Network


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

Hotspot


Dive into the research topics where Robert Kramer is active.

Publication


Featured researches published by Robert Kramer.


Pharmaceutical Biology | 2003

Novel Strategies for the Discovery of Plant-Derived Anticancer Agents.

Ad Kinghorn; Norman R. Farnsworth; Djaja D. Soejarto; Geoffrey A. Cordell; Steven M. Swanson; John M. Pezzuto; Mansukh C. Wani; Monroe E. Wall; Nicholas H. Oberlies; David J. Kroll; Robert Kramer; William C. Rose; Gregory D. Vite; Craig R. Fairchild; Russell Peterson; Robert Wild

Work has continued on the investigation of plants, collected mainly from tropical rainforests, as potential sources of new cancer chemotherapeutic agents. About 400 primary samples are obtained each year, with the chloroform-soluble extract of each being screened against a battery of in vitro assays housed at the three consortial sites in our current National Cooperative Drug Discovery Group (NCDDG) research project. An HPLC-MS dereplication procedure designed to screen out “nuisance” compounds has been refined. Several hundred secondary metabolites that are active in one or more of the primary assays utilized have been obtained in the project to date, and are representative of wide chemical diversity. Some of these are also active in various in vivo assays, inclusive of the hollow fiber assay, which was installed recently as part of our collaborative research effort. A number of bioactive compounds of interest to the project are described.


The FASEB Journal | 1998

Characterization of kringle domains of angiostatin as antagonists of endothelial cell migration, an important process in angiogenesis.

Weidong-Richard Ji; Francis J. Castellino; Yuan Chang; Melanie E. DeFord; Hilary Gray; Xavier Villarreal; Mohammad Eghtedarzadeh Kondri; Daniel Marti; Miguel Llinás; Johann Schaller; Robert Kramer; Pamela A. Trail

Angiogenesis is a complex process that involves endothelial cell proliferation, migration, basement membrane degradation, and neovessel organization. Angiostatin, consisting of four homologous triple‐disulfide bridged kringle domains, has previously been shown to exhibit profound inhibition of endothelial cell proliferation in vitro and angiogenesis in vivo. It was also demonstrated that angiostatin could suppress the growth of a variety of tumors via the blocking of angiogenesis. The primary aim of our study was to characterize the kringle domains of angiostatin for their inhibitory activities of endothelial cell migration in order to elucidate their contributions to the anti‐angiogenic function of angiostatin. In this report, we demonstrate for the first time that the kringles of angiostatin play different roles in inhibiting endothelial cell migration, a crucial process in angiogenesis. Kringle 4, which has only marginal anti‐proliferative activity, is among the most potent fragments in inhibiting endothelial cell migration (IC50 of approximately 500 nM). In contrast, kringle 1–3, which is equivalent to angiostatin in inhibiting endothelial cell proliferation, manifests only a modest anti‐migratory effect. The combination of kringle 1–3 and kringle 4 results in an anti‐migratory activity comparable to that of angiostatin. When kringle 1 is removed from kringle 1–3, the resulting kringle 2–3 becomes more potent than kringle 1–3. This implies that kringle 1, although virtually ineffective in inhibiting endothelial cell migration, may influence the conformation of kringle 1–3 to alter its anti‐migratory activity. We also show that disruption of the kringle structure by reducing/alkylating agents markedly attenuates the anti‐migratory activity of angiostatin, demonstrating the significance of kringle conformation in maintaining the anti‐angiogenic activity of angiostatin. Our data suggest that different kringle domains may contribute to the overall anti‐angiogenic function of angiostatin by their distinct anti‐migratory activities.—Ji, W. R., Castellino, F. J., Chang, Y., DeFord, M. E., Gray, H., Villarreal, X., Kondri, M. E., Marti, D. N., Llinás, M., Schaller, J., Kramer, R. A., and Trail, P. A. Characterization of kringle domains of angiostatin as antagonists of endothelial cell migration, an important process in angiogenesis. FASEB J. 12, 1731–1738 (1998)


Clinical Cancer Research | 2006

Dasatinib (BMS-354825) Pharmacokinetics and Pharmacodynamic Biomarkers in Animal Models Predict Optimal Clinical Exposure

Feng R. Luo; Zheng Yang; Amy Camuso; Richard Smykla; Kelly McGlinchey; Krista Fager; Christine Flefleh; Stephen Castaneda; Ivan Inigo; David Kan; Mei-Li Wen; Robert Kramer; Anne Blackwood-Chirchir; Francis Y. Lee

Purpose: Chronic myeloid leukemia (CML) is caused by reciprocal translocation between chromosomes 9 and 22, forming BCR-ABL, a constitutively activated tyrosine kinase. Imatinib mesylate, a selective inhibitor of BCR-ABL, represents current frontline therapy for CML; however, emerging evidence suggests that drug resistance to imatinib may limit its long-term success. To improve treatment options, dasatinib (BMS-354825) was developed as a novel, oral, multi-targeted kinase inhibitor of BCR-ABL and SRC family kinases. To date, dasatinib has shown promising anti-leukemic activity in preclinical models of CML and in phase I/II clinical studies in patients with imatinib-resistant or imatinib-intolerant disease. Experimental Design: The pharmacokinetic and pharmacodynamic biomarkers of dasatinib were investigated in K562 human CML xenografts grown s.c. in severe combined immunodeficient mice. Tumoral levels of phospho-BCR-ABL/phospho-CrkL were determined by Western blot. Results: Following a single oral administration of dasatinib at a preclinical efficacious dose of 1.25 or 2.5 mg/kg, tumoral phospho-BCR-ABL/phospho-CrkL were maximally inhibited at ∼3 hours and recovered to basal levels by 24 hours. The time course and extent of the inhibition correlated with the plasma levels of dasatinib in mice. Pharmacokinetic/biomarker modeling predicted that the plasma concentration of dasatinib required to inhibit 90% of phospho-BCR-ABL in vivo was 10.9 ng/mL in mice and 14.6 ng/mL in humans, which is within the range of concentrations achieved in CML patients who responded to dasatinib treatment in the clinic. Conclusions: Phospho-BCR-ABL/phospho-CrkL are likely to be useful clinical biomarkers for the assessment of BCR-ABL kinase inhibition by dasatinib.


Cancer Chemotherapy and Pharmacology | 2009

Preclinical efficacy spectrum and pharmacokinetics of ixabepilone

Francis Y. Lee; Richard Smykla; Kathy A. Johnston; Krista Menard; Kelly McGlinchey; Russell Peterson; Amy Wiebesiek; Gregory D. Vite; Craig R. Fairchild; Robert Kramer

PurposeIxabepilone, a semisynthetic analog of natural epothilone B, was developed for use in cancer treatment. This study extends previous findings regarding the efficacy of ixabepilone and its low susceptibility to tumor resistance mechanisms and describes the pharmacokinetics of this new antineoplastic agent.MethodsThe cytotoxicity of ixabepilone was assessed in vitro in breast, lung, and colon tumor cell lines and in vivo in human xenografts in mice. Antitumor activities of ixabepilone and taxanes were compared in multidrug-resistant models in vivo. Differential drug uptake of ixabepilone and paclitaxel was assessed in a P-glycoprotein (P-gp)-resistant colon cancer model in vitro. The pharmacokinetic profile of ixabepilone was established in mice and humans.ResultsIxabepilone demonstrated potent cytotoxicity in a broad range of human cancer cell lines in vitro and in a wide range of xenografts in vivo. Ixabepilone was ~3-fold more potent than docetaxel in the paclitaxel-resistant Pat-21 xenograft model (resistant due to overexpression of βIII-tubulin and a lack of βII-tubulin). Ixabepilone activity against P-gp-overexpressing breast and colon cancer was confirmed in in vivo models. Cellular uptake of ixabepilone, but not paclitaxel, was established in a P-gp-overexpressing model. The pharmacokinetics of ixabepilone was characterized by rapid tissue distribution and extensive tissue binding.ConclusionsCytotoxicity studies against a range of tumor types in vitro and in vivo demonstrate that ixabepilone has potent and broad-spectrum antineoplastic activity. This is accompanied by favorable pharmacokinetics. Ixabepilone has reduced susceptibility to resistance due to P-gp overexpression, tubulin mutations, and alterations in β-tubulin isotype expression.


Cancer Chemotherapy and Pharmacology | 2008

Preclinical discovery of ixabepilone, a highly active antineoplastic agent.

Francis Y. Lee; Robert M. Borzilleri; Craig R. Fairchild; Amrita Kamath; Richard Smykla; Robert Kramer; Gregory D. Vite

The epothilones and their analogs constitute a novel class of antineoplastic agents, produced by the myxobacterium Sorangium cellulosum. These antimicrotubule agents act in a similar manner to taxanes, stabilizing microtubules and resulting in arrested tumor cell division and apoptosis. Unlike taxanes, however, epothilones and their analogs are macrolide antibiotics, with a distinct tubulin binding mode and reduced susceptibility to a range of common tumor resistance mechanisms that limit the effectiveness of taxanes and anthracyclines. While natural epothilones A and B show potent antineoplastic activity in vitro, these effects were not seen in preclinical in vivo models due to their poor metabolic stability and unfavorable pharmacokinetics. A range of epothilone analogs was synthesized, therefore, with the aim of identifying those with more favorable characteristics. Here, we describe the preclinical characterization and selection of ixabepilone, a semi-synthetic epothilone B analog, among many other epothilone analogs. Ixabepilone demonstrated superior preclinical characteristics, including high metabolic stability, low plasma protein binding and low susceptibility to multidrug resistance protein-mediated efflux, all of which were predictive of potent in vivo cell-killing activity. Ixabepilone also demonstrated in vivo antitumor activity in a range of human tumor models, several of which displayed resistance to commonly used agents such as anthracyclines and taxanes. These favorable preclinical characteristics have since translated to the clinic. Ixabepilone has shown promising phase II clinical efficacy and acceptable tolerability in a wide range of cancers, including heavily pretreated and drug-resistant tumors. Based on these results, a randomized phase III trial was conducted in anthracycline-pretreated or resistant and taxane-resistant metastatic breast cancer to evaluate ixabepilone in combination with capecitabine. Ixabepilone combination therapy showed significantly superior progression-free survival and tumor responses over capecitabine alone.


Pure and Applied Chemistry | 2009

Discovery of anticancer agents of diverse natural origin.

A. Douglas Kinghorn; Hee Byung Chai; Jimmy Orjala; Norman R. Farnsworth; D. Doel Soejarto; Nicholas H. Oberlies; Mansukh C. Wani; David J. Kroll; Cedric J. Pearce; Steven M. Swanson; Robert Kramer; William C. Rose; Craig R. Fairchild; Gregory D. Vite; Stuart Emanuel; David Jarjoura; Frederick O. Cope

A collaborative multidisciplinary research project is described in which new natural product anticancer drug leads are obtained from a diverse group of organisms, constituted by tropical plants, aquatic cyanobacteria, and filamentous fungi. Information is provided on how these organisms are collected and processed. The types of bioassays are indicated in which crude extracts of these acquisitions are tested. Progress made in the isolation of lead bioactive secondary metabolites from three tropical plants is discussed.


Cancer Research | 2009

Discovery of BMS-641988, a Novel and Potent Inhibitor of Androgen Receptor Signaling for the Treatment of Prostate Cancer

Ricardo M. Attar; Maria Jure-Kunkel; Aaron Balog; Mary Ellen Cvijic; Janet Dell-John; Cheryl A. Rizzo; Liang Schweizer; Thomas Spires; J. Suso Platero; Mary T. Obermeier; Weifang Shan; Mark E. Salvati; William R. Foster; Joseph E. Dinchuk; Shen-Jue Chen; Gregory D. Vite; Robert Kramer; Marco M. Gottardis

Despite an excellent initial response to first-line hormonal treatment, most patients with metastatic prostate cancer will succumb to a hormone-refractory form of the disease. Because these tumors are still dependent on a functional androgen receptor (AR), there is a need to find novel and more potent antiandrogens. While searching for small molecules that bind to the AR and inhibit its transcriptional activity, BMS-641988 was discovered. This novel antiandrogen showed an increased (>1 log) potency compared with the standard antiandrogen, bicalutamide, in both binding affinity to the AR and inhibition of AR-mediated transactivation in cell-based reporter assays. In mature rats, BMS-641988 strongly inhibited androgen-dependent growth of the ventral prostate and seminal vesicles. In the CWR-22-BMSLD1 human prostate cancer xenograft model, BMS-641988 showed increased efficacy over bicalutamide (average percent tumor growth inhibition >90% versus <50%), even at exposure levels of bicalutamide 3-fold greater than what can be attained in humans. Furthermore, BMS-641988 was efficacious in CWR-22-BMSLD1 tumors initially refractory to treatment with bicalutamide. BMS-641988 was highly efficacious in the LuCaP 23.1 human prostate xenograft model, inducing stasis throughout the approximately 30-day dosing. To explore the functional mechanisms of BMS-641988, gene expression profiling analysis was done on CWR-22-BMSLD1 xenograft models in mice. Treatment with BMS-641988 resulted in a global gene expression profile more similar to castration compared with that of bicalutamide. Overall, these data highlight that the unique preclinical profile of BMS-641988 may provide additional understanding for the hormonal treatment of prostate cancer.


Clinical Cancer Research | 2008

Synergistic Antitumor Activity of Ixabepilone (BMS-247550) Plus Bevacizumab in Multiple In vivo Tumor Models

Francis Y. Lee; Kelly Covello; Stephen Castaneda; Donald R. Hawken; David Kan; Anne Lewin; Mei-Li Wen; Rolf-Peter Ryseck; Craig R. Fairchild; Joseph Fargnoli; Robert Kramer

Purpose: Angiogenesis is a critical step in the establishment, growth, and metastasis of solid tumors, and combination of antiangiogenic agents with chemotherapy is an attractive therapeutic option. We investigated the potential of ixabepilone, the first in a new class of antineoplastic agents known as epothilones, to synergize with antiangiogenic agents to inhibit tumor growth. Experimental Design:In vitro and in vivo cytotoxicity of ixabepilone as single agent and in combination with two targeted antiangiogenic agents, bevacizumab or sunitinib, were examined in preclinical tumor models. Direct effects of the agents against endothelial cells was also examined and compared with the effects of paclitaxel as single agent and in combination with bevacizumab. Results: Ixabepilone showed robust synergistic antitumor activity in combination with bevacizumab and sunitinib in preclinical in vivo models derived from breast, colon, lung, and kidney cancers. The synergistic antitumor effect was greater with ixabepilone compared with paclitaxel. Furthermore, ixabepilone was more effective than paclitaxel at killing endothelial cells expressing P-glycoprotein in vitro and inhibiting endothelial cell proliferation and tumor angiogenesis in vivo. Conclusions: Ixabepilone may enhance the antitumor effects of antiangiogenic therapy by direct cytotoxicity and also indirectly via the killing of tumor-associated endothelial cells. Given that ixabepilone has reduced susceptibility to drug efflux pumps compared with taxanes, these data may explain the increased antiangiogenic and antitumor activity of ixabepilone in combination with antiangiogenic agents. Phase II studies to assess the efficacy and safety of ixabepilone plus bevacizumab in locally recurrent or metastatic breast cancer are planned.


Clinical Cancer Research | 2005

Prediction of Active Drug Plasma Concentrations Achieved in Cancer Patients by Pharmacodynamic Biomarkers Identified from the Geo Human Colon Carcinoma Xenograft Model

Feng R. Luo; Zheng Yang; Huijin Dong; Amy Camuso; Kelly McGlinchey; Krista Fager; Christine Flefleh; David Kan; Ivan Inigo; Stephen Castaneda; Tai W. Wong; Robert Kramer; Robert Wild; Francis Y. Lee

Purpose: Epidermal growth factor receptor (EGFR), a protein tyrosine kinase expressed in many types of human cancers, has been strongly associated with tumor progression. Cetuximab is an IgG1 anti-EGFR chimeric mouse/human monoclonal antibody that has been approved for the treatment of advanced colon cancer. Using human tumor xenografts grown in nude mice, we have determined the in vivo pharmacodynamic response of cetuximab at efficacious doses. Three pharmacodynamic end points were evaluated: tumoral phospho-EGFR, tumoral mitogen-activated protein kinase (MAPK) phosphorylation, and Ki67 expression. Experimental Design: The pharmacodynamic study was conducted in nude mice bearing Geo tumors following a single i.p. administration of 0.25 and 0.04 mg. The tumors were analyzed by immunohistochemistry. The levels of phospho-EGFR were quantitated by an ELISA assay. Results: At 0.25 mg, phospho-EGFR was maximally inhibited by 91% at 24 hours, whereas the level of inhibition decreased to 72% by 72 hours. At 0.04 mg, the maximum inhibition of phospho-EGFR was 53% at 24 hours, whereas the level of inhibition decreased to 37% by 72 hours. The time course of phospho-EGFR inhibition and recovery seemed to correlate with the pharmacokinetics of cetuximab. Immunohistochemical analysis showed that phospho-MAPK and Ki67 expression were inhibited between 24 and 72 hours at 0.25 and 0.04 mg. A pharmacokinetic/pharmacodynamic model was established and predicted that the plasma concentration of cetuximab required to inhibit 90% of phospho-EGFR was 67.5 μg/mL. Conclusions: Phospho-EGFR/phospho-MAPK could be useful clinical biomarkers to assess EGFR inhibition by cetuximab.


Pharmaceutical Biology | 2003

New Anticancer Drugs from Cultured and Collected Marine Organisms

William Fenical; Paul R. Jensen; Christopher A. Kauffman; Stephanie Mayhead; D. Faulkner; Catherine Sincich; M. Rao; Eric Kantorowski; Lyndon M. West; Wendy Strangman; Yuzuru Shimizu; Bo Li; Sudhakararao Thammana; Katherine Drainville; Michael T. Davies-Coleman; Robert Kramer; Craig R. Fairchild; William C. Rose; Robert Wild; Gregory D. Vite; Russell Peterson

This paper provides an outline of a collaborative research project between researchers at the University of California, San Diego, University of Rhode Island, and the Bristol-Myers Squibb Pharmaceutical Research Institute, with participating members from the Developmental Therapeutics Branch of the National Cancer Institute. The program, formally funded by the National Cancer Institute under the National Cooperative Drug Discovery Groups (NCDDG) program, seeks to discover new anticancer drugs from marine organisms, in particular invertebrates such as sponges and ascidians, and marine microalgae, marine bacteria and fungi. In this report, the program and results obtained since its beginning in 2000 will be summarized.

Collaboration


Dive into the Robert Kramer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge