Network


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

Hotspot


Dive into the research topics where Timothy P. Heffernan is active.

Publication


Featured researches published by Timothy P. Heffernan.


Cell | 2011

BET Bromodomain Inhibition as a Therapeutic Strategy to Target c-Myc

Jake Delmore; Ghayas C Issa; Madeleine E. Lemieux; Peter B. Rahl; Junwei Shi; Hannah M. Jacobs; Efstathios Kastritis; Timothy Gilpatrick; Ronald M. Paranal; Jun Qi; Marta Chesi; Anna C. Schinzel; Michael R. McKeown; Timothy P. Heffernan; Christopher R. Vakoc; P. Leif Bergsagel; Irene M. Ghobrial; Paul G. Richardson; Richard A. Young; William C. Hahn; Kenneth C. Anderson; Andrew L. Kung; James E. Bradner; Constantine S. Mitsiades

MYC contributes to the pathogenesis of a majority of human cancers, yet strategies to modulate the function of the c-Myc oncoprotein do not exist. Toward this objective, we have targeted MYC transcription by interfering with chromatin-dependent signal transduction to RNA polymerase, specifically by inhibiting the acetyl-lysine recognition domains (bromodomains) of putative coactivator proteins implicated in transcriptional initiation and elongation. Using a selective small-molecule bromodomain inhibitor, JQ1, we identify BET bromodomain proteins as regulatory factors for c-Myc. BET inhibition by JQ1 downregulates MYC transcription, followed by genome-wide downregulation of Myc-dependent target genes. In experimental models of multiple myeloma, a Myc-dependent hematologic malignancy, JQ1 produces a potent antiproliferative effect associated with cell-cycle arrest and cellular senescence. Efficacy of JQ1 in three murine models of multiple myeloma establishes the therapeutic rationale for BET bromodomain inhibition in this disease and other malignancies characterized by pathologic activation of c-Myc.


Nature | 2011

Telomere dysfunction induces metabolic and mitochondrial compromise

Ergiin Sahin; Simona Colla; Marc Liesa; Javid Moslehi; Florian Muller; Mira Guo; Marcus P. Cooper; Darrell N. Kotton; Attila J. Fabian; Carl Walkey; Richard S. Maser; Giovanni Tonon; Friedrich Foerster; Robert Xiong; Y. Alan Wang; Sachet A. Shukla; Mariela Jaskelioff; Eric Martin; Timothy P. Heffernan; Alexei Protopopov; Elena Ivanova; John E. Mahoney; Maria Kost-Alimova; Samuel R. Perry; Roderick T. Bronson; Ronglih Liao; Richard C. Mulligan; Orian S. Shirihai; Lynda Chin; Ronald A. DePinho

Telomere dysfunction activates p53-mediated cellular growth arrest, senescence and apoptosis to drive progressive atrophy and functional decline in high-turnover tissues. The broader adverse impact of telomere dysfunction across many tissues including more quiescent systems prompted transcriptomic network analyses to identify common mechanisms operative in haematopoietic stem cells, heart and liver. These unbiased studies revealed profound repression of peroxisome proliferator-activated receptor gamma, coactivator 1 alpha and beta (PGC-1α and PGC-1β, also known as Ppargc1a and Ppargc1b, respectively) and the downstream network in mice null for either telomerase reverse transcriptase (Tert) or telomerase RNA component (Terc) genes. Consistent with PGCs as master regulators of mitochondrial physiology and metabolism, telomere dysfunction is associated with impaired mitochondrial biogenesis and function, decreased gluconeogenesis, cardiomyopathy, and increased reactive oxygen species. In the setting of telomere dysfunction, enforced Tert or PGC-1α expression or germline deletion of p53 (also known as Trp53) substantially restores PGC network expression, mitochondrial respiration, cardiac function and gluconeogenesis. We demonstrate that telomere dysfunction activates p53 which in turn binds and represses PGC-1α and PGC-1β promoters, thereby forging a direct link between telomere and mitochondrial biology. We propose that this telomere–p53–PGC axis contributes to organ and metabolic failure and to diminishing organismal fitness in the setting of telomere dysfunction.


Nature | 2012

Melanoma genome sequencing reveals frequent PREX2 mutations

Michael F. Berger; Eran Hodis; Timothy P. Heffernan; Yonathan Lissanu Deribe; Michael S. Lawrence; Alexei Protopopov; Elena S Ivanova; Ian R. Watson; Elizabeth Nickerson; Papia Ghosh; Hailei Zhang; Rhamy Zeid; Xiaojia Ren; Kristian Cibulskis; Andrey Sivachenko; Nikhil Wagle; Antje Sucker; Carrie Sougnez; Robert C. Onofrio; Lauren Ambrogio; Daniel Auclair; Timothy Fennell; Scott L. Carter; Yotam Drier; Petar Stojanov; Meredith A. Singer; Douglas Voet; Rui Jing; Gordon Saksena; Jordi Barretina

Melanoma is notable for its metastatic propensity, lethality in the advanced setting and association with ultraviolet exposure early in life. To obtain a comprehensive genomic view of melanoma in humans, we sequenced the genomes of 25 metastatic melanomas and matched germline DNA. A wide range of point mutation rates was observed: lowest in melanomas whose primaries arose on non-ultraviolet-exposed hairless skin of the extremities (3 and 14 per megabase (Mb) of genome), intermediate in those originating from hair-bearing skin of the trunk (5–55 per Mb), and highest in a patient with a documented history of chronic sun exposure (111 per Mb). Analysis of whole-genome sequence data identified PREX2 (phosphatidylinositol-3,4,5-trisphosphate-dependent Rac exchange factor 2)—a PTEN-interacting protein and negative regulator of PTEN in breast cancer—as a significantly mutated gene with a mutation frequency of approximately 14% in an independent extension cohort of 107 human melanomas. PREX2 mutations are biologically relevant, as ectopic expression of mutant PREX2 accelerated tumour formation of immortalized human melanocytes in vivo. Thus, whole-genome sequencing of human melanoma tumours revealed genomic evidence of ultraviolet pathogenesis and discovered a new recurrently mutated gene in melanoma.


Nature | 2014

Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function

Andrea Viale; Piergiorgio Pettazzoni; Costas A. Lyssiotis; Haoqiang Ying; Nora Sanchez; Matteo Marchesini; Alessandro Carugo; Tessa Green; Sahil Seth; Virginia Giuliani; Maria Kost-Alimova; Florian Muller; Simona Colla; Luigi Nezi; Giannicola Genovese; Angela K. Deem; Avnish Kapoor; Wantong Yao; Emanuela Brunetto; Ya’an Kang; Min Yuan; John M. Asara; Y. Alan Wang; Timothy P. Heffernan; Alec C. Kimmelman; Huamin Wang; Jason B. Fleming; Lewis C. Cantley; Ronald A. DePinho; Giulio Draetta

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in western countries, with a median survival of 6 months and an extremely low percentage of long-term surviving patients. KRAS mutations are known to be a driver event of PDAC, but targeting mutant KRAS has proved challenging. Targeting oncogene-driven signalling pathways is a clinically validated approach for several devastating diseases. Still, despite marked tumour shrinkage, the frequency of relapse indicates that a fraction of tumour cells survives shut down of oncogenic signalling. Here we explore the role of mutant KRAS in PDAC maintenance using a recently developed inducible mouse model of mutated Kras (KrasG12D, herein KRas) in a p53LoxP/WT background. We demonstrate that a subpopulation of dormant tumour cells surviving oncogene ablation (surviving cells) and responsible for tumour relapse has features of cancer stem cells and relies on oxidative phosphorylation for survival. Transcriptomic and metabolic analyses of surviving cells reveal prominent expression of genes governing mitochondrial function, autophagy and lysosome activity, as well as a strong reliance on mitochondrial respiration and a decreased dependence on glycolysis for cellular energetics. Accordingly, surviving cells show high sensitivity to oxidative phosphorylation inhibitors, which can inhibit tumour recurrence. Our integrated analyses illuminate a therapeutic strategy of combined targeting of the KRAS pathway and mitochondrial respiration to manage pancreatic cancer.


Cancer Discovery | 2016

Loss of PTEN promotes resistance to T cell–mediated immunotherapy

Weiyi Peng; Jie Qing Chen; Chengwen Liu; Shruti Malu; Caitlin Creasy; Michael T. Tetzlaff; Chunyu Xu; Jodi A. McKenzie; Chunlei Zhang; Xiaoxuan Liang; Leila Williams; Wanleng Deng; Guo Chen; Rina M. Mbofung; Alexander J. Lazar; Carlos A. Torres-Cabala; Zachary A. Cooper; Pei-Ling Chen; Trang Tieu; Stefani Spranger; Xiaoxing Yu; Chantale Bernatchez; Marie-Andree Forget; Cara Haymaker; Rodabe N. Amaria; Jennifer L. McQuade; Isabella C. Glitza; Tina Cascone; Haiyan S. Li; Lawrence N. Kwong

UNLABELLED T cell-mediated immunotherapies are promising cancer treatments. However, most patients still fail to respond to these therapies. The molecular determinants of immune resistance are poorly understood. We show that loss of PTEN in tumor cells in preclinical models of melanoma inhibits T cell-mediated tumor killing and decreases T-cell trafficking into tumors. In patients, PTEN loss correlates with decreased T-cell infiltration at tumor sites, reduced likelihood of successful T-cell expansion from resected tumors, and inferior outcomes with PD-1 inhibitor therapy. PTEN loss in tumor cells increased the expression of immunosuppressive cytokines, resulting in decreased T-cell infiltration in tumors, and inhibited autophagy, which decreased T cell-mediated cell death. Treatment with a selective PI3Kβ inhibitor improved the efficacy of both anti-PD-1 and anti-CTLA-4 antibodies in murine models. Together, these findings demonstrate that PTEN loss promotes immune resistance and support the rationale to explore combinations of immunotherapies and PI3K-AKT pathway inhibitors. SIGNIFICANCE This study adds to the growing evidence that oncogenic pathways in tumors can promote resistance to the antitumor immune response. As PTEN loss and PI3K-AKT pathway activation occur in multiple tumor types, the results support the rationale to further evaluate combinatorial strategies targeting the PI3K-AKT pathway to increase the efficacy of immunotherapy.


Molecular and Cellular Biology | 2002

An ATR- and Chk1-Dependent S Checkpoint Inhibits Replicon Initiation following UVC-Induced DNA Damage

Timothy P. Heffernan; Dennis A. Simpson; Alexandra R. Frank; Alexandra N. Heinloth; Richard S. Paules; Marila Cordeiro-Stone; William K. Kaufmann

ABSTRACT Inhibition of replicon initiation is a stereotypic DNA damage response mediated through S checkpoint mechanisms not yet fully understood. Studies were undertaken to elucidate the function of checkpoint proteins in the inhibition of replicon initiation following irradiation with 254 nm UV light (UVC) of diploid human fibroblasts immortalized by the ectopic expression of telomerase. Velocity sedimentation analysis of nascent DNA molecules revealed a 50% inhibition of replicon initiation when normal human fibroblasts were treated with a low dose of UVC (1 J/m2). Ataxia telangiectasia (AT), Nijmegen breakage syndrome (NBS), and AT-like disorder fibroblasts, which lack an S checkpoint response when exposed to ionizing radiation, responded normally when exposed to UVC and inhibited replicon initiation. Pretreatment of normal and AT fibroblasts with caffeine or UCN-01, inhibitors of ATR (AT mutated and Rad3 related) and Chk1, respectively, abolished the S checkpoint response to UVC. Moreover, overexpression of kinase-inactive ATR in U2OS cells severely attenuated UVC-induced Chk1 phosphorylation and reversed the UVC-induced inhibition of replicon initiation, as did overexpression of kinase-inactive Chk1. Taken together, these data suggest that the UVC-induced S checkpoint response of inhibition of replicon initiation is mediated by ATR signaling through Chk-1 and is independent of ATM, Nbs1, and Mre11.


Cell | 2014

Yap1 activation enables bypass of oncogenic KRAS addiction in pancreatic cancer

Avnish Kapoor; Wantong Yao; Haoqiang Ying; Sujun Hua; Alison Liewen; Qiuyun Wang; Yi Zhong; Chang Jiun Wu; Anguraj Sadanandam; Baoli Hu; Qing Chang; Gerald C. Chu; Ramsey Al-Khalil; Shan Jiang; Hongai Xia; Eliot Fletcher-Sananikone; Carol Lim; Gillian I. Horwitz; Andrea Viale; Piergiorgio Pettazzoni; Nora Sanchez; Huamin Wang; Alexei Protopopov; Jianhua Zhang; Timothy P. Heffernan; Randy L. Johnson; Lynda Chin; Y. Alan Wang; Giulio Draetta; Ronald A. DePinho

Activating mutations in KRAS are among the most frequent events in diverse human carcinomas and are particularly prominent in human pancreatic ductal adenocarcinoma (PDAC). An inducible Kras(G12D)-driven mouse model of PDAC has established a critical role for sustained Kras(G12D) expression in tumor maintenance, providing a model to determine the potential for and the underlying mechanisms of Kras(G12D)-independent PDAC recurrence. Here, we show that some tumors undergo spontaneous relapse and are devoid of Kras(G12D) expression and downstream canonical MAPK signaling and instead acquire amplification and overexpression of the transcriptional coactivator Yap1. Functional studies established the role of Yap1 and the transcriptional factor Tead2 in driving Kras(G12D)-independent tumor maintenance. The Yap1/Tead2 complex acts cooperatively with E2F transcription factors to activate a cell cycle and DNA replication program. Our studies, along with corroborating evidence from human PDAC models, portend a novel mechanism of escape from oncogenic Kras addiction in PDAC.


Cancer Discovery | 2015

Co-occurring genomic alterations define major subsets of KRAS - mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities

Ferdinandos Skoulidis; Lauren Averett Byers; Lixia Diao; Vassiliki Papadimitrakopoulou; Pan Tong; Julie Izzo; Carmen Behrens; Humam Kadara; Edwin R. Parra; Jaime Rodriguez Canales; Jianjun Zhang; Uma Giri; Jayanthi Gudikote; Maria Angelica Cortez; Chao Yang; You Hong Fan; Michael Peyton; Luc Girard; Kevin R. Coombes; Carlo Toniatti; Timothy P. Heffernan; Murim Choi; Garrett Michael Frampton; Vincent A. Miller; John N. Weinstein; Roy S. Herbst; Kwok-Kin Wong; Jianhua Zhang; Padmanee Sharma; Gordon B. Mills

UNLABELLED The molecular underpinnings that drive the heterogeneity of KRAS-mutant lung adenocarcinoma are poorly characterized. We performed an integrative analysis of genomic, transcriptomic, and proteomic data from early-stage and chemorefractory lung adenocarcinoma and identified three robust subsets of KRAS-mutant lung adenocarcinoma dominated, respectively, by co-occurring genetic events in STK11/LKB1 (the KL subgroup), TP53 (KP), and CDKN2A/B inactivation coupled with low expression of the NKX2-1 (TTF1) transcription factor (KC). We further revealed biologically and therapeutically relevant differences between the subgroups. KC tumors frequently exhibited mucinous histology and suppressed mTORC1 signaling. KL tumors had high rates of KEAP1 mutational inactivation and expressed lower levels of immune markers, including PD-L1. KP tumors demonstrated higher levels of somatic mutations, inflammatory markers, immune checkpoint effector molecules, and improved relapse-free survival. Differences in drug sensitivity patterns were also observed; notably, KL cells showed increased vulnerability to HSP90-inhibitor therapy. This work provides evidence that co-occurring genomic alterations identify subgroups of KRAS-mutant lung adenocarcinoma with distinct biology and therapeutic vulnerabilities. SIGNIFICANCE Co-occurring genetic alterations in STK11/LKB1, TP53, and CDKN2A/B-the latter coupled with low TTF1 expression-define three major subgroups of KRAS-mutant lung adenocarcinoma with distinct biology, patterns of immune-system engagement, and therapeutic vulnerabilities.


Cancer Cell | 2008

Feedback circuit among INK4 tumor suppressors constrains human glioblastoma development.

Ruprecht Wiedemeyer; Cameron Brennan; Timothy P. Heffernan; Yonghong Xiao; John T. Mahoney; Alexei Protopopov; Hongwu Zheng; Graham R. Bignell; Frank Furnari; Webster K. Cavenee; William C. Hahn; Koichi Ichimura; Peter Collins; Gerald C. Chu; Michael R. Stratton; Keith L. Ligon; Andrew Futreal; Lynda Chin

Summary We have developed a nonheuristic genome topography scan (GTS) algorithm to characterize the patterns of genomic alterations in human glioblastoma (GBM), identifying frequent p18INK4C and p16INK4A codeletion. Functional reconstitution of p18INK4C in GBM cells null for both p16INK4A and p18INK4C resulted in impaired cell-cycle progression and tumorigenic potential. Conversely, RNAi-mediated depletion of p18INK4C in p16INK4A-deficient primary astrocytes or established GBM cells enhanced tumorigenicity in vitro and in vivo. Furthermore, acute suppression of p16INK4A in primary astrocytes induced a concomitant increase in p18INK4C. Together, these findings uncover a feedback regulatory circuit in the astrocytic lineage and demonstrate a bona fide tumor suppressor role for p18INK4C in human GBM wherein it functions cooperatively with other INK4 family members to constrain inappropriate proliferation.


Cancer Research | 2015

The SMARCA2/4 ATPase domain surpasses the bromodomain as a drug target in SWI/SNF mutant cancers: Insights from cDNA rescue and PFI-3 inhibitor studies

Bhavatarini Vangamudi; Thomas A. Paul; Parantu K. Shah; Maria Kost-Alimova; Lisa Nottebaum; Xi Shi; Yanai Zhan; Elisabetta Leo; Harshad S. Mahadeshwar; Alexei Protopopov; Andrew Futreal; Trang Tieu; Mike Peoples; Timothy P. Heffernan; Joseph R. Marszalek; Carlo Toniatti; Alessia Petrocchi; Dominique Verhelle; Dafydd R. Owen; Giulio Draetta; Philip Jones; Wylie Solang Palmer; Shikhar Sharma; Jannik N. Andersen

The SWI/SNF multisubunit complex modulates chromatin structure through the activity of two mutually exclusive catalytic subunits, SMARCA2 and SMARCA4, which both contain a bromodomain and an ATPase domain. Using RNAi, cancer-specific vulnerabilities have been identified in SWI/SNF-mutant tumors, including SMARCA4-deficient lung cancer; however, the contribution of conserved, druggable protein domains to this anticancer phenotype is unknown. Here, we functionally deconstruct the SMARCA2/4 paralog dependence of cancer cells using bioinformatics, genetic, and pharmacologic tools. We evaluate a selective SMARCA2/4 bromodomain inhibitor (PFI-3) and characterize its activity in chromatin-binding and cell-functional assays focusing on cells with altered SWI/SNF complex (e.g., lung, synovial sarcoma, leukemia, and rhabdoid tumors). We demonstrate that PFI-3 is a potent, cell-permeable probe capable of displacing ectopically expressed, GFP-tagged SMARCA2-bromodomain from chromatin, yet contrary to target knockdown, the inhibitor fails to display an antiproliferative phenotype. Mechanistically, the lack of pharmacologic efficacy is reconciled by the failure of bromodomain inhibition to displace endogenous, full-length SMARCA2 from chromatin as determined by in situ cell extraction, chromatin immunoprecipitation, and target gene expression studies. Furthermore, using inducible RNAi and cDNA complementation (bromodomain- and ATPase-dead constructs), we unequivocally identify the ATPase domain, and not the bromodomain of SMARCA2, as the relevant therapeutic target with the catalytic activity suppressing defined transcriptional programs. Taken together, our complementary genetic and pharmacologic studies exemplify a general strategy for multidomain protein drug-target validation and in case of SMARCA2/4 highlight the potential for drugging the more challenging helicase/ATPase domain to deliver on the promise of synthetic-lethality therapy.

Collaboration


Dive into the Timothy P. Heffernan's collaboration.

Top Co-Authors

Avatar

Giulio Draetta

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Alessandro Carugo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher A. Bristow

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sahil Seth

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Joseph R. Marszalek

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lynda Chin

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Carlo Toniatti

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Piergiorgio Pettazzoni

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ronald A. DePinho

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Andrea Viale

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge