Network


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

Hotspot


Dive into the research topics where Jane B. Trepel is active.

Publication


Featured researches published by Jane B. Trepel.


The FASEB Journal | 2003

IL-1β-mediated up-regulation of HIF-1α via an NFκB/COX-2 pathway identifies HIF-1 as a critical link between inflammation and oncogenesis

Yunjin Jung; Jennifer S. Isaacs; Sunmin Lee; Jane B. Trepel; Len Neckers

Growing evidence indicates that inflammation is a contributing factor leading to cancer development. However, pathways involved in this progression are not well understood. To examine whether HIF‐1α is a factor linking inflammation and tumorigenesis, we investigated whether the HIF‐1 signaling pathway was stimulated by the pro‐inflammatory cytokine interleukin‐1β (IL‐1β) in A549 cells. We find that IL‐1β up‐regulated HIF‐1α protein under normoxia and activated the HIF‐1‐responsive gene vascular endothelial growth factor (VEGF) via a pathway dependent on nuclear factor κB (NFkB). Interestingly, although this pathway is stimulated by upstream signaling via AKT and mTOR and requires new transcription, IL‐1 mediated HIF‐1α induction also utilizes a post‐transcriptional mechanism that involves antagonism of VHL‐dependent HIF‐1α degradation, which results in increased HIF‐1α protein stability. IL‐1 mediated NFkB‐dependent cyclooxygenases‐2 (COX‐2) expression served as a positive effector for HIF‐1α induction. Although COX‐2 inhibitors attenuated IL‐1 mediated HIF‐1α induction, prostaglandin E2 (PGE2), a physiological product of COX‐2, induced HIF‐1α protein in a dose‐dependent manner. Our data, therefore, demonstrate that IL‐1β up‐regulates functional HIF‐1α protein through a classical inflammatory signaling pathway involving NFkB and COX‐2, culminating in up‐regulation of VEGF, a potent angiogenic factor required for tumor growth and metastasis. Thus, HIF‐1 is identified as a pivotal transcription factor linking the inflammatory and oncogenic pathways.


Journal of Clinical Oncology | 2005

Phase I and Pharmacokinetic Study of MS-275, a Histone Deacetylase Inhibitor, in Patients With Advanced and Refractory Solid Tumors or Lymphoma

Qin C. Ryan; Donna Headlee; Milin R. Acharya; Alex Sparreboom; Jane B. Trepel; Joseph Z. Ye; William D. Figg; Kyunghwa Hwang; Eun Joo Chung; Anthony J. Murgo; Giovanni Melillo; Yusri A. Elsayed; Manish Monga; Mikhail Kalnitskiy; James A. Zwiebel; Edward A. Sausville

PURPOSE The objective of this study was to define the maximum-tolerated dose (MTD), the recommended phase II dose, the dose-limiting toxicity, and determine the pharmacokinetic (PK) and pharmacodynamic profiles of MS-275. PATIENTS AND METHODS Patients with advanced solid tumors or lymphoma were treated with MS-275 orally initially on a once daily x 28 every 6 weeks (daily) and later on once every-14-days (q14-day) schedules. The starting dose was 2 mg/m2 and the dose was escalated in three- to six-patient cohorts based on toxicity assessments. RESULTS With the daily schedule, the MTD was exceeded at the first dose level. Preliminary PK analysis suggested the half-life of MS-275 in humans was 39 to 80 hours, substantially longer than predicted by preclinical studies. With the q14-day schedule, 28 patients were treated. The MTD was 10 mg/m2 and dose-limiting toxicities were nausea, vomiting, anorexia, and fatigue. Exposure to MS-275 was dose dependent, suggesting linear PK. Increased histone H3 acetylation in peripheral-blood mononuclear-cells was apparent at all dose levels by immunofluorescence analysis. Ten of 29 patients remained on treatment for > or = 3 months. CONCLUSION The MS-275 oral formulation on the daily schedule was intolerable at a dose and schedule explored. The q14-day schedule is reasonably well tolerated. Histone deacetylase inhibition was observed in peripheral-blood mononuclear-cells. Based on PK data from the q14-day schedule, a more frequent dosing schedule, weekly x 4, repeated every 6 weeks is presently being evaluated.


Journal of Biological Chemistry | 1998

p53 inhibits hypoxia-inducible factor-stimulated transcription.

Mikhail V. Blagosklonny; Won G. An; Larisa Y. Romanova; Jane B. Trepel; Tito Fojo; Len Neckers

p53 is required for hypoxia-induced apoptosisin vivo, although the mechanism by which this occurs is not known. Conversely, induction of the hypoxia-inducible factor-1 (HIF-1) transactivator stimulates transcription of a number of genes crucial to survival of the hypoxic state. Here we demonstrate that p53 represses HIF-1-stimulated transcription. Although higher levels of p53 are required to inhibit HIF than are necessary to transcriptionally activate p53 target genes, these levels of p53 are similar to those that stimulate cleavage of poly(ADP-ribose) polymerase, an early event in apoptosis. Transfection of full-length p300 stimulates both p53-dependent and HIF-dependent transcription but does not relieve p53-mediated inhibition of HIF. In contrast, a p300 fragment, which binds to p53 but not to HIF-1, prevents p53-dependent repression of HIF activity. Transcriptionally inactive p53, mutated in its DNA binding domain, retains the ability to block HIF transactivating activity, whereas a transcriptionally inactive double point mutant defective for p300 binding does not inhibit HIF. Finally, depletion of doxorubicin-induced endogenous p53 by E6 protein attenuates doxorubicin-stimulated inhibition of HIF, suggesting that a p53 level sufficient for HIF inhibition can be achieved in vivo. These data support a model in which stoichiometric binding of p53 to a HIF/p300 transcriptional complex mediates inhibition of HIF activity.


Journal of Biological Chemistry | 1996

P185ERBB2 BINDS TO GRP94 IN VIVO : DISSOCIATION OF THE P185ERBB2/GRP94 HETEROCOMPLEX BY BENZOQUINONE ANSAMYCINS PRECEDES DEPLETION OF P185ERBB2

Christine Chavany; Edward G. Mimnaugh; Penny Miller; Roberto Bitton; Phongmai Nguyen; Jane B. Trepel; Luke Whitesell; Rodney C. Schnur; James D. Moyer; Len Neckers

Treatment of SKBr3 cells with benzoquinone ansamycins, such as geldanamycin (GA), depletes p185, the receptor tyrosine kinase encoded by the erbB2 gene. In the same cells, a biologically active benzoquinone photoaffinity label specifically binds a protein of about 100 kDa, and the ability of various GA derivatives to reduce the intracellular level of p185 correlates with their ability to compete with the photoaffinity label for binding to this protein. In this report, we present evidence that the 100-kDa ansamycin-binding protein is GRP94. Membrane-associated p185 exists in a stable complex with GRP94. GA binding to GRP94 disrupts this complex, leading to degradation of pre-existing p185 protein, and resulting in an altered subcellular distribution of newly synthesized p185.


Molecular and Cellular Biology | 1996

Destabilization of Raf-1 by Geldanamycin Leads to Disruption of the Raf-1-MEK-Mitogen-Activated Protein Kinase Signalling Pathway

T W Schulte; M V Blagosklonny; L Romanova; J F Mushinski; B P Monia; J F Johnston; Phuongmai Nguyen; Jane B. Trepel; Leonard M. Neckers

The serine/threonine kinase Raf-1 functions downstream of Rats in a signal transduction cascade which transmits mitogenic stimuli from the plasma membrane to the nucleus. Raf-1 integrates signals coming from extracellular factors and, in turn, activates its substrate, MEK kinase. MEK activates mitogen-activated protein kinase (MAPK), which phosphorylates other kinases as well as transcription factors. Raf-1 exists in a complex with HSP90 and other proteins. The benzoquinone ansamycin geldanamycin (GA) binds to HSP90 and disrupts the Raf-1-HSP90 multimolecular complex, leading to destabilization of Raf-1. In this study, we examined whether Raf-1 destabilization is sufficient to block the Raf-1-MEK-MAPK signalling pathway and whether GA specifically inactivates the Raf-1 component of this pathway. Using the model system of NIH 3T3 cells stimulated with phorbol 12-myristate 13-acetate (PMA), we show that GA does not affect the ability of protein kinase C alpha to be activated by phorbol esters, but it does block activation of MEK and MAPK. Further, GA does not decrease the activity of constitutively active MEK in transiently transfected cells. Finally, disruption of the Raf-1-MEK-MAPK signalling pathway by GA prevents both the PMA-induced proliferative response and PMA-induced activation of a MAPK-sensitive nuclear transcription factor. Thus, we demonstrate that interaction between HSP90 and Raf-1 is a sine qua non for Raf stability and function as a signal transducer and that the effects observed cannot be attributed to a general impairment of protein kinase function.


Annals of Internal Medicine | 1988

Histopathologic staging at initial diagnosis of mycosis fungoides and the Sézary syndrome. Definition of three distinctive prognostic groups.

Edward A. Sausville; Joyce L. Eddy; Robert W. Makuch; A B Fischmann; Geraldine P. Schechter; Mary J. Matthews; Eli Glatstein; Daniel C. Ihde; Frederic J. Kaye; Stephen R. Veach; Ruby Phelps; Terry O'Connor; Jane B. Trepel; James D. Cotelingam; Adi F. Gazdar; John D. Minna; Paul A. Bunn

STUDY OBJECTIVE To determine the optimal staging evaluation at the time of initial diagnosis of mycosis fungoides or the Sézary syndrome. DESIGN Retrospective review of a uniformly staged inception cohort. SETTING Single-institution tertiary care center. PATIENTS 152 consecutive patients who had mycosis fungoides with or without the Sézary syndrome within 6 months of the initial definitive diagnosis. INTERVENTION A detailed staging evaluation including physical examination, routine laboratory studies, chest roentgenogram, lymphangiogram, peripheral blood smear, lymph node biopsy, bone marrow aspirate or biopsy, and liver biopsy in selected patients. MEASUREMENTS AND MAIN RESULTS Univariate adverse prognostic features at initial diagnosis in patients with mycosis fungoides included (P less than 0.01) one or more cutaneous tumors or generalized erythroderma, adenopathy, blood smear involvement with Sézary cells, lymph node effacement, eosinophilia, and visceral involvement. Important, independent prognostic factors in a multivariate analysis are the presence of visceral disease and type of skin involvement. CONCLUSIONS A staging system based on histopathologic evaluation of skin, lymph nodes, blood, and visceral sites provides more comprehensive prognostic information than clinical evaluation of skin disease and adenopathy. Patients may be divided at initial presentation into three prognostic groups: good-risk patients, who have plaque-only skin disease without lymph node, blood, or visceral involvement (median survival, greater than 12 years); intermediate-risk patients, who have cutaneous tumors, erythroderma, or plaque disease with node or blood involvement but no visceral disease or node effacement (median survival, 5 years); and poor-risk patients, who have visceral involvement or node effacement (median survival, 2.5 years).


Medicinal Chemistry | 2006

Curcumin is an inhibitor of p300 histone acetylatransferase.

Len Neckers; Jane B. Trepel; Sunmin Lee; Eun-Joo Chung; Min-Jung Lee; Yunjin Jung; Monica Marcu

Histone acetyltransferases (HATs), and p300/CBP in particular, have been implicated in cancer cell growth and survival, and as such, HATs represent novel, therapeutically relevant molecular targets for drug development. In this study, we demonstrate that the small molecule natural product curcumin, whose medicinal properties have long been recognized in India and Southeast Asia, is a selective HAT inhibitor. Furthermore the data indicate that alpha, beta unsaturated carbonyl groups in the curcumin side chain function as Michael reaction sites and that the Michael reaction acceptor functionality of curcumin is required for its HAT-inhibitory activity. In cells, curcumin promoted proteasome-dependent degradation of p300 and the closely related CBP protein without affecting the HATs PCAF or GCN5. In addition to inducing p300 degradation curcumin inhibited the acetyltransferase activity of purified p300 as assessed using either histone H3 or p53 as substrate. Radiolabeled curcumin formed a covalent association with p300, and tetrahydrocurcumin displayed no p300 inhibitory activity, consistent with a Michael reaction-dependent mechanism. Finally, curcumin was able to effectively block histone hyperacetylation in both PC3-M prostate cancer cells and peripheral blood lymphocytes induced by the histone deacetylase inhibitor MS-275. These data thus identify the medicinal natural product curcumin as a novel lead compound for development of possibly therapeutic, p300/CBP-specific HAT inhibitors.


Leukemia | 2000

Protease inhibitor-induced apoptosis: accumulation of wt p53, p21WAF1/CIP1, and induction of apoptosis are independent markers of proteasome inhibition.

An Wg; Hwang Sg; Jane B. Trepel; Mikhail V. Blagosklonny

inhibitors of proteases are currently emerging as a potential anti-cancer modality. nonselective protease inhibitors are cytotoxic to leukemia and cancer cell lines and we found that this cytotoxicity is correlated with their potency as inhibitors of the proteasome but not as inhibitors of calpain and cathepsin. highly selective inhibitors of the proteasome were more cytotoxic and fast-acting than less selective inhibitors (ps341>>alln>>allm). induction of wt p53 correlated with inhibition of the proteasome and antiproliferative effect in mcf-7, a breast cancer cell line, which was resistant to apoptosis caused by proteasome inhibitors. in contrast, inhibitors of the proteasome induced apoptosis in four leukemia cell lines lacking wt p53. the order of sensitivity of leukemia cells was: jurkat>hl60⩾u937>>K562. The highly selective proteasome inhibitor PS-341 induced cell death with an IC50 as low as 5 nM in apoptosis-prone leukemia cells. Cell death was preceded by p21WAF1/CIP1 accumulation, an alternative marker of proteasome inhibition, and by cleavage of PARP and Rb proteins and nuclear fragmentation. Inhibition of caspases abrogated PARP cleavage and nuclear fragmentation and delayed, but did not completely prevent cell death caused by PS-341. Reintroduction of wt p53 into p53-null PC3 prostate carcinoma cells did not increase their sensitivity to proteasome inhibitors. Likewise, comparison of parental and p21-deficient cells demonstrated that p21WAF1/CIP1 was dispensable for proteasome inhibitor-induced cytotoxicity. We conclude that accumulation of wt p53 and induction of apoptosis are independent markers of proteasome inhibition.


Journal of Clinical Oncology | 2013

Randomized Phase II, Double-Blind, Placebo-Controlled Study of Exemestane With or Without Entinostat in Postmenopausal Women With Locally Recurrent or Metastatic Estrogen Receptor-Positive Breast Cancer Progressing on Treatment With a Nonsteroidal Aromatase Inhibitor

Denise A. Yardley; Roohi Ismail-Khan; Bohuslav Melichar; Mikhail Lichinitser; Pamela N. Munster; Pamela Klein; Scott Cruickshank; Kathy D. Miller; Min J. Lee; Jane B. Trepel

PURPOSE Entinostat is an oral isoform selective histone deacetylase inhibitor that targets resistance to hormonal therapies in estrogen receptor-positive (ER+) breast cancer. This randomized, placebo-controlled, phase II study evaluated entinostat combined with the aromatase inhibitor exemestane versus exemestane alone. PATIENTS AND METHODS Postmenopausal women with ER+ advanced breast cancer progressing on a nonsteroidal aromatase inhibitor were randomly assigned to exemestane 25 mg daily plus entinostat 5 mg once per week (EE) or exemestane plus placebo (EP). The primary end point was progression-free survival (PFS). Blood was collected in a subset of patients for evaluation of protein lysine acetylation as a biomarker of entinostat activity. RESULTS One hundred thirty patients were randomly assigned (EE group, n = 64; EP group, n = 66). Based on intent-to-treat analysis, treatment with EE improved median PFS to 4.3 months versus 2.3 months with EP (hazard ratio [HR], 0.73; 95% CI, 0.50 to 1.07; one-sided P = .055; two-sided P = .11 [predefined significance level of .10, one-sided]). Median overall survival was an exploratory end point and improved to 28.1 months with EE versus 19.8 months with EP (HR, 0.59; 95% CI, 0.36 to 0.97; P = .036). Fatigue and neutropenia were the most frequent grade 3/4 toxicities. Treatment discontinuation because of adverse events was higher in the EE group versus the EP group (11% v 2%). Protein lysine hyperacetylation in the EE biomarker subset was associated with prolonged PFS. CONCLUSION Entinostat added to exemestane is generally well tolerated and demonstrated activity in patients with ER+ advanced breast cancer in this signal-finding phase II study. Acetylation changes may provide an opportunity to maximize clinical benefit with entinostat. Plans for a confirmatory study are underway.


Cancer Research | 2004

Enhanced Radiation-Induced Cell Killing and Prolongation of γH2AX Foci Expression by the Histone Deacetylase Inhibitor MS-275

Kevin Camphausen; William E. Burgan; Michael A. Cerra; Kelli A. Oswald; Jane B. Trepel; Min-Jung Lee; Philip J. Tofilon

Histone deacetylase (HDAC) inhibitors are undergoing clinical evaluation for cancer therapy. Because HDAC modulates chromatin structure and gene expression, parameters considered to influence radioresponse, we have investigated the effects of the HDAC inhibitor MS-275 on the radiosensitivity of two human tumor cell lines (DU145 prostate carcinoma and U251 glioma). Acetylation status of histones H3 and H4 was determined as a function of time after MS-275 addition to and removal from culture medium. Histone acetylation increased by 6 h after MS-275 addition, reaching a maximum between 24 and 48 h of exposure; providing fresh drug-free medium then resulted in a decrease in histone acetylation that began by 6 h and approached untreated levels by 16 h. Treatment of cells with MS-275 for 48 h followed by irradiation had little or no effect on radiation-induced cell death. However, exposure to MS-275 before and after irradiation resulted in an increase in radiosensitivity with dose enhancement factors of 1.9 and 1.3 for DU145 and U251 cells, respectively. This MS-275 treatment protocol did not result in a redistribution of the cells into a more radiosensitive phase of the cell cycle or in an increase in apoptosis. However, MS-275 did modify the time course of γH2AX expression in irradiated cells. Whereas there was no significant difference in radiation-induced γH2AX foci at 6 h, the number of cells expressing γH2AX foci was significantly greater in the MS-275-treated cells at 24 h after irradiation. These results indicate that MS-275 can enhance radiosensitivity and suggest that this effect may involve an inhibition of DNA repair.

Collaboration


Dive into the Jane B. Trepel's collaboration.

Top Co-Authors

Avatar

Min-Jung Lee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sunmin Lee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Len Neckers

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

William D. Figg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Seth M. Steinberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Leonard M. Neckers

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

James L. Gulley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Min Jung Lee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

William L. Dahut

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Phuongmai Nguyen

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge