConaill; Barbara Brandner; Monica L. Calicchio; Frank C. Kuo; Azra H. Ligon; Kristen E. Stevenson; Sarah M. Kehoe; Levi A. Garraway; William C. Hahn; Matthew Meyerson; Mark D. Fleming; Barrett J. Rollins

Langerhans cell histiocytosis (LCH) has a broad spectrum of clinical behaviors; some cases are self-limited, whereas others involve multiple organs and cause significant mortality. Although Langerhans cells in LCH are clonal, their benign morphology and their lack (to date) of reported recurrent genomic abnormalities have suggested that LCH may not be a neoplasm. Here, using 2 orthogonal technologies for detecting cancer-associated mutations in formalin-fixed, paraffin-embedded material, we identified the oncogenic BRAF V600E mutation in 35 of 61 archived specimens (57%). TP53 and MET mutations were also observed in one sample each. BRAF V600E tended to appear in younger patients but was not associated with disease site or stage. Langerhans cells stained for phospho-mitogen-activated protein kinase kinase (phospho-MEK) and phospho-extracellular signal-regulated kinase (phospho-ERK) regardless of mutation status. High prevalence, recurrent BRAF mutations in LCH indicate that it is a neoplastic disease that may respond to RAF pathway inhibitors.


Journal of Clinical Oncology | 2011

Integrative Genomic Analysis of Medulloblastoma Identifies a Molecular Subgroup That Drives Poor Clinical Outcome

Yoon-Jae Cho; Aviad Tsherniak; Pablo Tamayo; Sandro Santagata; Azra H. Ligon; Heidi Greulich; Rameen Berhoukim; Vladimir Amani; Liliana Goumnerova; Charles G. Eberhart; Ching C. Lau; James M. Olson; Richard J. Gilbertson; Amar Gajjar; Olivier Delattre; Marcel Kool; Keith L. Ligon; Matthew Meyerson; Jill P. Mesirov; Scott L. Pomeroy

PURPOSE Medulloblastomas are heterogeneous tumors that collectively represent the most common malignant brain tumor in children. To understand the molecular characteristics underlying their heterogeneity and to identify whether such characteristics represent risk factors for patients with this disease, we performed an integrated genomic analysis of a large series of primary tumors. PATIENTS AND METHODS We profiled the mRNA transcriptome of 194 medulloblastomas and performed high-density single nucleotide polymorphism array and miRNA analysis on 115 and 98 of these, respectively. Non-negative matrix factorization-based clustering of mRNA expression data was used to identify molecular subgroups of medulloblastoma; DNA copy number, miRNA profiles, and clinical outcomes were analyzed for each. We additionally validated our findings in three previously published independent medulloblastoma data sets. RESULTS Identified are six molecular subgroups of medulloblastoma, each with a unique combination of numerical and structural chromosomal aberrations that globally influence mRNA and miRNA expression. We reveal the relative contribution of each subgroup to clinical outcome as a whole and show that a previously unidentified molecular subgroup, characterized genetically by c-MYC copy number gains and transcriptionally by enrichment of photoreceptor pathways and increased miR-183∼96∼182 expression, is associated with significantly lower rates of event-free and overall survivals. CONCLUSION Our results detail the complex genomic heterogeneity of medulloblastomas and identify a previously unrecognized molecular subgroup with poor clinical outcome for which more effective therapeutic strategies should be developed.


Nature | 2008

CDK8 is a colorectal cancer oncogene that regulates β-catenin activity

Ron Firestein; Adam J. Bass; So Young Kim; Ian F. Dunn; Serena J. Silver; Isil Guney; Ellen Freed; Azra H. Ligon; Natalie Vena; Shuji Ogino; Milan G. Chheda; Pablo Tamayo; Stephen Finn; Yashaswi Shrestha; Jesse S. Boehm; Supriya K Jain; Emeric Bojarski; Craig H. Mermel; Jordi Barretina; Jennifer A. Chan; José Baselga; Josep Tabernero; David E. Root; Charles S. Fuchs; Massimo Loda; Ramesh A. Shivdasani; Matthew Meyerson; William C. Hahn

Aberrant activation of the canonical WNT/β-catenin pathway occurs in almost all colorectal cancers and contributes to their growth, invasion and survival. Although dysregulated β-catenin activity drives colon tumorigenesis, further genetic perturbations are required to elaborate full malignant transformation. To identify genes that both modulate β-catenin activity and are essential for colon cancer cell proliferation, we conducted two loss-of-function screens in human colon cancer cells and compared genes identified in these screens with an analysis of copy number alterations in colon cancer specimens. One of these genes, CDK8, which encodes a member of the mediator complex, is located at 13q12.13, a region of recurrent copy number gain in a substantial fraction of colon cancers. Here we show that the suppression of CDK8 expression inhibits proliferation in colon cancer cells characterized by high levels of CDK8 and β-catenin hyperactivity. CDK8 kinase activity was necessary for β-catenin-driven transformation and for expression of several β-catenin transcriptional targets. Together these observations suggest that therapeutic interventions targeting CDK8 may confer a clinical benefit in β-catenin-driven malignancies.


Cancer Cell | 2008

Acquisition of Granule Neuron Precursor Identity Is a Critical Determinant of Progenitor Cell Competence to Form Shh-Induced Medulloblastoma

Ulrich Schüller; Vivi M. Heine; Junhao Mao; Alvin T. Kho; Allison K. Dillon; Young-Goo Han; Emmanuelle Huillard; Tao Sun; Azra H. Ligon; Ying Qian; Qiufu Ma; Arturo Alvarez-Buylla; Andrew P. McMahon; David H. Rowitch; Keith L. Ligon

Whether the brain tumor medulloblastoma originates from stem cells or restricted progenitor cells is unclear. To investigate this, we activated oncogenic Hedgehog (Hh) signaling in multipotent and lineage-restricted central nervous system (CNS) progenitors. We observed that normal unipotent cerebellar granule neuron precursors (CGNPs) derive from hGFAP(+) and Olig2(+) rhombic lip progenitors. Hh activation in a spectrum of early- and late-stage CNS progenitors generated similar medulloblastomas, but not other brain cancers, indicating that acquisition of CGNP identity is essential for tumorigenesis. We show in human and mouse medulloblastoma that cells expressing the glia-associated markers Gfap and Olig2 are neoplastic and retain features of embryonic-type granule lineage progenitors. Thus, oncogenic Hh signaling promotes medulloblastoma from lineage-restricted granule cell progenitors.


Nature | 2001

Integration of cytogenetic landmarks into the draft sequence of the human genome

Vivian G. Cheung; N. Nowak; W. Jang; Irving Kirsch; S. Zhao; X. N. Chen; Terrence S. Furey; U. J. Kim; W. L. Kuo; M. Olvier; J. Conroy; A. Kasprzyk; Hillary Massa; R. Yonescu; S.S.J. Sait; C. Thoreen; A. Snijders; E. Lemyre; J. A. Bailey; A. Bruzel; W. D. Burrill; S. M. Clegg; Steven J. Collins; P. Dhami; Cynthia Friedman; C. S. Han; S. Herrick; Joyce Lee; Azra H. Ligon; S. Lowry

We have placed 7,600 cytogenetically defined landmarks on the draft sequence of the human genome to help with the characterization of genes altered by gross chromosomal aberrations that cause human disease. The landmarks are large-insert clones mapped to chromosome bands by fluorescence in situ hybridization. Each clone contains a sequence tag that is positioned on the genomic sequence. This genome-wide set of sequence-anchored clones allows structural and functional analyses of the genome. This resource represents the first comprehensive integration of cytogenetic, radiation hybrid, linkage and sequence maps of the human genome; provides an independent validation of the sequence map and framework for contig order and orientation; surveys the genome for large-scale duplications, which are likely to require special attention during sequence assembly; and allows a stringent assessment of sequence differences between the dark and light bands of chromosomes. It also provides insight into large-scale chromatin structure and the evolution of chromosomes and gene families and will accelerate our understanding of the molecular bases of human disease and cancer.


Lancet Oncology | 2016

Nivolumab for classical Hodgkin's lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial

Anas Younes; Armando Santoro; Margaret A. Shipp; Pier Luigi Zinzani; John M. Timmerman; Stephen M. Ansell; Philippe Armand; Michelle A. Fanale; Voravit Ratanatharathorn; John Kuruvilla; Jonathon B. Cohen; Graham P. Collins; Kerry J. Savage; Marek Trneny; Kazunobu Kato; Benedetto Farsaci; Susan M. Parker; Scott J. Rodig; Margaretha G. M. Roemer; Azra H. Ligon; Andreas Engert

Background Malignant cells of classical Hodgkin lymphoma (cHL) are characterised by genetic alterations at the 9p24·1 locus. This leads to overexpression of the programmed death 1 (PD-1) ligands and enables tumour cells to evade immune surveillance. A phase 1b study showed that nivolumab, a PD-1-blocking antibody, produced a high response rate in patients with relapsed and refractory cHL, with an acceptable safety profile. This phase 2 study assessed the clinical benefit of nivolumab monotherapy in patients with cHL after autologous stem-cell transplantation and brentuximab vedotin failure. Methods This ongoing phase 2 study (NCT02181738) assessed the efficacy and safety of nivolumab, administered intravenously over 60 minutes at 3 mg/kg every 2 weeks, in adult patients with cHL who had failed both autologous stem-cell transplantation and brentuximab vedotin. The primary endpoint was objective response rate by independent radiologic review committee (IRRC) assessment. Secondary and other endpoints included duration of response, safety, and assessment of PD-L1 and PD-L2 loci and PD-L1 and PD-L2 protein expression. Findings Among 80 treated patients, the median number of prior therapies was four (range 3–15). With a mean (SD) follow-up of 8·6 months (2·02), objective response rate per IRRC was 66·3% (53/80). The most common drug-related adverse events (≥15%) included fatigue, infusion-related reaction, and rash. The most common drug-related grade 3–4 adverse events were neutropenia and increased lipase levels (both n=4). The most common serious adverse event (any grade) was pyrexia (n=3). Interpretation Nivolumab demonstrated a high response rate and an acceptable safety profile in patients with cHL who progressed following autologous stem-cell transplantation and brentuximab vedotin. Nivolumab may therefore provide a novel treatment option for a patient population with a high unmet need. Ongoing follow-up will help to assess the durability of response. Funding Bristol-Myers Squibb.BACKGROUND Malignant cells of classical Hodgkins lymphoma are characterised by genetic alterations at the 9p24.1 locus, leading to overexpression of PD-1 ligands and evasion of immune surveillance. In a phase 1b study, nivolumab, a PD-1-blocking antibody, produced a high response in patients with relapsed and refractory classical Hodgkins lymphoma, with an acceptable safety profile. We aimed to assess the clinical benefit and safety of nivolumab monotherapy in patients with classical Hodgkins lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin. METHODS In this ongoing, single-arm phase 2 study, adult patients (aged ≥18 years) with recurrent classical Hodgkins lymphoma who had failed to respond to autologous stem-cell transplantation and had either relapsed after or failed to respond to brentuximab vedotin, and with an Eastern Cooperative Oncology Group performance status score of 0 or 1, were enrolled from 34 hospitals and academic centres across Europe and North America. Patients were given nivolumab intravenously over 60 min at 3 mg/kg every 2 weeks until progression, death, unacceptable toxicity, or withdrawal from study. The primary endpoint was objective response following a prespecified minimum follow-up period of 6 months, assessed by an independent radiological review committee (IRRC). All patients who received at least one dose of nivolumab were included in the primary and safety analyses. This trial is registered with ClinicalTrials.gov, number NCT02181738. FINDINGS Among 80 treated patients recruited between Aug 26, 2014, and Feb 20, 2015, the median number of previous therapies was four (IQR 4-7). At a median follow-up of 8·9 months (IQR 7·8-9·9), 53 (66·3%, 95% CI 54·8-76·4) of 80 patients achieved an IRRC-assessed objective response. The most common drug-related adverse events (those that occurred in ≥15% of patients) included fatigue (20 [25%] patients), infusion-related reaction (16 [20%]), and rash (13 [16%]). The most common drug-related grade 3 or 4 adverse events were neutropenia (four [5%] patients) and increased lipase concentrations (four [5%]). The most common serious adverse event (any grade) was pyrexia (three [4%] patients). Three patients died during the study; none of these deaths were judged to be treatment related. INTERPRETATION Nivolumab resulted in frequent responses with an acceptable safety profile in patients with classical Hodgkins lymphoma who progressed after autologous stem-cell transplantation and brentuximab vedotin. Therefore, nivolumab might be a new treatment option for a patient population with a high unmet need. Ongoing follow-up will help to assess the durability of response. FUNDING Bristol-Myers Squibb.

Collaboration


Dive into the Azra H. Ligon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sandro Santagata

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Ian F. Dunn

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Neal I. Lindeman

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian M. Alexander

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Wenya Linda Bi

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge