National Cancer Institute U.S. National Institutes of Health | www.cancer.gov
SPORES Logo - Translational Research, Clinical, Pre-Clinical, Basic, Pre-Clinical SPOREs - Specialized Programs of Research Excellence
Home Organization of the OSB Organ-Specific SPORE Programs Information for the Public Patient Advocate Research Team Information for Applicants Address and Directions IntraSPORE Communications  

The University of Texas M.D. Anderson Cancer Center SPORE in Uterine Cancer


Karen H. Lu, M.D.
Associate Professor
Department of Gynecology Oncology
M.D. Anderson Cancer Center


Purpose and Intent

Endometrial cancer is the most common gynecologic malignancy and the fourth most common cancer in women. For 2004, there will be an estimated 40,320 women diagnosed with this disease and 7,090 deaths. Significantly, there has been no decrease in the incidence or mortality of this cancer over the last 15 years.

The goal of the Gynecological Cancer SPORE at the University of Texas – M.D. Anderson Cancer Center is to conduct innovative translational research for the prevention and treatment of uterine tumors. Tumors arising from the epithelial (endometrium) and smooth muscle (myometrium) compartments of the uterus are important, yet under-funded, causes of morbidity and mortality in the United States. Endometrial cancer is the most common gynecologic malignancy and the fourth most common cancer in women. For 2004, there will be an estimated 40,320 women diagnosed with this disease and 7,090 deaths. Significantly, there has been no decrease in the incidence or mortality of this cancer over the last 15 years.

The proposed Gynecological Cancer SPORE is a truly multidisciplinary program that includes clinicians and basic scientists with both oncologic and non-oncologic backgrounds. Such a multidisciplinary team is necessary to achieve a more thorough understanding of the pathogenesis, prevention, and treatment of these tumors.

Encompassed within this overall goal are the following goals of the program:

  • Decipher the fundamental molecular differences between Type 1 and Type 2 endometrial cancers.
  • Provide a panel of molecular markers that will be useful in endometrial cancer prognosis and in identifying patients at risk for developing the malignancy.
  • Dissect the molecular pathways involved in estrogen and progesterone mediated growth regulation of the uterine endometrium and smooth muscle.
  • Promote novel strategies in the chemoprevention of endometrial cancer.
  • Understand, at the molecular level, the complex mechanism of action of selective estrogen receptor modulators (SERMs) such as tamoxifen, raloxifene, and a new third generation SERM, Arzoxifene, in the epithelial and smooth muscle compartments of the uterus.

Project 1
ENDOMETRIAL CARCINOMA: EFFICACY AND MECHANISM OF ACTION OF THE SERM ARZOXIFENE

Principal Investigators: Thomas W. Burke, M.D.; Cheryl L. Walker, Ph.D.
Co-Investigators: Leslie Gold, Ph.D.; Charles Levenback, M.D.; Lois Ramondetta, M.D.

Background: Epidemiologic evidence has identified unopposed estrogen exposure as a risk factor associated with endometrial carcinogenesis. A significant proportion of endometrial adenocarcinomas express immunohistochemically detectable estrogen (ER) and progesterone (PR) receptors, and objective clinical responses to “antiestrogenic” therapy using progestational agents (Megace) are well documented. The availability of SERMs that exhibit tissue-specific agonist or antagonist profiles now permits a more targeted approach to interference with (ER) signaling. One of these SERMs, LY353381 (Arzoxifene) developed by Eli Lilly, has been shown to function as an antagonist in the uterus in preclinical studies, although the molecular mechanisms response for this antagonism are unknown. Two small phase II trials have shown a response rate to Arzoxifene of >30% among women with receptor positive metastatic disease. We propose to perform a multi-institutional trial of Arzoxifene in women with advanced or recurrent endometrial adenocarcinoma and to combine these clinical investigations with laboratory-based studies to identify potential mechanisms of action of this SERM.

Hypothesis and Specific Aims: Our aims are three fold:

  • Aim 1: To measure and compare the objective clinical response rates of Megace and Arzoxifene in women with advanced or recurrent adenocarcinoma of the endometrium. We will conduct a randomized phase II study with two treatment arms (Arzoxifene and Megace) and measure time to progression, response duration and survival of treated patients.
  • Aim 2: Correlate tumor response with receptor status and signaling pathways present in the tumors to identify potential biomarkers of response to Arzoxifene and Megace. Patient materials from the clinical study in Aim 1 will be characterized for the presence of estrogen receptors (ER), progesterone receptors (PR), and transforming growth factor b receptors I and II ( TGF b RI/II).
  • Aim 3: Determine the mechanism of action of SERMs that function as antagonists in the endometrium. We hypothesize that “agonist marker genes” expressed in response to agonists but not antagonists act as critical cellular gatekeepers, enabling agonist but not antagonist treated cells to enter the cell cycle. Using primary cell cultures as a novel in vitro model system, expression profiling with microarrays will be performed on endometrial carcinoma cells to identify genes differentially expressed in response to agonists (estrogen or tamoxifen) versus antagonist (Arzoxifene).

Significance: Completion of the clinical trial will substantiate the effectiveness of Arzoxifene as a therapeutic agent. Laboratory correlates performed in conjunction with this trial will provide insight into the molecular mechanism of antagonism of Arzoxifene in endometrial carcinoma.


Project 2
A NOVEL ENDOMETRIAL CANCER CHEMOPREVENTION STRATEGY FOR OBESE, INSULIN-RESISTANT WOMEN – AN AT-RISK POPULATION

Principal Investigators: Karen H. Lu, M.D.; Peter J.A. Davies, M.D., Ph.D.
Co-Investigators: Diane C. Bodurka, M.D.; Michele Follen, M.D., Ph.D.

Background: Our studies are based on the observation that obesity is a strong risk factor for the development of endometrial cancer in post-menopausal women. Although part of this increased risk appears to be linked with hyper-estrogenization associated with obesity, it is our hypothesis that hyper-insulinemia also contributes to the development of endometrial cancer. Drugs that suppress insulin resistance, such as the thiazolidinedione insulin sensitizers, may have activity in suppressing abnormal endometrial proliferation in obese post-menopausal women. We speculate that insulin sensitizer therapy may constitute a novel, safe effective form of chemopreventive therapy for this disease.

Hypotheses and Specific Aims: Our approach involves three distinct experimental modalities.

  • Aim 1: To test the hypothesis that obesity and insulin resistance increase estrogen-dependent proliferation in the endometrium on animal models of obesity and insulinresistance. In addition, the ability of insulin sensitizers to reverse this effect will be studied. Specific genes involved in estrogen-regulated proliferation pathways will be examined by quantitative PCR, including hormone receptors and co-activators, and genes involved in the Wnt pathway, IGF pathway, TGF- b and retinoid pathway.
  • Aim 2: To identify novel genes and pathways associated with insulin resistance and estrogenization, and to identify relevant and specific biomarkers that are modulated by normalization of insulin resistance. Expression profiling with cDNA microarrays will be used to explore the effects of obesity and insulin resistance on both estrogen-dependent and estrogen-independent endometrial gene expression. In addition, specific biomarkers of the endometrium that are modulated by insulin-sensitizers will be identified.
  • Aim 3: To assess the ability of an insulin-sensitizing agent to modulate surrogate endometrial biomarkers in a post-menopausal cohort of obese, insulin resistant women. The post-menopausal cohort includes obese women (body mass index > 30kg/m2) who demonstrate insulin resistance based on an oral glucose tolerance test, but who do not fulfill criteria for Type II Diabetes. We hypothesize that rosiglitazone, an insulin-sensitizing agent, will modulate relevant endometrial proliferation biomarkers. In pre-and post–treatment endometrial biopsies, we will assess expression levels of genes involved in estrogen-regulated growth pathways, specific biomarkers as defined in Aim 2, and histologic and proliferation markers. As secondary endpoints, we will establish a point estimate of the baseline frequency of endometrial abnormalities in this cohort and we will determine changes in serum levels of estradiol, estrone, testosterone, DHEA-S and SHBG in obese, post-menopausal women taking rosiglitazone.

Significance: Increased endometrial cancer risk most closely associated with obesity, but there is also strong evidence that obesity increases risk of post-menopausal breast cancer. This proposal will examine the contribution of obesity and insulin resistance to the increased risk of estrogen associated cancers, using endometrial cancer as a model. Certainly the ability to decrease breast cancer risk, as well as endometrial cancer risk, with insulin sensitizers would have a major public health impact. This study also proposes a novel chemoprevention strategy. Rather than using an agent that focuses on prevention of a single cancer, we propose to treat an underlying metabolic syndrome that may decrease the risk of several hormonally associated cancers. In addition, use of an insulin-sensitizing agent may have a more favorable risk: benefit ratio than other options, including aromatase inhibitors or selective estrogen receptor modulators.


Project 3
METHYLATION PROFILING OF ENDOMETRIAL CANCER

Principal Investigators: Russell R. Broaddus, M.D., Ph.D.; Jean-Pierre Issa, M.D.

Background: Methylation of CpG islands is an epigenetic mechanism of regulating gene expression that has been described in great detail for colon cancer. Methylation is an important mechanism of gene silencing that can suppress the expression of tumor suppressor genes and other growth regulatory genes important in neoplasia. We hypothesize that methylation is an important mechanism of gene silencing in a subset of endometrial cancers. Endometrial cancer is a complex, heterogeneous disease that can be broadly classified into two groups according to histopathological, molecular, and clinical characteristics. Type 1 endometrial cancer is generally associated with estrogen exposure and is less clinically less aggressive. Type 2 cancers are not dependent on estrogen and are associated with local recurrence, distant metastasis, and shortened survival. There are numerous patient populations at risk for developing endometrial cancer. High-risk characteristics include endometrial hyperplasia, anovulatory cycle, tamoxifen use, obesity with insulin-resistance, and exposure to environmental estrogens.

Hypotheses and Specific Aims: For the first three specific aims, a large panel of markers known to be methylated in various cancers will be used to generate molecular fingerprints of endometrial tissues.

  • Aim 1 will examine CpG island methylation in Type 1 and Type 2 endometrial cancers to support the hypothesis that methylation profiling can provide molecular clues to the differences between Type 1 and Type 2 cancers.
  • Aim 2 will use methylation profiling of endometrial hyperplasia, a precursor to endometrial cancer, to determine if CpG island methylation is an early event.
  • Aim 3 will determine if methylation profiling can accurately identify “at-risk” endometrium.
  • Aim 4 will clone genes differentially methylated in endometrial cancer. This aim will allow for the discovery of novel markers of methylation that can potentially be used as therapeutic targets or tissue prognostic indicators.

Significance: The main goal of the proposed project is to evaluate CpG island methylation as a mechanism of gene silencing in endometrial cancer. As a result of the experiments proposed for this project, we expect to use methylation profiling as unique molecular fingerprints of the different types of endometrial cancer. These methylation profiles will then be tested as possible prognostic indicators in women at risk for the development of endometrial cancer. Groups at risk for the development of endometrial cancer include women with endometrial hyperplasia, women treated with tamoxifen for breast cancer, and women with anovulatory cycles. The methylation profiles tested in the cancers will be applied to each of these high-risk groups. Finally, we will use well-established techniques to clone and identify genes differentially methylated in endometrial cancer. Identification of such genes with differentiated methylation patterns will hopefully provide molecular clues as to the fundamental differences between type 1 endometrial cancers (less aggressive) and type 2 endometrial cancer (more aggressive).


Project 4
MOLECULAR PROGRESSION OF ENDOMETRIAL CANCER

Principal Investigators: David Loose, M.D.; Judith Wolf, M.D.
Co-Investigators: Gregory L. Shipley, Ph.D.; George M. Stancel, Ph.D.

Background: Estrogen exposure is well documented to be important in the pathogenesis of endometrial cancer. We hypothesize that normal growth control pathways induced by estrogen are dysregulated in endometrial hyperplasia and cancer. By identifying genes that are dysregulated, we can 1) further our understanding of the step-wise progression in endometrial carcinogenesis and 2) identify genes that can be used as surrogate biomarkers of endometrial cancer risk. Identification of these tissue biomarkers is crucial for the successful design of chemoprevention trials. In addition, these biomarkers potentially can be used clinically as tissue indicators of risk in high-risk groups. High-risk groups include women using SERMs such as tamoxifen, women with HNPCC, and women with obesity and type 2 diabetes. We have recently analyzed gene expression changes in post-menopausal endometrium exposed to different estrogen preparation using DNA microchip array analysis and real-time quantitative PCR (Q-PCR). The array analysis identified several hundred genes that are regulated by estrogen. From this large set of estrogen regulated genes, and additional genes selected for their role in the development of endometrial hyperplasia, a set of 33 transcripts of identified that are involved in proliferation of the endometrium.

Hypothesis and Specific Aims: We hypothesize that normal growth control pathways induced by estrogen are dysregulated in endometrial hyperplasia and cancer. Our approach will be to make quantitative measurements of mRNA levels of this set of potential biomarkers in endometrial biopsy samples in different states of proliferation: normal, hyperplasia, and cancer.

  • Aim 1 : Quantitate the set of 33 candidate biomarkers in women treated with conjugated estrogens; normal secretory and normal proliferative pre-menopausal endometrium; hyperplastic endometrium both with and without nearby cancer; type I endometrial cancer; and type 2 endometrial cancer. Using laser capture microdissection, we will focus on the precancerous lesion endometrial intraepithelial neoplasia (EIN) and assess transcript levels in EIN and in normal adjacent regions.
  • Aim 2 : High density microarrays will be screened to identify new genes whose expression is altered in hyperplasia and cancer compared to normal endometrium. Genes that are identified by the microchip analysis will be validated by Q-PCR. The combination of aims 1 and 2 will identify a set of “abnormal proliferation” biomarker transcripts.
  • Aim 3 : We will test the ability of the biomarkers to predict changes in proliferation by quantitating transcript levels in women treated with progestins for atypical hyperplasia. We hypothesize that biomarkers that are elevated in abnormal proliferation will be reduced by progestin treatment. Biomarkers will also be quantitated in endometrial biopsies from women treated with tamoxifen, a group at risk for developing endometrial cancer. This panel of biomarkers may be extended for use in the future analysis of endometrial effects of newer SERMs.

Significance: The overall goal of this project is to identify and validate an essemble of biomarkers whose expression in the endometrium changes during the progression from normal quiescent to normal proliferative through hyperplastic states into carcinoma. A combination of qualitative and quantitative measurements will be made to identify candidate biomarkers. Such a genetic essemble will be useful for identifying women at risk for endometrial cancer and for providing molecular indicators of prognosis.


ADMINISTRATIVE CORE
Directors: Thomas W. Burke, M.D.; George M. Stancel, Ph.D.; Karen H. Lu, M.D.; Russell R. Broaddus, M.D., Ph.D.

The overall goal of the Administrative Core is the effective management of all activities relating to the SPORE. The Administrative Core will actively work to communicate and disseminate information to both SPORE investigators and to external constituencies. Dr. Thomas Burke, Principal Investigator of the Administrative Core and Overall Project, will direct these activities. Specific aims are to:

  • Plan and oversee all SPORE activities.
  • Provide administrative support for the Developmental Research and Career Development Programs.
  • Communicate and consult with NCI Chief of Organ System Branch, Dr. Jorge Gomez, and Program Director, Dr. Jane Fountain, to assure the accurate and timely submission of all required reports and publications and to promptly notify the project officer of important events that affect the management of the SPORE.
  • Coordinate quality assurance activities, including data quality control through the Principal Investigators Committee, and Internal and External Advisory Committees with support from the Biostatistics and Pathology Cores.
  • Oversee all fiscal and budgetary issues.
  • Convene and provide administrative support for meetings of the Principal Investigators Committee, Internal Advisory Committee, and External Advisory Committee.
  • Assure compliance with all general, governmental, NCI, and institutional regulations and requirements.
  • Support policies for recruitment of women and minorities.
  • Coordinate research projects

PATHOLOGY CORE
Director: Russell R. Broaddus, M.D., Ph.D.

The goal of the Pathology Core is to provide frozen tissue, paraffin-embedded tissue, and histopathological expertise related to the specific needs for the research projects within this SPORE program. To achieve this goal, the Pathology Core proposed the following specific aims:

  • Aim 1: To maintain a frozen and paraffin-embedded repository of endometrial cancers, hyperplasias, and normal endometrial samples.
  • Aim 2: To provide pathological review for all clinical specimens utilized in the SPORE projects and to provide histopathological technical services as necessary. Such technical services include immunohistochemistry, in situ hybridization, and microdissection of tissue sections.
  • Aim 3: To establish a blood/urine/ascites fluid repository from patients undergoing hysterectomy for endometrial cancer and endometrial hyperplasia. These fluids will provide the resources for the systematic testing of putative prognostic and diagnostic markers isolated from endometrial tissues. Furthermore, using the novel technique of phage display, novel tumor markers can be discovered from the serum or ascites fluid of endometrial cancer patients.
  • Aim 4: To construct various endometrial tissue arrays using the Beecher Instruments microarray device. Such tissue arrays will provide for more rapid immunohistochemical analysis of protein expression.
  • Aim 5: To create a SPORE Database for all samples collected at M.D. Anderson Cancer Center. The database will provide for a virtual tissue repository that can be electronically shared with all SPORE investigators.

BIOMARKER CORE
Director: David S. Loose, Ph.D.

The Biomarker Core will provide a centralized resource for the rapid and high throughput quantitation of transcripts using fluorescent real-time reverse transcriptase-coupled quantitative PCR (Q-PCR). The use of the Biomarker Core will serve fundamental purposes:

  1. Quantitative assessment of gene expression at the mRNA level for known genes that are involved in proliferation and implicated in cancer progression, and genes that are known markers for estrogen action in either normal or malignant endometrium.
  2. Quantitative validation of the expression of genes that are initially identified in screening high-density microarrays. This is a critical adjunct to the microarray screening and will require development of a significant number of new Q-PCR assays.

The following specific aims represent interactions between projects in the SPORE and the Biomarker Core:

  • Aim 1 – Endometrial Carcinoma and SERMs (Burke/Walker): This project will examine the clinical response of women with endometrial cancer to the SERM Arzoxifene and an important component of this will be the identification of potential biomarkers of efficacy. Primary cultures of endometrial epithelial cells (normal and neoplastic) will be analyzed for changes in gene expression in response to SERMs. Total RNA will be prepared from these cells and Q-PCR will be performed using a broad panel of endometiral candidate biomarkers. In addition, novel genes identified by microarray screening in this study will be validated by Q-PCR.
  • Aim 2 – Insulin Resistance and Endometrial Cancer (Lu/Davies): This project will use a preclinical model of insulin resistance in the rat to examine biomarkers in the endometrium that are associated with the risk factors of obesity and insulin resistance. Expression of a panel of known genes will be used to initially examine endometrial response to estrogens following treatment the insulin sensitizer rosaglitazone. Additional biomarker candidates will be identified by microarray hybridization and subsequently validated by Q-PCR. The clinical arm of this study will examine candidate biomarker expression in obese, insulin-resistant post-menopausal women treated with rosaglitazone.
  • Aim 3 – Molecular Progression of Endometrial Cancer (Loose/Wolf): This project will identify potential biomarkers whose expression changes as cells progress from a normal phenotype, to the precancerous endometrial intraepithelial neoplasm, to hyperplasia, and to endometrial cancer. Our approach to identifying potential biomarkers will also use the 2-stage approach initially examining a panel of 33 known endometrial transcripts in the first years of the SPORE followed by microarray comparisons between the phenotypes. Newly discovered genes whose expression is altered during the progression will be validated by Q-PCR. The predictive ability of the candidate biomarkers will be tested in women with hyperplasia treated with Megace.

BIOSTATISTICS & DATA MANAGEMENT CORE
Director: Peter Mueller, Ph.D.

The Biostatistics and Data Management Core will serve the multiple needs for the planning and conduct of the SPORE’s translational research. This resource will be used for hypothesis refinement, experimental design, data management, quality control, result analysis and information presentation of results, and will function across all projects of the SPORE. Data from SPORE clinical trials and laboratory projects will be entered into a customized database application developed specifically for the SPORE. This computerized database will facilitate continuous monitoring of clinical trial results and will allow for automated data audits. Thus, from inception to reporting, translational experiments will benefit from SPORE resources that will be used to augment existing M.D. Anderson biostatistics resources. The specific aims of the Biostatistics and Data Management Core are:

  • To provide guidance in the design and conduct of clinical trials and other experiments arising from the ongoing research of the SPORE.
  • To provide innovative statistical modeling, simulation techniques, and data analyses needed by the Projects, Developmental Projects, and other Cores to achieve their Specific Aims.
  • To ensure that the results of all Projects are based on well-designed experiments and are appropriately interpreted.

DEVELOPMENTAL RESEARCH PROGRAM
Directors: Cheryl Walker, Ph.D.; Peter J.A. Davies, M.D., Ph.D.; Charles Levenback, M.D.

The overall aim of the Developmental Research Program is to provide initial funding to support innovative translational studies and exploratory research in gynecologic oncology. The support of translational research projects that can generate clinically testable hypotheses aimed at reducing the incidence of uterine cancer or leading to improved therapeutic outcomes or quality of life for gynecologic cancer patients will be emphasized. Pre-proposals in the form of project abstracts (1/2 – 1 page) will be solicited by the Program and applicants of responsive abstracts invited to submit Pilot Project proposals to the program. Proposals will follow an abbreviated R01 research grant format and be up to 5 pages in length. The Program Directors in conjunction with the Executive Committee of the SPORE will help investigators submitting proposals formulate relevant translational research aims and plans, as many investigators may not have previous expertise in this area. This process will therefore be a major educational activity that is anticipated to further stimulate translational research in gynecologic oncology and encourage the participation of both basic researchers and clinicians in translational research. Competing proposals will be evaluated and ranked by the SPORE Executive Committee. Projects will be funded for 1 year with the possibility of renewal for an additional year. It is anticipated that the Program will fund 2 pilot projects each year. All projects will be reviewed at the end of their first year with written progress reports and oral presentations at the SPORE annual retreat.


CAREER DEVELOPMENT PROGRAM
Directors: George M. Stancel, Ph.D.; David M. Gershenson, M.D.

The goal of the Career Development Program is to develop highly trained investigators dedicated to translational studies in human uterine cancer. The program will:

Recruit and train physician and basic science postdoctoral fellows to become highly skilled translational investigators in the field of uterine cancer;

  1. Provide specific knowledge of research in uterine cancer that will have the ability of awardees to conduct innovative translational science that will directly impact on the understanding and treatment of this disease,
  2. Redirect individuals who already have shown considerable scientific promise into uterine cancer research.

The unique educational environment that exists at M.D. Anderson Cancer Center, the University of Texas Health Science Center, and other institutions of the Texas Medical Center will aid in achieving these goals. Solicitations will be made for qualified candidates from M.D. Anderson Cancer Center, including physicians in our Gynecologic Oncology and Pathology fellowship programs, basic science graduates of our Graduate School of Biomedical Sciences, and residents/fellows in the Department of Obstetrics, Gynecology, and Reproductive Sciences at the U.T. Houston Medical School, as well as M.D. and Ph.D. candidates identified by national advertisements. The SPORE Executive Committee will interview candidates, make the final selection of fellows, and approve the placement of the trainee in a research laboratory, making this decision based upon the interests and research experience of the trainee and the availability of suitable resources. Minorities and women will be encouraged to apply by several specific mechanisms. In additional to research training via their individual projects, fellows will participate in activities designed to provide an overview of current knowledge, problems, and opportunities in uterine cancer research, state-of-the-art techniques and approaches for translational research, career development activities such as grant preparation and seminar presentations, and the opportunities to interact with established senior investigators in human uterine cancer research. Some of these enrichment activities will be open to the entire Texas Medical Center community to further increase knowledge and interest in uterine cancer issues within the wider biomedical community.


M.D. Anderson Gynecologic SPORE for Uterine Cancer List of Investigators

Diane C. Bodurka, M.D.
Associate Professor
M.D. Anderson Cancer Center
Department of Gynecologic Oncology
1515 Holcombe Blvd., Box 440
Houston , Texas 77030

Russell R. Broaddus, M.D., Ph.D.
Assistant Professor
M.D. Anderson Cancer Center
Department of Pathology
1515 Holcombe Blvd., Box 85
Houston , Texas 77030

Thomas W. Burke, M.D.
Professor
VP & Chief Medical Officer
M.D. Anderson Cancer Center
1515 Holcombe Blvd., Box 43
Houston , Texas 77030

Peter J.A. Davies, M.D., Ph.D.
Professor
UT Health Science Center at Houston
Department of Integrative Biology & Pharmacology
6431 Fannin, MSB 5.136
Houston , Texas 77030

Michele Follen, M.D., Ph.D.
Professor
M.D. Anderson Cancer Center
Biomedical Engineering Center
1515 Holcombe Blvd., Box 193
Houston , Texas 77030

David M. Gershenson, M.D.
Professor
Department Chair
M.D. Anderson Cancer Center
Department of Gynecology Oncology
1515 Holcombe Blvd., Box 440
Houston, Texas 77030

Leslie I. Gold, Ph.D.
Associate Professor
New York University School of Medicine
Department of Pathology
550 First Avenue, NB 16N28
New York , NY 10016

Jean-Pierre Issa, M.D.
Professor
M.D. Anderson Cancer Center
Department of Leukemia
1515 Holcombe Blvd., Box 428
Houston , Texas 77030

Charles Levenback, M.D.
Professor
M.D. Anderson Cancer Center
Department of Gynecologic Oncology
1515 Holcombe Blvd., Box 440
Houston , Texas 77030

David S. Loose, Ph.D.
Associate Professor
UT Health Science Center at Houston
Department of Integrative Biology & Pharmacology
6431 Fannin, MSB 5.136
Houston , Texas 77030

Karen H. Lu, M.D.
Assistant Professor
M.D. Anderson Cancer Center
Department of Gynecologic Oncology
1515 Holcombe Blvd., Box 440
Houston , Texas 77030

Peter Mueller, Ph.D.
Professor
M.D. Anderson Cancer Center
Department of Biostatistics
1515 Holcombe Blvd., Box 447
Houston , Texas 77030

Lois Ramondetta, M.D.
Assistant Professor
M.D. Anderson Cancer Center
Department of Gynecologic Oncology
1515 Holcombe Blvd., Box 440
Houston , Texas 77030

Gregory L. Shipley, Ph.D.
Assistant Professor
UT Health Science Center at Houston
Department of Integrative Biology & Pharmacology
P.O. Box 20708
Houston , Texas 77225-0708

George M. Stancel, Ph.D.
Professor
Dean of Graduate School of Biomedical Sciences
UT Houston Graduate School of Biomedical Sciences
GSBS Dean’s Office
6655 Travis St. , HMC-300/06, MSB 5.114
Houston , Texas 77030

Cheryl L. Walker, Ph.D.
Professor
M.D. Anderson Cancer Center Science Park Research Division
Department of Carcinogenesis
Park Road 1C
Smithville , Texas 78957

Judith K. Wolf, M.D.
Associate Professor
M.D. Anderson Cancer Center
Department of Gynecologic Oncology
1515 Holcombe Blvd., Box 440
Houston , Texas 77030

 


National Cancer Institute Department of Health and Human Services National Institutes of Health FirstGov.gov