Network


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

Hotspot


Dive into the research topics where James B. Rake is active.

Publication


Featured researches published by James B. Rake.


Investigational New Drugs | 1997

Discovery of cryptophycin-1 and BCN-183577 : Examples of strategies and problems in the detection of antitumor activity in mice

Thomas H. Corbett; Frederick A. Valeriote; Lisa Demchik; Nancy Lowichik; Lisa Polin; Chiab Panchapor; Susan Pugh; Kathryn White; Juiwanna Kushner; James B. Rake; Mark P. Wentland; Trimurtulu Golakoti; Carl Hetzel; Junichi Ogino; Gregory M. L. Patterson; Richard E. Moore

Historically, many new anticancer agents were first detected in a prescreen; usually consisting of a molecular/biochemical target or a cellular cytotoxicity assay. The agent then progressed to in vivo evaluation against transplanted human or mouse tumors. If the investigator had a large drug supply and ample resources, multiple tests were possible, with variations in tumor models, tumor and drug routes, dose-decrements, dose-schedules, number of groups, etc. However, in most large programs involving several hundred in vivo tests yearly, resource limitations and drug supply limitations have usually dictated a single trial. Under such restrictive conditions, we have implemented a flexible in vivo testing protocol. With this strategy, the tumor model is dictated by in vitro cellular sensitivity; drug route by water solubility (with water soluble agents injected intravenously); dosage decrement by drug supply, dose-schedule by toxicities encountered, etc. In this flexible design, many treatment parameters can be changed during the course of treatment (e.g., dose and schedule). The discovery of two active agents are presented (Cryptophycin-1, and Thioxanthone BCN 183577). Both were discovered by the intravenous route of administration. Both would have been missed if they were tested intraperitoneally, the usual drug route used in discovery protocols. It is also likely that they would have been missed with an easy to execute fixed protocol design, even if injected IV.


Archive | 1992

Discovery of Solid Tumor Active Agents Using a Soft-Agar-Colony-Formation Disk-Diffusion-Assay

Thomas H. Corbett; Frederick A. Valeriote; Lisa Polin; Chiab Panchapor; Susan Pugh; Kathryn White; Nancy Lowichik; Juiwanna Knight; Marie-Christine Bissery; Antoinette Wozniak; Patricia LoRusso; Laura Biernat; Daniel Polin; Lentawn Knight; Sandra Biggar; Darrell Looney; Lisa Demchik; Julie Jones; Lynne Jones; Scott C. Blair; Kerry Palmer; Sandra Essenmacher; Loretta Lisow; Ken Mattes; Paul F. Cavanaugh; James B. Rake; Laurence H. Baker

The history of antitumor drug discovery has essentially been the use of two lymphocytic leukemias of mice as selection funnels through which all agents needed to pass in order to advance toward clinical development (L1210 prior to 1975 and P388 after 1975). It is thus not surprising that agents in the clinic are highly active against these tumor systems. However, none of the agents discovered by these leukemias are tumor specific (i.e., active against all tumors), and none of the agents are broadly active against solid tumors of either rodents or humans (1, 2, 3). An example contrasting the responsiveness of transplantable solid tumors of mice and the two leukemias is shown in Table-1. The lack of responsiveness of these solid tumors of mice is not unlike those seen in human lung, pancreatic, colon, and prostate tumors. The point to emphasize is that the lack of solid tumor activity of available antitumor agents is not species related. The fault does not lie with the omission of human tumors in the initial selection process, but rather with the omission of solid tumors.


Pharmaceutical Biology | 1995

Tumor Models and the Discovery and Secondary Evaluation of Solid Tumor Active Agents

Thomas H. Corbett; Fred Valeriote; Patricia LoRusso; Lisa Polin; Chiab Panchapor; Susan Pugh; Kathryn White; Juiwanna Knight; Lisa Demchik; Julie Jones; Lynne Jones; Nancy Lowichik; Laura Biernat; Brenda J. Foster; Antoinette J. Wozniak; Loretta Lisow; Manuel Valdivieso; Lawrence H. Baker; Wilbur R. Leopold; Judith Sebolt; Marie Christine Bissery; Ken Mattes; Janet Dzubow; James B. Rake; Robert B. Perni; Mark P. Wentland; Susan A. Coughlin; J Michael Shaw; Gary G Liversidge; Elaine Liversidge

AbstractEach independently arising tumor is a separate and unique biologic entity with its own unique histologic appearance, biologic behavior, and drug response profile. Thus, in drug discovery, no single tumor has been a perfect predictor for any other tumor. For this reason, new agents are evaluated in a variety of tumor models which is known as breadth of activity testing. In recent years, human tumors implanted in athymic nude mice and SCID mice have also become available for breadth of activity testing. In studies carried out in these laboratories, it was found that 10 human tumors metastasized in the SCID mice, but failed to metastasize in nude mice. In addition, tumor growth and tumor takes were superior in the SCID mice. The strengths and weaknesses of xenograft model systems are discussed. For example, most human tumor xenograft models are excessively sensitive to alkylating agents as well as to a new class of DNA binders (XE840 and XP315). Using human tumor models that are the least sensitive t...


Investigational New Drugs | 1999

Preclinical efficacy of thioxanthone SR271425 against transplanted solid tumors of mouse and human origin

Thomas H. Corbett; Chiab Panchapor; Lisa Polin; Nancy Lowichik; Susan Pugh; Kathryn White; Juiwanna Kushner; Jennifer Meyer; Jennifer Czarnecki; Salina Chinnukroh; Matthew Edelstein; Patricia LoRusso; Lance K. Heilbrun; Jerome P. Horwitz; Charles Grieshaber; Robert Perni; Mark P. Wentland; Susan Coughlin; Steven Elenbaas; Richard Philion; James B. Rake

A highly active and broadly active thioxanthone has been identified: N-[[1-[[2-(Diethylamino)ethyl]amino]-7-methoxy-9-oxo-9H-thioxanthen-4-yl] methylformamide (SR271425, BCN326862, WIN71425). In preclinical testing against a variety of subcutaneously growing solid tumors, the following %T/C and Log10 tumor cell kill (LK) values were obtained: Panc-03 T/C = 0, 5/5 cures; Colon-38 (adv. stage) T/C = 0, 3/5 cures, 4.9 LK; Mam-16/C T/C = 0, 3.5 LK; Mam-17/0 T/C = 0, 2.8 LK; Colon-26 T/C = 0, 1/5 cures, 3.2 LK; Colon-51 T/C= 0, 2.7 LK; Panc-02 T/C = 0, 3.1 LK; B16 Melanoma T/C = 13%, 4.0 LK; Squamous Lung-LC12 (adv. stage) T/C = 14%, 4.9 LK; BG-1 human ovarian T/C = 16%, 1.3 LK; WSU-Br1 human breast T/C = 25%, 0.8 LK. The agent was modestly active against doxorubicin (Adr)-resistant solid tumors: Mam-17/Adr T/C =23%, 0.8 LK; and Mam-16/C/Adr T/C = 25%, 1.0 LK, but retained substantial activity against a taxol-resistant tumor: Mam-16/C/taxol T/C = 3%, 2.4 LK. SR271425 was highly active against IV implanted leukemias, L1210 6.3 LK and AML1498 5.3 LK. The agent was equally active both by the IV and oral routes of administration, although requiring approximately 30% higher dose by the oral route. Based on its preclinical antitumor profile, it may be appropriate to evaluate SR271425 in clinical trials.


Bioorganic & Medicinal Chemistry Letters | 1995

The antitumor activity of novel pyrazoloquinoline derivatives

Mark P. Wentland; Suzanne C. Aldous; Monte D. Gruett; Robert B. Perni; Ronald G. Powles; Deborah W. Danz; Kristina M. Klingbeil; A.Danielle Peverly; Ronald G. Robinson; Thomas H. Corbett; James B. Rake; Susan A. Coughlin

Abstract Mammalian topoisomerase II inhibition activity has been identified in a series of novel pyrazoloquinoline derivatives; potency for two analogues containing cyclohexyl groups at the 2-position was comparable to the reference agents, mAMSA and VP-16. In several instances, topo II inhibition translated to a high level of in vitro cytotoxicity and murine antitumor activity.


Biochemical Pharmacology | 1995

Mechanism of action and antitumor activity of (S)-10-(2,6-dimethyl-4-pyridinyl)-9-fluoro-3-methyl-7-oxo-2,3-dihydro-7H-pyridol [1,2,3-de]-[1,4]benzothiazine-6-carboxylic acid (WIN 58161)

Susan A. Coughlin; Deborah W. Danz; Ronald G. Robinson; Kristina M. Klingbeil; Mark P. Wentland; Thomas H. Corbett; William R. Waud; Leonard A. Zwelling; Elizabeth Altschuler; E.S. Bales; James B. Rake

(S)-10-(2,6-Dimethyl-4-pyridinyl)-9-fluoro-3-methyl-7-oxo-2,3-dihydro-7H - pyrido[1,2,3-de][1,4]benzothiazine-6-carboxylic acid (WIN 58161) is an enantiomerically pure quinolone with outstanding bacterial topoisomerase II (DNA gyrase, EC 5.99.1.3) inhibitory and antibacterial activity. Unlike most quinolones, WIN 58161 also exhibits significant inhibitory activity against mammalian topoisomerase II (EC 5.99.1.3). DNA gyrase and topoisomerase II inhibitory activities are enantioselective. Consequently, WIN 58161 and its enantiomer (WIN 58161-2) provide useful tools to probe the contribution of topoisomerase II inhibition to the mechanism of cytotoxicity of quinolones and the potential utility of quinolone-topoisomerase II inhibitors as antitumor agents. WIN 58161 inhibited both highly purified Escherichia coli DNA gyrase and HeLa cell topoisomerase II by the promotion of enzyme-DNA covalent complexes. WIN 58161 did not bind stably to DNA via intercalation and did not enhance the formation of topoisomerase I (EC 5.99.1.2)-DNA covalent complexes. At drug concentrations that are cytotoxic to P388 murine leukemia cells, WIN 58161 promoted intracellular DNA single-strand breaks (SSBs) that exhibited the hallmarks of being mediated by topoisomerase. DNA fragments were complexed with protein, and SSBs were readily resealed at 37 degrees following drug removal. WIN 58161-2 was neither cytotoxic nor did it promote intracellular SSBs in P388. These observations suggest that the mechanism of cytotoxicity of WIN 58161 is predominantly, if not exclusively, a result of topoisomerase II inhibition. When studied in tumor-bearing mice, WIN 58161 exhibited a significant antitumor effect against each of five tumors tested, whereas neither toxicity nor antitumor activity was observed with WIN 58161-2. We conclude from these studies that WIN 58161 represents the prototype of a novel chemical class of topoisomerase II inhibitor with potential clinical utility in treating cancer.


Archive | 2011

Transplantable Syngeneic Rodent Tumors: Solid Tumors in Mice

Lisa Polin; Thomas H. Corbett; Bill J. Roberts; Alfred J. Lawson; Wilbur R. Leopold; Kathryn White; Juiwanna Kushner; Stuart T. Hazeldine; Richard E. Moore; James B. Rake; Jerome P. Horwitz

As preclinical chemotherapists, we are often asked to identify experimental tumor models that can accurately predict for the drug response characteristics of all tumors of a given cellular subtype or molecular target. Unfortunately, it is impossible to give satisfactory answers to these inquiries. Because of the unique character of each independently arising tumor (whether spontaneous or induced), it does not take very long to realize that each tumor is a unique biologic entity with its own tumor growth behavior, histological appearance, drug response and molecular expression profiles. This is true whether the tumor is an experimental animal model or one originally derived from a patient. Further, many factors can influence the tumor growth and therapy response of experimental tumor models. Still, in vivo models are needed to adequately assess pharmacodynamics, toxicity and efficacy of any potential novel therapy. Presented herein is what we hope will be useful information regarding the transplant characteristics of tumor models, with some of the “pitfalls” to look out for when using any given tumor model for chemotherapy evaluations. Although most of the examples given use syngeneic models, the methodologies for assessing the predictive worth and maintaining model usefulness can be applied to almost any given transplantable tumor system (whether syngeneic or xenograft).


Archive | 2002

Transplantable Syngeneic Rodent Tumors

Thomas H. Corbett; Lisa Polin; Bill J. Roberts; Alfred J. Lawson; Wilbur R. Leopold; Kathryn White; Juiwanna Kushner; Jennifer Paluch; Stuart T. Hazeldine; Richard E. Moore; James B. Rake; Jerome P. Horwitz

For many decades, the results from transplantable tumor models have been viewed with considerable skepticism. The perception has long been that these models are excessively sensitive, and not predictive of the human disease. Although we do not intend to debate the many issues involved, it is our view that a better understanding of the transplant properties (e.g., take-rate) of the models—as well as a better understanding of the potential shortcomings in data presentation—will greatly aid the reader in the interpretation of these data. This chapter is an effort to summarize some of the basic operating characteristics of a wide range of solid-tumor models. Since this is a chemotherapy group, we can best explain some of the behavioral characteristics of these models within therapeutic experiments. Most of the data is drawn from the use of transplantable, syngeneic mouse tumors, but a few human tumors have been used for contrast.


Cancer Chemotherapy and Pharmacology | 1999

Evidence of enhanced in vivo activity using tirapazamine with paclitaxel and paraplatin regimens against the MV-522 human lung cancer xenograft

Steven Weitman; Gina Mangold; Jennifer Marty; Daniel L. Dexter; Susan G. Hilsenbeck; James B. Rake; Paul E. Juniewicz; Daniel D. Von Hoff

Purpose: Tirapazamine (3-amino-1,2,4-benzotriazine 1,4-dioxide; SR 4233) is a bioreductive agent that exhibits relatively selective cytotoxicity towards cells under hypoxic conditions and can enhance the antitumor activity of many standard oncolytics. In the present study we examined the interaction between tirapazamine in vivo with paclitaxel and paraplatin in two- and three-way combination studies using the MV-522 human lung carcinoma xenograft model. Methods: Agents were administered as a single i.p. bolus, with tirapazamine being given 3 h prior to paclitaxel, paraplatin, or their combination. Tumor growth inhibition (TGI), final tumor weights, partial and complete responses, and time to tumor doubling were determined after drug administration. Results: Tirapazamine as a single agent was ineffective against this human lung tumor model. A substantial increase in TGI was seen in animals treated with the triple-agent regimen (tirapazamine-paclitaxel-paraplatin) compared to animals treated with double-agent regimens that did not include tirapazamine. The addition of tirapazamine to paclitaxel-paraplatin therapy resulted in a 50% complete response rate; there were no complete responses seen when only the paclitaxel-paraplatin combination was administered. Time to tumor doubling was also significantly improved with the addition of tirapazamine to the paclitaxel and paraplatin combinations. Tirapazamine did not increase the toxicity of paclitaxel, paraplatin, or their combinations as judged by its minimal impact on body weight and the fact that no toxic deaths were observed with tirapazamine-containing regimens. Conclusions: These results are important since recent studies have suggested that the combination of paclitaxel and paraplatin may be particularly active in patients with advanced stage non-small-cell lung cancer. Since tirapazamine can significantly improve efficacy, but does not appear to enhance the toxicity of paclitaxel and paraplatin, its evaluation in future clinical trials in combination with paclitaxel-paraplatin-based therapy appears warranted.


Journal of Chromatography B: Biomedical Sciences and Applications | 2001

High-performance liquid chromatographic method for the estimation of the novel investigational anti-cancer agent SR271425 and its metabolites in mouse plasma

Srinivasu Poondru; Sen-Lin Zhou; James B. Rake; Gareth Shackleton; Thomas H. Corbett; Ralph E. Parchment; Bhaskara R. Jasti

A simple and reliable HPLC method was developed for the estimation of a new anti-cancer agent that belongs to the thioxanthone class, SR271425 in mouse plasma. SR271425, its metabolites and internal standard (SR233377) were separated from plasma by liquid-liquid extraction using dichloromethane after quenching the plasma proteins with acetonitrile. Chromatography was performed on a reversed-phase C18 column using methanol-10 mM phosphate buffer, pH 3.5 (45:55) as mobile phase at a flow-rate of 0.8 ml/min for first 10 min and 1.4 ml/min for the next 15 min with UV-Vis detection at 264 nm and SR233377 as internal standard. The retention times of SR271425 and internal standard were 18.6 and 14.8 min, respectively. The limit of detection was 40 ng/ml and the limit of quantification was 78 ng/ml. This method was also able to detect the three metabolites of SR271425. The intra- and inter-day relative standard deviations were less than 13% at all concentrations. This analytical method was precise and reproducible for pharmacokinetics and metabolism studies of the drug in mice. SR271425 is proceeding to phase I clinical trials in 2001.

Collaboration


Dive into the James B. Rake's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark P. Wentland

Rensselaer Polytechnic Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lisa Polin

Wayne State University

View shared research outputs
Top Co-Authors

Avatar

Robert B. Perni

Rensselaer Polytechnic Institute

View shared research outputs
Top Co-Authors

Avatar

Patricia LoRusso

Harper University Hospital

View shared research outputs
Top Co-Authors

Avatar

Susan Pugh

Wayne State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

George Y. Lesher

Rensselaer Polytechnic Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge