Network


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

Hotspot


Dive into the research topics where Gary J. Kelloff is active.

Publication


Featured researches published by Gary J. Kelloff.


The New England Journal of Medicine | 2000

THE EFFECT OF CELECOXIB, A CYCLOOXYGENASE-2 INHIBITOR, IN FAMILIAL ADENOMATOUS POLYPOSIS

Gideon Steinbach; Patrick M. Lynch; Robin K. S. Phillips; Marina Wallace; Ernest T. Hawk; Gary B. Gordon; Naoki Wakabayashi; Brian Saunders; Yu Shen; Takashi Fujimura; Li Kuo Su; Bernard Levin; Louis Godio; Sherri Patterson; Miguel A. Rodriguez-Bigas; Susan L. Jester; Karen L. King; Marta Schumacher; James L. Abbruzzese; Raymond N. DuBois; Walter N. Hittelman; Stuart O. Zimmerman; Jeffrey W. Sherman; Gary J. Kelloff

BACKGROUND Patients with familial adenomatous polyposis have a nearly 100 percent risk of colorectal cancer. In this disease, the chemopreventive effects of nonsteroidal antiinflammatory drugs may be related to their inhibition of cyclooxygenase-2. METHODS We studied the effect of celecoxib, a selective cyclooxygenase-2 inhibitor, on colorectal polyps in patients with familial adenomatous polyposis. In a double-blind, placebo-controlled study, we randomly assigned 77 patients to treatment with celecoxib (100 or 400 mg twice daily) or placebo for six months. Patients underwent endoscopy at the beginning and end of the study. We determined the number and size of polyps from photographs and videotapes; the response to treatment was expressed as the mean percent change from base line. RESULTS At base line, the mean (+/-SD) number of polyps in focal areas where polyps were counted was 15.5+/-13.4 in the 15 patients assigned to placebo, 11.5+/-8.5 in the 32 patients assigned to 100 mg of celecoxib twice a day, and 12.3+/-8.2 in the 30 patients assigned to 400 mg of celecoxib twice a day (P=0.66 for the comparison among groups). After six months, the patients receiving 400 mg of celecoxib twice a day had a 28.0 percent reduction in the mean number of colorectal polyps (P=0.003 for the comparison with placebo) and a 30.7 percent reduction in the polyp burden (the sum of polyp diameters) (P=0.001), as compared with reductions of 4.5 and 4.9 percent, respectively, in the placebo group. The improvement in the extent of colorectal polyposis in the group receiving 400 mg twice a day was confirmed by a panel of endoscopists who reviewed the videotapes. The reductions in the group receiving 100 mg of celecoxib twice a day were 11.9 percent (P=0.33 for the comparison with placebo) and 14.6 percent (P=0.09), respectively. The incidence of adverse events was similar among the groups. CONCLUSIONS In patients with familial adenomatous polyposis, six months of twice-daily treatment with 400 mg of celecoxib, a cyclooxygenase-2 inhibitor, leads to a significant reduction in the number of colorectal polyps.


Clinical Cancer Research | 2005

Progress and promise of FDG-PET imaging for cancer patient management and oncologic drug development

Gary J. Kelloff; John M. Hoffman; Bruce E. Johnson; Howard I. Scher; Barry A. Siegel; Edward Y. Cheng; Bruce D. Cheson; Joyce O'Shaughnessy; Kathryn Z. Guyton; David A. Mankoff; Lalitha K. Shankar; Steven M. Larson; Caroline C. Sigman; Richard L. Schilsky; Daniel C. Sullivan

2-[18F]Fluoro-2-deoxyglucose positron emission tomography (FDG-PET) assesses a fundamental property of neoplasia, the Warburg effect. This molecular imaging technique offers a complementary approach to anatomic imaging that is more sensitive and specific in certain cancers. FDG-PET has been widely applied in oncology primarily as a staging and restaging tool that can guide patient care. However, because it accurately detects recurrent or residual disease, FDG-PET also has significant potential for assessing therapy response. In this regard, it can improve patient management by identifying responders early, before tumor size is reduced; nonresponders could discontinue futile therapy. Moreover, a reduction in the FDG-PET signal within days or weeks of initiating therapy (e.g., in lymphoma, non–small cell lung, and esophageal cancer) significantly correlates with prolonged survival and other clinical end points now used in drug approvals. These findings suggest that FDG-PET could facilitate drug development as an early surrogate of clinical benefit. This article reviews the scientific basis of FDG-PET and its development and application as a valuable oncology imaging tool. Its potential to facilitate drug development in seven oncologic settings (lung, lymphoma, breast, prostate, sarcoma, colorectal, and ovary) is addressed. Recommendations include initial validation against approved therapies, retrospective analyses to define the magnitude of change indicative of response, further prospective validation as a surrogate of clinical benefit, and application as a phase II/III trial end point to accelerate evaluation and approval of novel regimens and therapies.


Clinical Pharmacology & Therapeutics | 2009

I‐SPY 2: An Adaptive Breast Cancer Trial Design in the Setting of Neoadjuvant Chemotherapy

Anna Barker; Cc Sigman; Gary J. Kelloff; Nola M. Hylton; Da Berry; Laura Esserman

I‐SPY 2 (investigation of serial studies to predict your therapeutic response with imaging and molecular analysis 2) is a process targeting the rapid, focused clinical development of paired oncologic therapies and biomarkers. The framework is an adaptive phase II clinical trial design in the neoadjuvant setting for women with locally advanced breast cancer. I‐SPY 2 is a collaborative effort among academic investigators, the National Cancer Institute, the US Food and Drug Administration, and the pharmaceutical and biotechnology industries under the auspices of the Foundation for the National Institutes of Health Biomarkers Consortium.


Molecular Carcinogenesis | 1999

CHEMOPREVENTIVE ACTIVITY OF CELECOXIB, A SPECIFIC CYCLOOXYGENASE-2 INHIBITOR, AND INDOMETHACIN AGAINST ULTRAVIOLET LIGHT-INDUCED SKIN CARCINOGENESIS

Susan M. Fischer; Herng-Hsang Lo; Gary B. Gordon; Karen Seibert; Gary J. Kelloff; Ronald A. Lubet; Claudio J. Conti

Epidemiological and dietary studies suggest that nonsteroidal anti‐inflammatory drugs (NSAIDs) reduce the risk of colon cancer, possibly through a mechanism involving inhibition of cyclooxygenase (COX)‐2, which is overexpressed in premalignant adenomatous polyps and colon cancer. Because ultraviolet light (UV) can induce COX‐2 and nonspecific NSAIDs can decrease UV‐induced skin cancer, we evaluated the ability of two compounds, celecoxib (a specific COX‐2 inhibitor) and indomethacin (a nonspecific NSAID), to block UV‐induced skin tumor development in SKH:HR‐1‐hrBr hairless mice. Mice fed 150 or 500 ppm celecoxib showed a dose‐dependent reduction (60% and 89%, respectively) in tumor yield. Indomethacin (4 ppm) reduced tumor yield by 78%. Although both acute and chronic UV exposure increased cell proliferation and edema, neither compound reduced these parameters. In contrast, UV‐induced prostaglandin synthesis in the epidermis was effectively blocked by both compounds. UV‐induced increases in COX‐2 expression in skin were also not altered in any of the treatment groups. Similarly, tumors that constitutively express high levels of COX‐2 displayed no reduction by treatment with celecoxib or indomethacin. The dramatic protective effects of celecoxib suggests that specific COX‐2 inhibitors may offer a way to safely reduce the risk of skin cancer in humans. Mol. Carcinog. 25:231–240, 1999.


Cancer Prevention Research | 2008

Difluoromethylornithine plus sulindac for the prevention of sporadic colorectal adenomas: a randomized placebo-controlled, double-blind trial.

Frank L. Meyskens; Christine E. McLaren; Daniel Pelot; Sharon Fujikawa-Brooks; Philip M. Carpenter; Ernest T. Hawk; Gary J. Kelloff; Michael J. Lawson; Jayashri Kidao; John McCracken; C. Gregory Albers; Dennis J. Ahnen; D. Kim Turgeon; Steven Goldschmid; Peter Lance; Curt H. Hagedorn; Daniel L. Gillen; Eugene W. Gerner

Abstract Preclinical studies of chemoprevention drugs given in combination at low doses show remarkable efficacy in preventing adenomas with little additional toxicities, suggesting a strategy to improve risk to benefit ratios for preventing recurrent adenomas. Three hundred seventy-five patients with history of resected (≥3 mm) adenomas were randomly assigned to receive oral difluoromethylornithine (DFMO) 500 mg and sulindac 150 mg once daily or matched placebos for 36 months, stratified by use of low-dose aspirin (81 mg) at baseline and clinical site. Follow-up colonoscopy was done 3 years after randomization or off-study. Colorectal adenoma recurrence was compared among the groups with log-binomial regression. Comparing the outcome in patients receiving placebos to those receiving active intervention, (a) the recurrence of one or more adenomas was 41.1% and 12.3% (risk ratio, 0.30; 95% confidence interval, 0.18-0.49; P < 0.001); (b) 8.5% had one or more advanced adenomas, compared with 0.7% of patients (risk ratio, 0.085; 95% confidence interval, 0.011-0.65; P < 0.001); and (c) 17 (13.2%) patients had multiple adenomas (>1) at the final colonoscopy, compared with 1 (0.7%; risk ratio, 0.055; 0.0074-0.41; P < 0.001). Serious adverse events (grade ≥3) occurred in 8.2% of patients in the placebo group, compared with 11% in the active intervention group (P = 0.35). There was no significant difference in the proportion of patients reporting hearing changes from baseline. Recurrent adenomatous polyps can be markedly reduced by a combination of low oral doses of DFMO and sulindac and with few side effects.


Advances in Cancer Research | 1999

Perspectives on cancer chemoprevention research and drug development.

Gary J. Kelloff

Publisher Summary This chapter focuses on the nature of carcinogenesis and provides an overview of the progress in this field, and presents successful strategies in chemoprevention. Chemoprevention is the use of agents to slow the progression of, reverse, or inhibit carcinogenesis. Chemopreventive agents modulate cancer risk at the molecular, cellular, tissue, and clinical levels. The mechanisms of these agents are defined by activities seen at the molecular and cellular levels, while chemopreventive efficacy is evaluated at the tissue and clinical levels. The chapter discusses principles of molecular target-based chemopreventive drug discovery and early development of five prototypical agent classes that showed significant chemopreventive activity, and reviews the rationale for their development. The chapter discusses the specific classes to the generality of molecular target- and empirical-based screening assays useful for discovery and early development of candidate chemopreventive agents. The rationale and methods for the development of a sequential drug development program, built on the rational mechanism-based and empirical agent discovery approach, are summarized. This program identifies and evaluates intermediate biomarkers as surrogate end points for cancer incidence. The chapter reviews the need for, the progress of, and the promise of chemoprevention at major cancer targets and describes a strategy for effectively using surrogate end points in defining chemopreventive efficacy. The chapter concludes with a discussion of major issues and challenges of chemoprevention drug development, and the tremendous opportunity it promises for reduction of the human cancer problem.


Clinical Cancer Research | 2006

Progress in Chemoprevention Drug Development: The Promise of Molecular Biomarkers for Prevention of Intraepithelial Neoplasia and Cancer—A Plan to Move Forward

Gary J. Kelloff; Scott M. Lippman; Andrew J. Dannenberg; Caroline C. Sigman; Homer L. Pearce; Brian J. Reid; Eva Szabo; V. Craig Jordan; Margaret R. Spitz; Gordon B. Mills; Vali Papadimitrakopoulou; Reuben Lotan; Bharat B. Aggarwal; Robert S. Bresalier; Jeri Kim; Banu Arun; Karen H. Lu; Melanie Thomas; Helen E. Rhodes; Molly Brewer; Michele Follen; Dong M. Shin; Howard L. Parnes; Jill M. Siegfried; Alison A. Evans; William J. Blot; Wong Ho Chow; Patricia L. Blount; Carlo C. Maley; Kenneth K. Wang

This article reviews progress in chemopreventive drug development, especially data and concepts that are new since the 2002 AACR report on treatment and prevention of intraepithelial neoplasia. Molecular biomarker expressions involved in mechanisms of carcinogenesis and genetic progression models of intraepithelial neoplasia are discussed and analyzed for how they can inform mechanism-based, molecularly targeted drug development as well as risk stratification, cohort selection, and end-point selection for clinical trials. We outline the concept of augmenting the risk, mechanistic, and disease data from histopathologic intraepithelial neoplasia assessments with molecular biomarker data. Updates of work in 10 clinical target organ sites include new data on molecular progression, significant completed trials, new agents of interest, and promising directions for future clinical studies. This overview concludes with strategies for accelerating chemopreventive drug development, such as integrating the best science into chemopreventive strategies and regulatory policy, providing incentives for industry to accelerate preventive drugs, fostering multisector cooperation in sharing clinical samples and data, and creating public-private partnerships to foster new regulatory policies and public education.


Carcinogenesis | 2000

Retinoids in chemoprevention and differentiation therapy

Laura Hansen; Caroline C. Sigman; Fausto Andreola; Sharon A. Ross; Gary J. Kelloff; Luigi M. De Luca

Retinoids are essential for the maintenance of epithelial differentiation. As such, they play a fundamental role in chemoprevention of epithelial carcinogenesis and in differentiation therapy. Physiological retinoic acid is obtained through two oxidation steps from dietary retinol, i.e. retinol-->retinal-->retinoic acid. The latter retinal-->retinoic acid step is irreversible and eventually marks disposal of this essential nutrient, through cytochrome P450-dependent oxidative steps. Mutant mice deficient in aryl hydrocarbon receptor (AHR) accumulate retinyl palmitate, retinol and retinoic acid. This suggests a direct connection between the AHR and retinoid homeostasis. Retinoids control gene expression through the nuclear retinoic acid receptors (RARs) alpha, beta and gamma and 9-cis-retinoic acid receptors alpha, beta and gamma, which bind with high affinity the natural ligands all-trans-retinoic acid and 9-cis-retinoic acid, respectively. Retinoids are effective chemopreventive agents against skin, head and neck, breast, liver and other forms of cancer. Differentiation therapy of acute promyelocytic leukemia (APL) is based on the ability of retinoic acid to induce differentiation of leukemic promyelocytes. Patients with relapsed, retinoid-resistant APL are now being treated with arsenic oxide, which results in apoptosis of the leukemic cells. Interestingly, induction of differentiation in promyelocytes and consequent remission of APL following retinoid therapy depends on expression of a chimeric PML-RAR alpha fusion protein resulting from a t(15;17) chromosomal translocation. This protein functions as a dominant negative against the function of both PML and RARs and its overexpression is able to recreate the phenotypes of the disease in transgenic mice. The development of new, more effective and less toxic retinoids, alone or in combination with other drugs, may provide additional avenues for cancer chemoprevention and differentiation therapy.


Journal of Translational Medicine | 2012

Considerations in the development of circulating tumor cell technology for clinical use

David R. Parkinson; Nicholas Dracopoli; Brenda Gumbs Petty; Carolyn C. Compton; Massimo Cristofanilli; Albert Deisseroth; Daniel F. Hayes; Gordon Kapke; Prasanna Kumar; Jerry S. H. Lee; Minetta C. Liu; Robert McCormack; Stanislaw Mikulski; Larry A. Nagahara; Klaus Pantel; Sonia Pearson-White; Elizabeth Punnoose; Lori T Roadcap; Andrew E. Schade; Howard I. Scher; Caroline C. Sigman; Gary J. Kelloff

This manuscript summarizes current thinking on the value and promise of evolving circulating tumor cell (CTC) technologies for cancer patient diagnosis, prognosis, and response to therapy, as well as accelerating oncologic drug development. Moving forward requires the application of the classic steps in biomarker development–analytical and clinical validation and clinical qualification for specific contexts of use. To that end, this review describes methods for interactive comparisons of proprietary new technologies, clinical trial designs, a clinical validation qualification strategy, and an approach for effectively carrying out this work through a public-private partnership that includes test developers, drug developers, clinical trialists, the US Food & Drug Administration (FDA) and the US National Cancer Institute (NCI).


Clinical Cancer Research | 2005

The Progress and Promise of Molecular Imaging Probes in Oncologic Drug Development

Gary J. Kelloff; Kenneth A. Krohn; Steven M. Larson; Ralph Weissleder; David A. Mankoff; John M. Hoffman; Jeanne M. Link; Kathryn Z. Guyton; William C. Eckelman; Howard I. Scher; Joyce O'Shaughnessy; Bruce D. Cheson; Caroline C. Sigman; James L. Tatum; George Q. Mills; Daniel C. Sullivan; Janet Woodcock

As addressed by the recent Food and Drug Administration Critical Path Initiative, tools are urgently needed to increase the speed, efficiency, and cost-effectiveness of drug development for cancer and other diseases. Molecular imaging probes developed based on recent scientific advances have great potential as oncologic drug development tools. Basic science studies using molecular imaging probes can help to identify and characterize disease-specific targets for oncologic drug therapy. Imaging end points, based on these disease-specific biomarkers, hold great promise to better define, stratify, and enrich study groups and to provide direct biological measures of response. Imaging-based biomarkers also have promise for speeding drug evaluation by supplementing or replacing preclinical and clinical pharmacokinetic and pharmacodynamic evaluations, including target interaction and modulation. Such analyses may be particularly valuable in early comparative studies among candidates designed to interact with the same molecular target. Finally, as response biomarkers, imaging end points that characterize tumor vitality, growth, or apoptosis can also serve as early surrogates of therapy success. This article outlines the scientific basis of oncology imaging probes and presents examples of probes that could facilitate progress. The current regulatory opportunities for new and existing probe development and testing are also reviewed, with a focus on recent Food and Drug Administration guidance to facilitate early clinical development of promising probes.

Collaboration


Dive into the Gary J. Kelloff's collaboration.

Top Co-Authors

Avatar

Vernon E. Steele

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Charles W. Boone

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ronald A. Lubet

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Caroline C. Sigman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

James A. Crowell

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ernest T. Hawk

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Greenwald

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Raymond V. Gilden

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Richard C. Moon

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge