33 results on '"Joy Bauch"'
Search Results
2. Data from ABT-263 and rapamycin act cooperatively to kill lymphoma cells in vitro and in vivo
- Author
-
Alex R. Shoemaker, Steven W. Elmore, Saul H. Rosenberg, Stephen W. Fesik, David J. Frost, Christin Tse, Joy Bauch, Sanjay R. Chemburkar, Baole Wang, Sally Schlessinger, Marion Refici, Anatol Oleksijew, Kelly Foster, Michael J. Mitten, Yu Xiao, and Scott Ackler
- Abstract
ABT-263 is a potent, orally bioavailable inhibitor of the antiapoptotic Bcl-2 family members Bcl-2, Bcl-xL, and Bcl-w, which is currently in phase I clinical trials. Previous work has shown that this compound has low nanomolar cell-killing activity in a variety of lymphoma and leukemia cell lines, many of which overexpress Bcl-2 through a variety of mechanisms. Rapamycin is a macrolide antibiotic that inhibits the mammalian target of rapamycin complex, leading to cell cycle arrest and inhibition of protein translation. Rapamycin (and its analogues) has shown activity in a variety of tumor cell lines primarily through induction of cell cycle arrest. Activity has also been shown clinically in mantle cell lymphoma and advanced renal cell carcinoma. Here, we show that treatment of the follicular lymphoma lines DoHH-2 and SuDHL-4 with 100 nmol/L rapamycin induces substantial G0-G1 arrest. Addition of as little as 39 nmol/L ABT-263 to the rapamycin regimen induced a 3-fold increase in sub-G0 cells. Combination of these agents also led to a significant increase in Annexin V staining over ABT-263 alone. In xenograft models of these tumors, rapamycin induced a largely cytostatic response in the DoHH-2 and SuDHL-4 models. Coadministration with ABT-263 induced significant tumor regression, with DoHH-2 and SuDHL-4 tumors showing 100% overall response rates. Apoptosis in these tumors was significantly enhanced by combination therapy as measured by staining with an antibody specific for cleaved caspase-3. These data suggest that combination of ABT-263 and rapamycin or its analogues represents a promising therapeutic strategy for the treatment of lymphoma. [Mol Cancer Ther 2008;7(10):3265–74]
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Table 1 from A Small-Molecule Inhibitor of Bcl-XL Potentiates the Activity of Cytotoxic Drugs In vitro and In vivo
- Author
-
Steven W. Elmore, Saul H. Rosenberg, Stephen W. Fesik, Haichao Zhang, Robert Warner, Baole Wang, Stephen K. Tahir, Jason Stavropoulos, Wang Shen, Weiguo Qing, Tilman Oltersdorf, Jacqueline M. O'Connor, Paul Nimmer, ShiChung Ng, Hugh Nellans, William McClellan, Kennan Marsh, Mary K. Joseph, Ken Jarvis, David J. Frost, Thomas Deckwirth, Milan Bruncko, Tony Borre, Barbara A. Belli, Joy Bauch, Anatol Oleksijew, and Alex R. Shoemaker
- Abstract
Supplementary Table 1 from A Small-Molecule Inhibitor of Bcl-XL Potentiates the Activity of Cytotoxic Drugs In vitro and In vivo
- Published
- 2023
- Full Text
- View/download PDF
4. Data from A Small-Molecule Inhibitor of Bcl-XL Potentiates the Activity of Cytotoxic Drugs In vitro and In vivo
- Author
-
Steven W. Elmore, Saul H. Rosenberg, Stephen W. Fesik, Haichao Zhang, Robert Warner, Baole Wang, Stephen K. Tahir, Jason Stavropoulos, Wang Shen, Weiguo Qing, Tilman Oltersdorf, Jacqueline M. O'Connor, Paul Nimmer, ShiChung Ng, Hugh Nellans, William McClellan, Kennan Marsh, Mary K. Joseph, Ken Jarvis, David J. Frost, Thomas Deckwirth, Milan Bruncko, Tony Borre, Barbara A. Belli, Joy Bauch, Anatol Oleksijew, and Alex R. Shoemaker
- Abstract
Inhibition of the prosurvival members of the Bcl-2 family of proteins represents an attractive strategy for the treatment of cancer. We have previously reported the activity of ABT-737, a potent inhibitor of Bcl-2, Bcl-XL, and Bcl-w, which exhibits monotherapy efficacy in xenograft models of small-cell lung cancer and lymphoma and potentiates the activity of numerous cytotoxic agents. Here we describe the biological activity of A-385358, a small molecule with relative selectivity for binding to Bcl-XL versus Bcl-2 (Ki's of 0.80 and 67 nmol/L for Bcl-XL and Bcl-2, respectively). This compound efficiently enters cells and co-localizes with the mitochondrial membrane. Although A-385358 shows relatively modest single-agent cytotoxic activity against most tumor cell lines, it has an EC50 of L for survival. In addition, A-385358 enhances the in vitro cytotoxic activity of numerous chemotherapeutic agents (paclitaxel, etoposide, cisplatin, and doxorubicin) in several tumor cell lines. In A549 non–small-cell lung cancer cells, A-385358 potentiates the activity of paclitaxel by as much as 25-fold. Importantly, A-385358 also potentiated the activity of paclitaxel in vivo. Significant inhibition of tumor growth was observed when A-385358 was added to maximally tolerated or half maximally tolerated doses of paclitaxel in the A549 xenograft model. In tumors, the combination therapy also resulted in a significant increase in mitotic arrest followed by apoptosis relative to paclitaxel monotherapy. (Cancer Res 2006; 66(17): 8731-9)
- Published
- 2023
- Full Text
- View/download PDF
5. ABT-263 and rapamycin act cooperatively to kill lymphoma cells in vitro and in vivo
- Author
-
Scott L. Ackler, Saul H. Rosenberg, Joy Bauch, Sally Schlessinger, Marion Refici, Kelly Foster, Christin Tse, Sanjay R. Chemburkar, Anatol Oleksijew, David Frost, Michael J. Mitten, Baole Wang, Alex R. Shoemaker, Stephen W. Fesik, Steven W. Elmore, and Yu Xiao
- Subjects
Cancer Research ,Programmed cell death ,Cell cycle checkpoint ,Follicular lymphoma ,Antineoplastic Agents ,Mice, SCID ,Biology ,Pharmacology ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Sirolimus ,Sulfonamides ,Aniline Compounds ,Cell Death ,Cell Cycle ,Remission Induction ,Drug Synergism ,Cell cycle ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Lymphoma ,Oncology ,Apoptosis ,Mantle cell lymphoma ,Lymphoma, Large B-Cell, Diffuse ,medicine.drug - Abstract
ABT-263 is a potent, orally bioavailable inhibitor of the antiapoptotic Bcl-2 family members Bcl-2, Bcl-xL, and Bcl-w, which is currently in phase I clinical trials. Previous work has shown that this compound has low nanomolar cell-killing activity in a variety of lymphoma and leukemia cell lines, many of which overexpress Bcl-2 through a variety of mechanisms. Rapamycin is a macrolide antibiotic that inhibits the mammalian target of rapamycin complex, leading to cell cycle arrest and inhibition of protein translation. Rapamycin (and its analogues) has shown activity in a variety of tumor cell lines primarily through induction of cell cycle arrest. Activity has also been shown clinically in mantle cell lymphoma and advanced renal cell carcinoma. Here, we show that treatment of the follicular lymphoma lines DoHH-2 and SuDHL-4 with 100 nmol/L rapamycin induces substantial G0-G1 arrest. Addition of as little as 39 nmol/L ABT-263 to the rapamycin regimen induced a 3-fold increase in sub-G0 cells. Combination of these agents also led to a significant increase in Annexin V staining over ABT-263 alone. In xenograft models of these tumors, rapamycin induced a largely cytostatic response in the DoHH-2 and SuDHL-4 models. Coadministration with ABT-263 induced significant tumor regression, with DoHH-2 and SuDHL-4 tumors showing 100% overall response rates. Apoptosis in these tumors was significantly enhanced by combination therapy as measured by staining with an antibody specific for cleaved caspase-3. These data suggest that combination of ABT-263 and rapamycin or its analogues represents a promising therapeutic strategy for the treatment of lymphoma. [Mol Cancer Ther 2008;7(10):3265–74]
- Published
- 2008
- Full Text
- View/download PDF
6. Activity of the Bcl-2 Family Inhibitor ABT-263 in a Panel of Small Cell Lung Cancer Xenograft Models
- Author
-
Baole Wang, Joy Bauch, David Frost, Christin Tse, Steven K. Tahir, Kennan C. Marsh, Xiufen Yang, Anatol Oleksijew, Jacqueline M. O'Connor, Debra Ferguson, Michael J. Mitten, Stephen W. Fesik, Scott L. Ackler, Saul H. Rosenberg, Jessica Adickes, Alex R. Shoemaker, Steven W. Elmore, and Marion Refici
- Subjects
Cancer Research ,Lung Neoplasms ,Mice, Nude ,Antineoplastic Agents ,Mice ,chemistry.chemical_compound ,medicine ,Carcinoma ,Animals ,Humans ,Carcinoma, Small Cell ,Lung cancer ,Sulfonamides ,Aniline Compounds ,Navitoclax ,Dose-Response Relationship, Drug ,business.industry ,Bcl-2 family ,Cancer ,Biological activity ,medicine.disease ,Xenograft Model Antitumor Assays ,Clinical trial ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,chemistry ,Apoptosis ,Immunology ,Cancer research ,business - Abstract
Purpose: The purpose of this study was to characterize the activity of the Bcl-2 protein family inhibitor ABT-263 in a panel of small cell lung cancer (SCLC) xenograft models. Experimental Design: A panel of 11 SCLC xenograft models was established to evaluate the efficacy of ABT-263. Single agent activity was examined on a continuous dosing schedule in each of these models. The H146 model was used to further evaluate dose and schedule, comparison to standard cytotoxic agents, and induction of apoptosis. Results: ABT-263 exhibited a range of antitumor activity, leading to complete tumor regression in several models. Significant regressions of tumors as large as 1 cc were also observed. The efficacy of ABT-263 was also quite durable; in several cases, minimal tumor regrowth was noted several weeks after the cessation of treatment. Antitumor effects were equal or superior to that of several clinically approved cytotoxic agents. Regression of large established tumors was observed through several cycles of therapy and efficacy was retained in a Pgp-1 overexpressing line. Significant efficacy was observed on several dose and therapeutic schedules and was associated with significant induction of apoptosis. Conclusions: ABT-263 is a potent, orally bioavailable inhibitor of Bcl-2 family proteins that has recently entered clinical trials. The efficacy data reported here suggest that SCLC is a promising area of clinical investigation with this agent.
- Published
- 2008
- Full Text
- View/download PDF
7. Zotarolimus, a Novel Sirolimus Analogue With Potent Anti-proliferative Activity on Coronary Smooth Muscle Cells and Reduced Potential for Systemic Immunosuppression
- Author
-
Stevan W. Djuric, Kennan C. Marsh, Gin C. Hsieh, Michael P. Sheets, Yung-Wu Chen, Thomas A. Fey, Cindy Henry, Karl W. Mollison, Rolf Wagner, Donna M. Gauvin, George W. Carter, James M. Trevillyan, Teresa A. Rosenberg, Sandra E. Burke, Steve J. Ballaron, Joy Bauch, and Morey L. Smith
- Subjects
Graft Rejection ,Male ,Encephalomyelitis, Autoimmune, Experimental ,T-Lymphocytes ,medicine.medical_treatment ,Myocytes, Smooth Muscle ,Tacrolimus Binding Protein 1A ,Pharmacology ,Binding, Competitive ,Drug Hypersensitivity ,Rats, Sprague-Dawley ,Inhibitory Concentration 50 ,Restenosis ,In vivo ,Rats, Inbred BN ,Angioplasty ,Animals ,Humans ,Medicine ,Hypersensitivity, Delayed ,Zotarolimus ,cardiovascular diseases ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Sirolimus ,Dose-Response Relationship, Drug ,biology ,business.industry ,equipment and supplies ,medicine.disease ,Coronary Vessels ,Rats ,Disease Models, Animal ,surgical procedures, operative ,Animals, Newborn ,Rats, Inbred Lew ,Concanavalin A ,Allograft rejection ,cardiovascular system ,biology.protein ,Heart Transplantation ,Lymphocyte Culture Test, Mixed ,Cardiology and Cardiovascular Medicine ,business ,Immunosuppressive Agents ,Half-Life ,medicine.drug - Abstract
Sirolimus (rapamycin) is an immunosuppressant used in preventing allograft rejection and in drug-eluting stents to prevent restenosis after angioplasty. Zotarolimus, an analogue of sirolimus, was designed to have a shorter in vivo half-life. Zotarolimus was found to be mechanistically similar to sirolimus in having high-affinity binding to the immunophilin FKBP12 and comparable potency for inhibiting in vitro proliferation of both human and rat T cells. Rat pharmacokinetic studies with intravenous dosing demonstrated terminal elimination half-lives of 9.4 hours and 14.0 hours for zotarolimus and sirolimus, respectively. Given orally, T1/2 values were 7.9 hours and 33.4 hours, respectively. Consistent with its shorter duration, zotarolimus showed a corresponding and statistically significant 4-fold reduction in potency for systemic immunosuppression in 3 rat disease models. Pharmacokinetic studies in cynomolgus monkey underpredicted the half-life difference between zotarolimus and sirolimus apparent from recent clinical data. In vitro inhibition of human coronary artery smooth muscle cell proliferation by zotarolimus was comparable to sirolimus. Drug-eluting stents for local delivery of zotarolimus to the vessel wall of coronary arteries are in clinical development. The pharmacological profile of zotarolimus suggests it may be advantageous for preventing restenosis with a reduced potential for causing systemic immunosuppression or other side effects.
- Published
- 2007
- Full Text
- View/download PDF
8. A farnesyltransferase inhibitor improves disease phenotypes in mice with a Hutchinson-Gilford progeria syndrome mutation
- Author
-
Margarita Meta, David Frost, Shao H. Yang, Joy Bauch, Sharmila Majumdar, Catherine Coffinier, Loren G. Fong, Xin Qiao, Martin O. Bergo, and Stephen G. Young
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Osteoporosis ,Adipose tissue ,Biology ,medicine.disease_cause ,Bone and Bones ,Mice ,Progeria ,medicine ,Animals ,Farnesyltranstransferase ,Enzyme Inhibitors ,Genetics ,Mutation ,integumentary system ,Farnesyltransferase inhibitor ,nutritional and metabolic diseases ,General Medicine ,medicine.disease ,Progerin ,Phenotype ,Radiography ,Disease Models, Animal ,Cancer research ,Bone Diseases ,Restrictive dermopathy ,Research Article - Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is caused by the production of a truncated prelamin A, called progerin, which is farnesylated at its carboxyl terminus. Progerin is targeted to the nuclear envelope and causes misshapen nuclei. Protein farnesyltransferase inhibitors (FTI) mislocalize progerin away from the nuclear envelope and reduce the frequency of misshapen nuclei. To determine whether an FTI would ameliorate disease phenotypes in vivo, we created gene-targeted mice with an HGPS mutation (LmnaHG/+) and then examined the effect of an FTI on disease phenotypes. LmnaHG/+ mice exhibited phenotypes similar to those in human HGPS patients, including retarded growth, reduced amounts of adipose tissue, micrognathia, osteoporosis, and osteolytic lesions in bone. Osteolytic lesions in the ribs led to spontaneous bone fractures. Treatment with an FTI increased adipose tissue mass, improved body weight curves, reduced the number of rib fractures, and improved bone mineralization and bone cortical thickness. These studies suggest that FTIs could be useful for treating humans with HGPS.
- Published
- 2006
- Full Text
- View/download PDF
9. Thienopyrimidine Ureas as Novel and Potent Multitargeted Receptor Tyrosine Kinase Inhibitors
- Author
-
Teresa Barlozzari, Shannon S Arries, Keith B. Glaser, Michael L. Curtin, Lori J. Pease, Patrick A. Marcotte, Robin R. Frey, Daniel H. Albert, Neil Wishart, George A. Cunha, Yan Guo, Jennifer J. Bouska, Lee D. Arnold, Jun Guo, Michael R. Michaelides, Kennan C. Marsh, Steven K. Davidsen, Joy Bauch, Asma A Ahmed, Peter F. Bousquet, Maria D Moskey, Paul Tapang, Kent D. Stewart, Yujia Dai, Zhiqin Ji, Vincent S. Stoll, and Junling Li
- Subjects
Models, Molecular ,Platelet-derived growth factor ,Antineoplastic Agents ,Mice, SCID ,Receptor tyrosine kinase ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Adenosine Triphosphate ,Growth factor receptor ,Drug Discovery ,Animals ,Edema ,Humans ,Urea ,Receptors, Platelet-Derived Growth Factor ,Phosphorylation ,Mice, Inbred BALB C ,Estradiol ,biology ,Uterus ,Vascular Endothelial Growth Factor Receptor-2 ,Xenograft Model Antitumor Assays ,Vascular endothelial growth factor ,Pyrimidines ,chemistry ,Biochemistry ,Enzyme inhibitor ,NIH 3T3 Cells ,biology.protein ,Molecular Medicine ,Female ,Signal transduction ,Platelet-derived growth factor receptor - Abstract
A series of novel thienopyrimidine-based receptor tyrosine kinase inhibitors has been discovered. Investigation of structure-activity relationships at the 5- and 6-positions of the thienopyrimidine nucleus led to a series of N,N'-diaryl ureas that potently inhibit all of the vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptor tyrosine kinases. A kinase insert domain-containing receptor (KDR) homology model suggests that these compounds bind to the "inactive conformation" of the enzyme with the urea portion extending into the back hydrophobic pocket adjacent to the adenosine 5'-triphosphate (ATP) binding site. A number of compounds have been identified as displaying excellent in vivo potency. In particular, compounds 28 and 76 possess favorable pharmacokinetic (PK) profiles and demonstrate potent antitumor efficacy against the HT1080 human fibrosarcoma xenograft tumor growth model (tumor growth inhibition (TGI) = 75% at 25 mg/kg.day, per os (po)).
- Published
- 2005
- Full Text
- View/download PDF
10. Antitumor Activity of Orally Bioavailable Farnesyltransferase Inhibitor, ABT-100, Is Mediated by Antiproliferative, Proapoptotic, and Antiangiogenic Effects in Xenograft Models
- Author
-
Joy Bauch, Gerard M. Sullivan, Luis E. Rodriguez, Ingrid B.J.K. Joseph, Kennan C. Marsh, Jacqueline M. O'Connor, David Frost, Joann P. Palma, Nan-Horng Lin, Kenneth Jarvis, Marion Refici, Saul H. Rosenberg, Haiying Zhang, Debra Ferguson, and Hing L. Sham
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Cancer Research ,Angiogenesis ,Farnesyltransferase ,Basic fibroblast growth factor ,Administration, Oral ,Biological Availability ,Mice, Nude ,Angiogenesis Inhibitors ,Antineoplastic Agents ,Apoptosis ,Mice, SCID ,Biology ,Pharmacology ,Mice ,chemistry.chemical_compound ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Farnesyltranstransferase ,Humans ,Enzyme Inhibitors ,Clonogenic assay ,Cell Proliferation ,Alkyl and Aryl Transferases ,Dose-Response Relationship, Drug ,Neovascularization, Pathologic ,Interleukin-8 ,Farnesyltransferase inhibitor ,Imidazoles ,Xenograft Model Antitumor Assays ,Vascular endothelial growth factor ,Oncology ,chemistry ,Area Under Curve ,biology.protein ,Fibroblast Growth Factor 2 ,Tipifarnib ,medicine.drug - Abstract
Purpose: To evaluate the preclinical pharmacokinetics, antitumor efficacy, and mechanism of action of a novel orally active farnesyltransferase inhibitor, ABT-100. Experimental Design: In vitro sensitivity of a panel of human cell lines was determined using proliferation and clonogenic assays. In vivo efficacy of ABT-100 was evaluated in xenograft models (flank or orthotopic) by assessing angiogenesis, proliferation, and apoptosis in correlation with pharmacokinetics. Efficacy of the racemate of ABT-100 (A-367074) was also compared with R115777 (tipifarnib). Results: ABT-100 inhibited proliferation of cells in vitro carrying oncogenic H-Ras (EJ-1 bladder; IC50 2.2 nmol/L), Ki-Ras (DLD-1 colon, MDA-MB-231 breast, HCT-116 colon, and MiaPaCa-2 pancreatic; IC50 range, 3.8-9.2 nmol/L), and wild-type Ras (PC-3 and DU-145; IC50, 70 and 818 nmol/L, respectively) as well as clonogenic potential. ABT-100 shows 70% to 80% oral bioavailability in mice. ABT-100 regressed EJ-1 tumors (2-12.5 mg/kg/d s.c., every day for 21 days) and showed significant efficacy in DLD-1, LX-1, MiaPaCa-2, or PC-3 tumor-bearing mice (6.25-50 mg/kg/d s.c. once daily or twice daily orally). A-367074 showed equivalent efficacy to R115777 given at approximately one-fourth the total dose of R115777 for a shorter duration (EJ-1 and LX-1). Antitumor activity was associated with decreased cell proliferation (Ki-67), increased apoptosis (terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling), and decreased angiogenesis. A reduction in tumor angiogenic cytokine levels (vascular endothelial growth factor, basic fibroblast growth factor, and interleukin-8) correlated with a reduction in tumor vascularity (CD31). Conclusions: Overall, ABT-100 has an acceptable pharmacokinetic profile, is well tolerated, and possesses broad-spectrum antitumor activity against a series of xenograft models similar to farnesyltransferase inhibitors in clinical development; therefore, it is an attractive candidate for clinical evaluation.
- Published
- 2005
- Full Text
- View/download PDF
11. Synthesis of 1H-pyridin-2-one derivatives as potent and selective farnesyltransferase inhibitors
- Author
-
Kennan C. Marsh, Jerome Cohen, Nan-Horng Lin, Joy Bauch, Wen-Zhen Gu, Qun Li, Haiying Zhang, Rodger F. Henry, Le Wang, Saul H. Rosenberg, and Hing L. Sham
- Subjects
Cellular activity ,Pyridines ,Stereochemistry ,Farnesyltransferase ,Clinical Biochemistry ,Administration, Oral ,Pharmaceutical Science ,Crystallography, X-Ray ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Dogs ,Drug Discovery ,Animals ,Farnesyltranstransferase ,Molecular Biology ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Organic Chemistry ,Bioavailability ,Enzyme ,chemistry ,Enzyme inhibitor ,biology.protein ,Lactam ,Molecular Medicine - Abstract
Two novel series of potent and selective FTase inhibitors have been synthesized using structure-based design. Medicinal chemistry efforts led to the discovery of compound 4e with potent cellular activity and good oral bioavailability in dog. A synthetic route toward novel heterocycles 1,5-dimethyl-6-oxo-4-aryl-1,6-dihydro-pyridine-2-carbonitrile was established. The structure of compound 5c was confirmed by X-ray crystallography.
- Published
- 2004
- Full Text
- View/download PDF
12. Discovery of potent imidazole and cyanophenyl containing farnesyltransferase inhibitors with improved oral bioavailability
- Author
-
Hing L. Sham, Lisa A. Hasvold, Jerry Cohen, Weibo Wang, Yunsong Tong, Wen-Zhen Gu, Qun Li, Joy Bauch, Saul H. Rosenberg, Tongmei Li, Nicholas M Leonard, Le Wang, Vincent S. Stoll, Haiying Zhang, Nan-Horng Lin, and Kennan C. Marsh
- Subjects
Models, Molecular ,Stereochemistry ,Farnesyltransferase ,Clinical Biochemistry ,Administration, Oral ,Biological Availability ,Pharmaceutical Science ,Crystallography, X-Ray ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Dogs ,Nitriles ,Drug Discovery ,Animals ,Farnesyltranstransferase ,Imidazole ,Moiety ,heterocyclic compounds ,Enzyme Inhibitors ,Molecular Biology ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,Bicyclic molecule ,biology ,organic chemicals ,Organic Chemistry ,Imidazoles ,Stereoisomerism ,Haplorhini ,Bioavailability ,chemistry ,Enzyme inhibitor ,Drug Design ,biology.protein ,Molecular Medicine - Abstract
A pyridyl moiety was introduced into a previously developed series of farnesyltransferase inhibitors containing imidazole and cyanophenyl (such as 4), resulting in potent inhibitors with improved pharmacokinetics.
- Published
- 2003
- Full Text
- View/download PDF
13. Aryl tetrahydropyridine inhibitors of farnesyltransferase: glycine, phenylalanine and histidine derivatives
- Author
-
Shi-Chung Ng, Gerard M. Sullivan, Weibo Wang, Hovis M. Imade, Stephen J. O'connor, Haiying Zhang, Hing L. Sham, Stephen L. Gwaltney, Saul H. Rosenberg, Lisa A. Hasvold, Jerome Cohen, Wen-Zhen Gu, Joy Bauch, Qun Li, Stephen K. Tahir, Steve Muchmore, Charles W. Hutchins, Kennan C. Marsh, Nelson Lissa T, Clarissa G. Jakob, Vincent S. Stoll, and David Frost
- Subjects
Models, Molecular ,Pyridines ,Stereochemistry ,Phenylalanine ,Farnesyltransferase ,Clinical Biochemistry ,Glycine ,Molecular Conformation ,Biological Availability ,Pharmaceutical Science ,Crystallography, X-Ray ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Farnesyltranstransferase ,Histidine ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Aryl ,Organic Chemistry ,Genes, ras ,Enzyme ,chemistry ,biology.protein ,Molecular Medicine - Abstract
Inhibitors of farnesyltransferase are effective against a variety of tumors in mouse models of cancer. Clinical trials to evaluate these agents in humans are ongoing. In our effort to develop new farnesyltransferase inhibitors, we have discovered a series of aryl tetrahydropyridines that incorporate substituted glycine, phenylalanine and histidine residues. The design, synthesis, SAR and biological properties of these compounds will be discussed.
- Published
- 2003
- Full Text
- View/download PDF
14. New Antimitotic Agents with Activity in Multi-Drug-Resistant Cell Lines and in Vivo Efficacy in Murine Tumor Models
- Author
-
Akiyo Claiborne, Gang Liu, David Frost, Ken Jarvis, Jeffery D. Alder, Thomas W. von Geldern, and Qun Li, Joy Bauch, Laura Gehrke, William J. Chiou, Gunawardana Indrani W, Saul H. Rosenberg, Andrew Tasker, Jae Hwan-Soo, Bruce G. Szczepankiewicz, R. Bruce Credo, Yu Hua Hui, Michael A. Nukkala, Nicolette A. Zielinski, J. Ruth Wu-Wong, Stephen L. Gwaltney, and Karl W. Mollison
- Subjects
Male ,Magnetic Resonance Spectroscopy ,Transplantation, Heterologous ,Antineoplastic Agents ,Pharmacology ,Mass Spectrometry ,Mice ,Structure-Activity Relationship ,Pharmacokinetics ,In vivo ,Drug Discovery ,Tumor Cells, Cultured ,Animals ,Cytotoxic T cell ,Structure–activity relationship ,Oxazoles ,Chromatography, High Pressure Liquid ,Chemistry ,Cell growth ,Drug Resistance, Multiple ,In vitro ,Mice, Inbred C57BL ,Biochemistry ,Drug Resistance, Neoplasm ,Mice, Inbred DBA ,Cell culture ,Molecular Medicine ,Female ,Antimitotic Agent ,Drug Screening Assays, Antitumor ,Colchicine - Abstract
During a screen for compounds that could inhibit cell proliferation, a series of new tubulin-binding compounds was identified with the discovery of oxadiazoline 1 (A-105972). This compound showed good cytotoxic activity against non-multi-drug-resistant and multi-drug-resistant cancer cell lines, but its utility in vivo was limited by a short half-life. Medicinal chemistry efforts led to the discovery of indolyloxazoline 22g (A-259745), which maintained all of the in vitro activity seen with oxadiazoline 1, but also demonstrated a better pharmacokinetic profile, and dose-dependent in vivo activity. Over a 28 day study, indolyloxazoline 22g increased the life span of tumor-implanted mice by up to a factor of 3 upon oral dosing. This compound, and others of its structural class, may prove to be useful in the development of new chemotherapeutic agents to treat human cancers.
- Published
- 2001
- Full Text
- View/download PDF
15. Second-Generation Peptidomimetic Inhibitors of Protein Farnesyltransferase Demonstrating Improved Cellular Potency and Significant in Vivo Efficacy
- Author
-
Said M. Sebti, Juan F. M. Leal, Robert B. Warner, Patricia Ewing, Sajeev Cherian, Le Wang, Stephen Tahir, Edward Devine, Jerome Cohen, Kenneth J. Barr, Hing L. Vernon Hills Sham, Shi-Chung Ng, Haichao Zhang, Michael Mitten, Badr Saeed, Bryan K. Sorensen, Daniel J. Hoffman, Saul H. Rosenberg, Jeffrey Alder, Kennan Marsh, Peter Kovar, Jang Yun Lee, A. S. Tasker, Stephen J. O'Connor, and Joy Bauch
- Subjects
Benzylamines ,Peptidomimetic ,Farnesyltransferase ,Mice, Nude ,Antineoplastic Agents ,Receptors, Tumor Necrosis Factor ,Mice ,In vivo ,Drug Discovery ,Tumor Cells, Cultured ,Animals ,Humans ,Potency ,fas Receptor ,Enzyme Inhibitors ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Cellular Assay ,Neuropeptides ,Enzyme ,chemistry ,Biochemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Ethers ,Signal Transduction - Abstract
The synthesis and evaluation of analogues of previously reported farnesyltransferase inhibitors, pyridyl benzyl ether 3 and pyridylbenzylamine 4, are described. Substitution of 3 at the 5-position of the core aryl ring resulted in inhibitors of equal or less potency against the enzyme and decreased efficacy in a cellular assay against Ras processing by the enzyme. Substitution of 4 at the benzyl nitrogen yielded 26, which showed improved efficacy and potency and yet presented a poor pharmacokinetic profile. Further modification afforded 30, which demonstrated a dramatically improved pharmacokinetic profile. Compounds 26 and 29 demonstrated significant in vivo efficacy in nude mice inoculated with MiaPaCa-2, a human pancreatic tumor-derived cell line.
- Published
- 1999
- Full Text
- View/download PDF
16. Ex VivoAssessment of Immunosuppression in Undiluted Whole Blood from Pigs Dosed with Tacrolimus (FK506)
- Author
-
Melissa S. Pong, Michael P. Sheets, Thomas A. Fey, James M. Trevillyan, Morey L. Smith, Karl W. Mollison, Jay R. Luly, Joy Bauch, Stevan W. Djuric, Stephen J. Ballaron, Yung-Wu Chen, Kennan C. Marsh, Gin C. Hsieh, Robin M Kolano, Donna M. Gauvin, and George W. Carter
- Subjects
Swine ,medicine.medical_treatment ,Immunology ,Pharmacology ,Biology ,Lymphocyte Activation ,Peripheral blood mononuclear cell ,Tacrolimus ,chemistry.chemical_compound ,medicine ,Animals ,Immunology and Allergy ,Potency ,Lymphocytes ,Dosing ,Whole blood ,Ionomycin ,Immunosuppression ,Blood proteins ,chemistry ,Leukocytes, Mononuclear ,Interleukin-2 ,Tetradecanoylphorbol Acetate ,Immunosuppressive Agents ,Ex vivo - Abstract
To assess the duration of immunosuppression in FK506-dosed pigs, an undiluted whole blood assay was established to measure reactivities of T cells in their physiological milieu. PMA and ionomycin were shown to induce IL-2 production in swine blood. The IC50of FK506 in inhibiting IL-2 production in whole blood and isolated PBMC stimulated with PMA and ionomycin measured 1.2 and 0.04 nM, respectively. These data underscore the influence of red blood cells and plasma proteins on drug potency. IL-2 levels were determined in blood drawn immediately before and 1, 24, 48, and 72 h after iv dosing. For pigs dosed with 0.05 mg/kg, 50% recovery of IL-2 production was observed at 16 h and 100% at 35 h after dosing. For pigs dosed with 0.15 mg/kg, 50% recovery was observed at 38 h and 100% at 72 h. Blood concentrations of FK506 at 50 and 100% recovery of IL-2 production measured 10.8 and 2.2 nM for pigs dosed with 0.05 mg/kg and 6.1 and 1.1 nM for pigs dosed with 0.15 mg/kg, respectively. These concentrations are severalfold higher than predicted from the IC50of FK506 for inhibiting IL-2 production in the whole blood assay. These data suggest that the true potency of FK506 in blood after dosing is influenced by additional factors, which could include plasma protein binding, the presence of active or interfering metabolites, serum interfering factors, and sequestration of drug in blood cells. Our results demonstrate the utility of an undiluted whole blood assay for assessing the duration of immunosuppression in drug-dosed animals and emphasize the importance of assessing drug potency in the whole blood environmentex vivo.
- Published
- 1999
- Full Text
- View/download PDF
17. Plasma and cerebrospinal fluid pharmacokinetics of ABT-888 after oral administration in non-human primates
- Author
-
Robert C. Dauser, Susan M. Blaney, Leticia McGuffey, Jed G. Nuchtern, Vincent L. Giranda, Terzah M. Horton, Patrick A. Thompson, Brian D. Dayton, Jodi A. Muscal, Jack Su, Brian W. Gibson, and Joy Bauch
- Subjects
Male ,Cancer Research ,Time Factors ,Metabolic Clearance Rate ,medicine.medical_treatment ,Poly ADP ribose polymerase ,Administration, Oral ,Pharmacology ,Poly(ADP-ribose) Polymerase Inhibitors ,Toxicology ,Poly (ADP-Ribose) Polymerase Inhibitor ,Article ,Cerebrospinal fluid ,Pharmacokinetics ,Oral administration ,Blood plasma ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Chemotherapy ,biology ,Molecular Structure ,Macaca mulatta ,Oncology ,Enzyme inhibitor ,Area Under Curve ,Immunology ,biology.protein ,Leukocytes, Mononuclear ,Benzimidazoles ,Poly(ADP-ribose) Polymerases - Abstract
ABT-888 inhibits poly(ADP-ribose) polymerase (PARP) and may enhance the efficacy of chemotherapy and radiation in CNS tumors. We studied the plasma and cerebrospinal fluid (CSF) pharmacokinetics (PK) of ABT-888 in a non-human primate (NHP) model that is highly predictive of human CSF penetration.ABT-888, 5 mg/kg, was administered orally to three NHPs. Serial blood and CSF samples were obtained. Plasma and CSF concentrations of ABT-888 were measured using LC/MS/MS, and the resulting concentration versus time data were evaluated using non-compartmental and compartmental PK methods.The CSF penetration of ABT-888 was 57+/-7% (mean+/-SD). The peak ABT-888 concentration in the plasma was 0.62+/-0.18 microM. Plasma and CSF AUC0-infinity were 3.7+/-1.7 and 2.1+/-0.8 microM h. PARP inhibition in peripheral blood mononuclear cells was evident 2 h after ABT-888 administration.The CSF penetration of ABT-888 after oral administration was 57%. Plasma and CSF concentrations were in the range that has been shown to inhibit PARP activity in vivo in humans.
- Published
- 2009
18. Discovery of an orally bioavailable small molecule inhibitor of prosurvival B-cell lymphoma 2 proteins
- Author
-
Hong Ding, Kunzer Aaron R, Kennan C. Marsh, Saul H. Rosenberg, Jessica Adickes, Wang Xilu, Steven W. Elmore, Stephen W. Fesik, Haichao Zhang, Alexander R. Shoemaker, Joy Bauch, Stephen K. Tahir, Xiufen Yang, Paul Nimmer, Michael D. Wendt, Cheol-Min Park, Xiaohong Song, Milan Bruncko, and Christin Tse
- Subjects
BH3 Mimetic ABT-737 ,Cell Survival ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,Administration, Oral ,Pharmacology ,medicine.disease_cause ,Cell Line ,Mice ,Structure-Activity Relationship ,In vivo ,Oral administration ,Drug Discovery ,medicine ,Animals ,Humans ,B-cell lymphoma ,Chemotherapy ,Sulfonamides ,Molecular Structure ,Chemistry ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Proto-Oncogene Proteins c-bcl-2 ,Apoptosis ,Molecular Medicine ,Carcinogenesis - Abstract
Overexpression of prosurvival proteins such as Bcl-2 and Bcl-X L has been correlated with tumorigenesis and resistance to chemotherapy, and thus, the development of antagonists of these proteins may provide a novel means for the treatment of cancer. We recently described the discovery of 1 (ABT-737), which binds Bcl-2, Bcl-X L, and Bcl-w with high affinity, shows robust antitumor activity in murine tumor xenograft models, but is not orally bioavailable. Herein, we report that targeted modifications at three key positions of 1 resulted in a 20-fold improvement in the pharmacokinetic/pharmacodynamic relationship (PK/PD) between oral exposure (AUC) and in vitro efficacy in human tumor cell lines (EC 50). The resulting compound, 2 (ABT-263), is orally efficacious in an established xenograft model of human small cell lung cancer, inducing complete tumor regressions in all animals. Compound 2 is currently in multiple phase 1 clinical trials in patients with small cell lung cancer and hematological malignancies.
- Published
- 2008
19. ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models
- Author
-
Thomas McGonigal, Gui Dong Zhu, Nayereh S. Ghoreishi-Haack, Yanping Luo, Bryan F. Cox, Jennifer J. Bouska, Amanda M. Olson, Joann P. Palma, David Frost, Boris Hristov, Lawrence Kleinberg, Yan Shi, Ken Jarvis, Ran Guan, Jason Stavropoulos, Saul H. Rosenberg, Yan Luo, Jonathan A. Meulbroek, Velitchka Bontcheva-Diaz, Loren M. Lasko, Debra Ferguson, Edward K. Han, Xuesong Liu, Kenneth B. Idler, Joy Bauch, Larry E. Dillehay, Eric F. Johnson, Vincent L. Giranda, Luis E. Rodriguez, Theodore L. DeWeese, Cherrie K. Donawho, David R. Grimm, Thomas D. Penning, Kennan C. Marsh, Rhonda R. Holley-Shanks, Vered Klinghofer, and Alan C. Tsurutani
- Subjects
Male ,Cancer Research ,Veliparib ,Administration, Oral ,Biological Availability ,Mice, Inbred Strains ,Pharmacology ,Poly(ADP-ribose) Polymerase Inhibitors ,Poly (ADP-Ribose) Polymerase Inhibitor ,chemistry.chemical_compound ,Mice ,Dogs ,In vivo ,Glioma ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,Antineoplastic Agents, Alkylating ,Cisplatin ,Temozolomide ,business.industry ,Drug Synergism ,Rats, Inbred Strains ,Haplorhini ,medicine.disease ,Xenograft Model Antitumor Assays ,Carboplatin ,Rats ,Disease Models, Animal ,Oncology ,chemistry ,Tumor progression ,Blood-Brain Barrier ,Benzimidazoles ,Female ,business ,medicine.drug ,DNA Damage - Abstract
Purpose: To evaluate the preclinical pharmacokinetics and antitumor efficacy of a novel orally bioavailable poly(ADP-ribose) polymerase (PARP) inhibitor, ABT-888. Experimental Design: In vitro potency was determined in a PARP-1 and PARP-2 enzyme assay. In vivo efficacy was evaluated in syngeneic and xenograft models in combination with temozolomide, platinums, cyclophosphamide, and ionizing radiation. Results: ABT-888 is a potent inhibitor of both PARP-1 and PARP-2 with Kis of 5.2 and 2.9 nmol/L, respectively. The compound has good oral bioavailability and crosses the blood-brain barrier. ABT-888 strongly potentiated temozolomide in the B16F10 s.c. murine melanoma model. PARP inhibition dramatically increased the efficacy of temozolomide at ABT-888 doses as low as 3.1 mg/kg/d and a maximal efficacy achieved at 25 mg/kg/d. In the 9L orthotopic rat glioma model, temozolomide alone exhibited minimal efficacy, whereas ABT-888, when combined with temozolomide, significantly slowed tumor progression. In the MX-1 breast xenograft model (BRCA1 deletion and BRCA2 mutation), ABT-888 potentiated cisplatin, carboplatin, and cyclophosphamide, causing regression of established tumors, whereas with comparable doses of cytotoxic agents alone, only modest tumor inhibition was exhibited. Finally, ABT-888 potentiated radiation (2 Gy/d × 10) in an HCT-116 colon carcinoma model. In each model, ABT-888 did not display single-agent activity. Conclusions: ABT-888 is a potent inhibitor of PARP, has good oral bioavailability, can cross the blood-brain barrier, and potentiates temozolomide, platinums, cyclophosphamide, and radiation in syngeneic and xenograft tumor models. This broad spectrum of chemopotentiation and radiopotentiation makes this compound an attractive candidate for clinical evaluation.
- Published
- 2007
20. Discovery of N-(4-(3-amino-1H-indazol-4-yl)phenyl)-N'-(2-fluoro-5-methylphenyl)urea (ABT-869), a 3-aminoindazole-based orally active multitargeted receptor tyrosine kinase inhibitor
- Author
-
Patrick A. Marcotte, Steven K. Davidsen, Joy Bauch, Junling Li, Yujia Dai, Niru B. Soni, David R. Reuter, Lori J. Pease, Paul Tapang, Dean Hickman, Christopher M. Harris, Donald J. Osterling, Amanda M. Olson, Vincent S. Stoll, Ruth L. Martin, Kent D. Stewart, Asma A Ahmed, George A. Cunha, Kresna Hartandi, Daniel H. Albert, Zhiqin Ji, Maria D Moskey, Jennifer J. Bouska, Peter F. Bousquet, Jun Guo, Michael R. Michaelides, Kennan C. Marsh, and Keith B. Glaser
- Subjects
Male ,Models, Molecular ,Platelet-derived growth factor ,Indazoles ,Receptor Protein-Tyrosine Kinases ,Administration, Oral ,Angiogenesis Inhibitors ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Adenosine Triphosphate ,Growth factor receptor ,Drug Discovery ,Animals ,Edema ,Humans ,Phosphorylation ,Binding Sites ,biology ,Estradiol ,Phenylurea Compounds ,Uterus ,Xenograft Model Antitumor Assays ,Linifanib ,chemistry ,Biochemistry ,Enzyme inhibitor ,biology.protein ,NIH 3T3 Cells ,Molecular Medicine ,Female ,Signal transduction ,Tyrosine kinase ,Hydrophobic and Hydrophilic Interactions - Abstract
In our continued efforts to search for potent and novel receptor tyrosine kinase (RTK) inhibitors as potential anticancer agents, we discovered, through a structure-based design, that 3-aminoindazole could serve as an efficient hinge-binding template for kinase inhibitors. By incorporating an N,N'-diaryl urea moiety at the C4-position of 3-aminodazole, a series of RTK inhibitors were generated, which potently inhibited the tyrosine kinase activity of the vascular endothelial growth factor receptor and the platelet-derived growth factor receptor families. A number of compounds with potent oral activity were identified by utilizing an estradiol-induced mouse uterine edema model and an HT1080 human fibrosarcoma xenograft tumor model. In particular, compound 17p (ABT-869) was found to possess favorable pharmacokinetic profiles across different species and display significant tumor growth inhibition in multiple preclinical animal models.
- Published
- 2007
21. A small-molecule inhibitor of Bcl-XL potentiates the activity of cytotoxic drugs in vitro and in vivo
- Author
-
Paul Nimmer, David Frost, Wang Shen, Mcclellan William J, Jacqueline M. O'Connor, Robert B. Warner, Stephen K. Tahir, Joy Bauch, Alex R. Shoemaker, Shi-Chung Ng, Anatol Oleksijew, Steven W. Elmore, Tony Borre, Tilman Oltersdorf, Haichao Zhang, Mary K. Joseph, Milan Bruncko, Jason Stavropoulos, Kennan C. Marsh, Saul H. Rosenberg, Weiguo Qing, Hugh N. Nellans, Barbara A. Belli, Stephen W. Fesik, Ken Jarvis, Thomas Deckwirth, and Baole Wang
- Subjects
Male ,Cancer Research ,Lung Neoplasms ,Paclitaxel ,Transplantation, Heterologous ,bcl-X Protein ,Bcl-xL ,Antineoplastic Agents ,Mice, SCID ,Biology ,Pharmacology ,Piperazines ,Nitrophenols ,chemistry.chemical_compound ,Mice ,In vivo ,Cell Line, Tumor ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Doxorubicin ,Etoposide ,Cisplatin ,Sulfonamides ,Aniline Compounds ,Biphenyl Compounds ,Biological activity ,Drug Synergism ,Kinetics ,Oncology ,chemistry ,biology.protein ,medicine.drug - Abstract
Inhibition of the prosurvival members of the Bcl-2 family of proteins represents an attractive strategy for the treatment of cancer. We have previously reported the activity of ABT-737, a potent inhibitor of Bcl-2, Bcl-XL, and Bcl-w, which exhibits monotherapy efficacy in xenograft models of small-cell lung cancer and lymphoma and potentiates the activity of numerous cytotoxic agents. Here we describe the biological activity of A-385358, a small molecule with relative selectivity for binding to Bcl-XL versus Bcl-2 (Ki's of 0.80 and 67 nmol/L for Bcl-XL and Bcl-2, respectively). This compound efficiently enters cells and co-localizes with the mitochondrial membrane. Although A-385358 shows relatively modest single-agent cytotoxic activity against most tumor cell lines, it has an EC50 of
- Published
- 2006
22. Plasma and cerebrospinal fluid pharmacokinetics of intravenously administered ABT-751 in non-human primates
- Author
-
Elizabeth Fox, Joy Bauch, Steve Y. Cho, Kennan C. Marsh, Cynthia McCully, and Frank M. Balis
- Subjects
Pharmacology ,Cancer Research ,Sulfonamides ,biology ,Sulfonamide (medicine) ,Toxicology ,Macaca mulatta ,Microtubule polymerization ,Bioavailability ,Tubulin ,Cerebrospinal fluid ,Oncology ,Pharmacokinetics ,Csf penetration ,Blood plasma ,Immunology ,biology.protein ,medicine ,Animals ,Pharmacology (medical) ,Infusions, Intravenous ,medicine.drug - Abstract
ABT-751 is an orally bioavailable sulfonamide that binds to the colchicine binding site on beta-tubulin and inhibits microtubule polymerization. The plasma and cerebrospinal fluid (CSF) pharmacokinetics of ABT-751, after a short intravenous infusion, were evaluated in a non-human primate (Macaca mulatta) model that is highly predictive of the CSF penetration of drugs in humans.Plasma and CSF samples were collected over 24 h after 7.5 mg/kg (150 mg/m2) ABT-751 infused over 0.25-0.70 h, and ABT-751 concentrations in plasma and CSF were quantified using a validated HPLC-MS/MS assay. Pharmacokinetic parameters in plasma and CSF were derived using non-compartmental methods.Plasma disappearance was bi-exponential with a terminal half-life of 13 h. The mean +/- SD clearance was 100 +/- 18 ml/min m2, the mean +/- SD volume of distribution at steady state was 1.3 +/- 0.5 l/kg, and the mean +/- SD mean residence time was 4.6 +/- 1.8 h. The mean +/- SD peak ABT-751 concentration in CSF was 0.26 +/- 0.08 microM, and the mean +/- SD CSF half-life of 1.3 +/- 0.3 h. CSF penetration was limited (mean +/- SD AUC(CSF):AUC(plasma), 1.1 +/- 0.3%) relative to total (protein-bound + free) plasma drug concentrations, but the CSF concentrations approximated the estimated free drug concentrations in plasma.
- Published
- 2006
23. Discovery and structure-activity relationship of antagonists of B-cell lymphoma 2 family proteins with chemopotentiation activity in vitro and in vivo
- Author
-
Hong Ding, Andrew M. Petros, Wendt Michael D, Milan Bruncko, Kunzer Aaron R, Alexander R. Shoemaker, Shi-Chung Ng, Kennan C. Marsh, Anatol Oleksijew, Thorsten Oost, Tilman Oltersdorf, Darlene Martineau, Joy Bauch, Haichao Zhang, Mcclellan William J, Paul Nimmer, Mary K. Joseph, Stephen W. Fesik, Wang Shen, Steven W. Elmore, Saul H. Rosenberg, and Barbara A. Belli
- Subjects
Serum ,Magnetic Resonance Spectroscopy ,Paclitaxel ,Ultraviolet Rays ,Transplantation, Heterologous ,bcl-X Protein ,Biological Availability ,Antineoplastic Agents ,Fluorescence Polarization ,Plasma protein binding ,Mice, SCID ,chemistry.chemical_compound ,Mice ,Piperidines ,In vivo ,Cell Line, Tumor ,Drug Discovery ,medicine ,Structure–activity relationship ,Animals ,Humans ,Serum Albumin ,Sulfonamides ,Aniline Compounds ,Chemistry ,Drug Synergism ,Stereoisomerism ,Human serum albumin ,In vitro ,Protein Structure, Tertiary ,Biochemistry ,Mechanism of action ,Cell culture ,Molecular Medicine ,medicine.symptom ,Drug Screening Assays, Antitumor ,medicine.drug ,Protein Binding - Abstract
Development of a rationally designed potentiator of cancer chemotherapy, via inhibition of Bcl-X(L) function, is described. Lead compounds generated by NMR screening and directed parallel synthesis displayed sub-microM binding but were strongly deactivated in the presence of serum. The dominant component of serum deactivation was identified as domain III of human serum albumin (HSA); NMR solution structures of inhibitors bound to both Bcl-X(L) and HSA domain III indicated two potential optimization sites for separation of affinities. Modifications at both sites resulted in compounds with improved Bcl-X(L) binding and greatly increased activity in the presence of human serum, culminating in 73R, which bound to Bcl-X(L) with a K(i) of 0.8 nM. In a cellular assay 73R reversed the protection afforded by Bcl-X(L) overexpression against cytokine deprivation in FL5.12 cells with an EC(50) of 0.47 microM. 73R showed little effect on the viability of the human non small cell lung cancer cell line A549. However, consistent with the proposed mechanism, 73R potentiated the activity of paclitaxel and UV irradiation in vitro and potentiated the antitumor efficacy of paclitaxel in a mouse xenograft model.
- Published
- 2006
24. Discovery of trans-3,4'-bispyridinylethylenes as potent and novel inhibitors of protein kinase B (PKB/Akt) for the treatment of cancer: Synthesis and biological evaluation
- Author
-
Vincent L. Giranda, Tilman Oltersdorf, Vincent S. Stoll, Shannon S Arries, Eric F. Johnson, Xuesong Liu, Yan Luo, Kennan C. Marsh, Jianchun Gong, Vered Klinghofer, Jennifer J. Bouska, Yan Shi, Qun Li, Chris Dalton, Ron De Jong, Clarissa G. Jakob, Akiyo Claibone, Saul H. Rosenberg, Tongmei Li, Joy Bauch, and Gui-Dong Zhu
- Subjects
Clinical Biochemistry ,Pharmaceutical Science ,AKT1 ,Antineoplastic Agents ,Protein Serine-Threonine Kinases ,Biochemistry ,Glycogen Synthase Kinase 3 ,Structure-Activity Relationship ,Adenosine Triphosphate ,Neoplasms ,Drug Discovery ,Humans ,Enzyme Inhibitors ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Protein kinase B ,CAMK ,Cell Proliferation ,Binding Sites ,Kinase ,Chemistry ,Organic Chemistry ,Ethylenes ,Protein-Tyrosine Kinases ,Calcium-Calmodulin-Dependent Protein Kinases ,Molecular Medicine ,Signal transduction ,Tyrosine kinase ,Proto-Oncogene Proteins c-akt - Abstract
A novel series of Akt/PKB inhibitors derived from a screening lead (1) has been prepared. The novel trans-3,4′-bispyridinylethylenes described herein are potent inhibitors of Akt/PKB with IC50 values in the low double-digit nanomolar range against Akt1. Compound 2q shows excellent selectivity against distinct families of kinases such as tyrosine kinases and CAMK, and displays poor to modest selectivity against closely related kinases in the AGC and CMGC families. The cellular activities including inhibition of cell growth and phosphorylation of downstream target GSK3 are also described. The X-ray structure of compound 2q complexed with PKA in the ATP binding site was determined.
- Published
- 2005
25. Design, synthesis, and biological activity of 4-[(4-cyano-2-arylbenzyloxy)-(3-methyl-3H-imidazol-4-yl)methyl]benzonitriles as potent and selective farnesyltransferase inhibitors
- Author
-
Charles W. Hutchins, Jerry Cohen, Kennan C. Marsh, David C. Park, Wang Xilu, Gerry Sullivan, Gary T. Wang, Le Wang, Nan-Horng Lin, Vincent S. Stoll, Qun Li, Nicholas M Leonard, Wen-Zhen Gu, Joy Bauch, Haiying Zhang, Saul H. Rosenberg, Hing L. Sham, Yunsong Tong, and Clarissa G. Jakob
- Subjects
Models, Molecular ,Cell Membrane Permeability ,Nitrile ,Stereochemistry ,Farnesyltransferase ,Administration, Oral ,Biological Availability ,Crystallography, X-Ray ,Chemical synthesis ,chemistry.chemical_compound ,Structure-Activity Relationship ,Dogs ,Drug Discovery ,Nitriles ,Structure–activity relationship ,Animals ,Farnesyltranstransferase ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Molecular Structure ,Aryl ,Imidazoles ,Biological activity ,chemistry ,Enzyme inhibitor ,Drug Design ,Benzamides ,biology.protein ,Molecular Medicine - Abstract
A novel series of 4-[(4-cyano-2-arylbenzyloxy)-(3-methyl-3H-imidazol-4-yl)methyl]benzonitriles have been synthesized as selective farnesyltransferase inhibitors using structure-based design. X-ray cocrystal structures of compound 20-FTase-HFP and A313326-FTase-HFP confirmed our initial design. The decreased interaction between the aryl groups and Ser 48 in GGTase-I binding site could be one possible reason to explain the improved selectivity for this new series of FTase inhibitors. Medicinal chemistry efforts led to the discovery of compound 64 with potent cellular activity (EC(50) = 3.5 nM) and outstanding pharmacokinetic profiles in dog (96% bioavailable, 18.4 h oral t(1/2), and 0.19 L/(h x kg) plasma clearance).
- Published
- 2004
26. Pyridone-containing farnesyltransferase inhibitors: synthesis and biological evaluation
- Author
-
Wen-Zhen Gu, Saul H. Rosenberg, Le Wang, Todd W. Rockway, Robert A. Mantei, Joy Bauch, Haiying Zhang, Hing L. Sham, Lisa A. Hasvold, Stephen L. Gwaltney, Stephen A. Fakhoury, Nelson Lissa T, Weibo Wang, Nan-Horng Lin, Qun Li, Kennan C. Marsh, Jerome Cohen, and Gerard M. Sullivan
- Subjects
Pyridones ,Farnesyltransferase ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Animals ,Farnesyltranstransferase ,Humans ,Enzyme Inhibitors ,Molecular Biology ,Biological evaluation ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Molecular Structure ,Organic Chemistry ,In vitro ,Enzyme ,chemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Cell Division - Abstract
Farnesyltransferase inhibitors (FTIs) have been developed as potential anti-cancer agents due to their efficacy in blocking malignant growth in a variety of murine models of human tumors. To that end, we have developed a series of pyridone farnesyltransferase inhibitors with potent in vitro and cellular activity. The synthesis, SAR and biological properties of these compounds will be discussed.
- Published
- 2003
27. Aryl tetrahydropyridine inhibitors of farnesyltransferase: bioavailable analogues with improved cellular potency
- Author
-
Charles W. Hutchins, Kennan C. Marsh, Stephen L. Gwaltney, Jerome Cohen, Vincent S. Stoll, Lisa A. Hasvold, Joy Bauch, Steve Muchmore, Gerard M. Sullivan, Qun Li, Weibo Wang, David Frost, Wen-Zhen Gu, Nelson Lissa T, Shi-Chung Ng, Stephen J. O'connor, Stephen K. Tahir, Clarissa G. Jakob, Hovis M. Imade, Saul H. Rosenberg, Haiying Zhang, and Hing L. Sham
- Subjects
Models, Molecular ,Alkylation ,Pyridines ,Farnesyltransferase ,Clinical Biochemistry ,Pharmaceutical Science ,Biological Availability ,Biochemistry ,Chemical synthesis ,chemistry.chemical_compound ,Structure-Activity Relationship ,Dogs ,Drug Discovery ,medicine ,Animals ,Farnesyltranstransferase ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Aryl ,Organic Chemistry ,Cancer ,medicine.disease ,In vitro ,Enzyme ,chemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Half-Life - Abstract
Inhibitors of farnesyltransferase are effective against a variety of tumors in mouse models of cancer. Clinical trials to evaluate these agents in humans are ongoing. In our effort to develop new farnesyltransferase inhibitors, we have discovered bioavailable aryl tetrahydropyridines that are potent in cell culture. The design, synthesis, SAR and biological properties of these compounds will be discussed.
- Published
- 2003
28. Discovery and characterization of the potent, selective and orally bioavailable MMP inhibitor ABT-770
- Author
-
Terrance J. Magoc, Douglas W. Morgan, Robert B. Garland, Michael L. Curtin, Douglas H. Steinman, Yan Guo, Paul Tapang, H. Robin Heyman, Carol K. Wada, James H. Holms, Ildiko B. Elmore, Steven K. Davidsen, Joy Bauch, Alan S. Florjancic, Patrick A. Marcotte, Daniel H. Albert, Joseph F. Dellaria, Carole L. Goodfellow, Jane Gong, Jennifer J. Bouska, Michael R. Michaelides, and Kennan C. Marsh
- Subjects
Matrix metalloproteinase inhibitor ,Stereochemistry ,Metabolic Clearance Rate ,Clinical Biochemistry ,Pharmaceutical Science ,Administration, Oral ,Biological Availability ,Antineoplastic Agents ,Matrix Metalloproteinase Inhibitors ,Hydroxamic Acids ,Biochemistry ,Chemical synthesis ,chemistry.chemical_compound ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Dogs ,In vivo ,Oral administration ,Drug Discovery ,Tumor Cells, Cultured ,Structure–activity relationship ,Animals ,Enzyme Inhibitors ,Molecular Biology ,Hydroxamic acid ,biology ,Organic Chemistry ,Biphenyl Compounds ,Biological activity ,Haplorhini ,Neoplasms, Experimental ,Rats ,chemistry ,Enzyme inhibitor ,Area Under Curve ,Injections, Intravenous ,biology.protein ,Molecular Medicine ,Cell Division ,Half-Life - Abstract
Modification of the biphenyl portion of MMP inhibitor 2a gave analogue 2i which is greater than 1000-fold selective against MMP-2 versus MMP-1. The stereospecific synthesis of both enantiomers of 2i was achieved beginning with (S)- or (R)-benzyl glycidyl ether. The (S)-enantiomer, 11 (ABT-770), is orally bioavailable and efficacious in an in vivo model of tumor growth.
- Published
- 2001
29. Potent inhibitors of protein farnesyltransferase: heteroarenes as cysteine replacements
- Author
-
Sajeev Cherian, Shi-Chung Ng, Joy Bauch, Haichao Zhang, Jerry Cohen, Greg Donner, Badr Saeed, Stephen K. Tahir, Wang Shen, Robert B. Warner, Hing L. Sham, Kenneth James Junior Henry, Kennan C. Marsh, Saul H. Rosenberg, Peter Kovar, Stephen A. Fakhoury, and Jang Lee
- Subjects
Stereochemistry ,Farnesyltransferase ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Chemical synthesis ,Mice ,Structure-Activity Relationship ,In vivo ,Drug Discovery ,Moiety ,Animals ,heterocyclic compounds ,Enzyme Inhibitors ,Molecular Biology ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Chemistry ,organic chemicals ,Organic Chemistry ,3T3 Cells ,In vitro ,Rats ,Genes, ras ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Epoxy Compounds ,Cysteine - Abstract
Synthesis and biological evaluation of heteroarenes as reduced cysteine replacements are described. Of the heteroaryl groups examined with respect to FT inhibitor FTI-276 (1), pyridyl was the replacement found to be most effective. Substitutions at C4 of the pyridyl moiety did not affect the in vitro activity. Compound 9a was found to have moderate in vivo bioavailability.
- Published
- 1999
30. Bcl-2 Family Protein Inhibitors Induce a Unique Thrombocytopenia In Vivo
- Author
-
Chris Tse, Joy Bauch, Kennan C. Marsh, Steven W. Elmore, Richard E. Nelson, Sherry J. Morgan, Glenn A. Reinhart, Saul H. Rosenberg, Ryan M. Fryer, Morey L. Smith, and Laurie Iciek
- Subjects
Disseminated intravascular coagulation ,Immunology ,Bcl-2 family ,Antagonist ,Cell Biology ,Hematology ,Biology ,Pharmacology ,Fibrinogen ,medicine.disease ,Biochemistry ,medicine.anatomical_structure ,In vivo ,medicine ,Platelet ,Bone marrow ,Mean platelet volume ,medicine.drug - Abstract
We recently disclosed the discovery of ABT-737, a potent antagonist of antiapoptotic Bcl-2 family proteins that induced tumor regression in murine xenograft models. ABT-263 is a related, orally bioavailable Bcl-2 family protein inhibitor that is currently in clinical development. For both agents, multiple daily dosing from two to four weeks was well tolerated in all species evaluated. The primary preclinical toxicological finding in mouse, rat and dog was a unique thrombocytopenia characterized by a rapid clearance of circulating platelets. A series of in vivo studies that were performed to better characterize this phenomenon are described here. After a single intravenous or oral dose in dogs, circulating platelet counts decreased rapidly and concentration dependently with a nadir at approximately six hours post administration. Platelet counts returned to near normal levels within several days post dose, accompanied by an increase in both mean platelet volume and percent of reticulated platelets. Analysis of platelets by aggregometry during this rebound period indicated that returning platelets were fully functional. Following multiple doses, platelet counts also decreased rapidly after initial dosing, but exhibited evidence of rebound during the dosing period that appeared to be dose dependant. Evaluation of fibrinogen, d-dimer, antithrombin-III, PT and APTT did not support a mechanism involving consumptive coagulopathy. Whole body scintography utilizing [111] Indium labeled platelets in dogs indicated that platelet clearance after compound administration is primarily mediated by the liver. These data suggest that ABT-737 and ABT-263 preferentially affect circulating platelets rather than abrogating platelet production in the bone marrow. In fact, the observed rebound in platelet count in the face of continued dosing reflects the capacity for the animals to compensate for the effect. The enantiomer of ABT-737 (which has significantly lower activity against Bcl-2 family proteins) had no effect on circulating platelets in animals suggesting an underlying mechanism that is dependent on inhibition of antiapoptotic Bcl-2 family proteins. The unusually rapid kinetics of platelet clearance and recovery suggests that the drug-induced thrombocytopenia elicited by these Bcl-2 family protein inhibitors is unique compared to that observed with conventional cytotoxic chemotherapy.
- Published
- 2006
- Full Text
- View/download PDF
31. The Bcl-2 Family Inhibitor ABT-263 Shows Significant but Reversible Thrombocytopenia in Mice
- Author
-
Saul H. Rosenberg, Joy Bauch, Michael J. Mitten, Stephen W. Fesik, Haichao Zhang, David Madar, Jessica Adickes, Alex R. Shoemaker, Steven W. Elmore, Kennan C. Marsh, Anatol Oleksijew, Christin Tse, and David Frost
- Subjects
medicine.medical_specialty ,Severe combined immunodeficiency ,Hematology ,medicine.diagnostic_test ,business.industry ,Immunology ,Spleen ,Cell Biology ,medicine.disease ,Biochemistry ,Lymphoma ,medicine.anatomical_structure ,Endocrinology ,Bleeding time ,Internal medicine ,Toxicity ,medicine ,Cytotoxic T cell ,Platelet ,business - Abstract
ABT-263 is an orally bioavailable small molecule inhibitor of Bcl-2 family proteins with a Ki of ≤ 1 nM against Bcl-2, Bcl-XL, and Bcl-w. Potent cytotoxic effects have been demonstrated against small cell lung carcinomas and lymphomas (EC50 of less than 500 nM in vitro against numerous cell lines). Tumor growth stasis and regression in xenograft models confirmed this activity. However, a unique thrombocytopenia has been observed in mice, rats, and dogs. Using a mouse model to characterize the extent and duration of decreased circulating platelets, outbred CF-1 or inbred scid-bg mice were treated with increasing doses of ABT-263. Blood samples were collected at various times post dosing and platelet numbers were determined using automated hematology analysis. The results showed a rapid, dose-dependent reduction in circulating platelet counts with a platelet nadir (>80% reduction relative to baseline) occurring approximately 6 hours after a single 100 mg/kg oral dose of ABT-263. Platelet counts returned to normal levels by 72 hrs post-dose. Analysis of bleeding times using a tail-nick method demonstrated a correlation between reduced platelet count and increased bleeding time. At doses of 50 mg/kg and higher, bleed times were increased by more than 5-fold relative to vehicle controls. In multi-dose studies, platelet levels did not continue to decline relative to the first dose of ABT-263. Mice treated with ABT-263 at 100 mg/kg/day for as long as 21 days exhibited no overt signs of toxicity (bleeding, significant weight loss, etc). Because the spleen is a potential site for clearing of damaged or aggregated platelets, a study using splenectomized mice was also conducted. The kinetics of platelet reduction and recovery were similar in splenectomized vs. intact mice with no additional adverse events noted. Evaluations of plasma drug levels associated with preclinical anti-tumor efficacy vs. thrombocytopenic effects indicate that a reasonable therapeutic window exists for ABT-263.
- Published
- 2006
- Full Text
- View/download PDF
32. Discovery of an Orally Bioavailable Small Molecule Inhibitor of Prosurvival B-Cell Lymphoma 2 Proteins.
- Author
-
Cheol-Min Park, Milan Bruncko, Jessica Adickes, Joy Bauch, Hong Ding, Aaron Kunzer, Kennan C. Marsh, Paul Nimmer, Alexander R. Shoemaker, Xiaohong Song, Stephen K. Tahir, Christin Tse, Xilu Wang, Michael D. Wendt, Xiufen Yang, Haichao Zhang, Stephen W. Fesik, Saul H. Rosenberg, and Steven W. Elmore
- Published
- 2008
- Full Text
- View/download PDF
33. Endothelin receptor antagonist has limited access to the fetal compartment during chronic maternal administration late in pregnancy
- Author
-
Joy Bauch, Sylvia Synowiec, Brian D. Dayton, Mark G. Neerhof, Saira Khan, and Larry G. Thaete
- Subjects
Litter (animal) ,medicine.medical_specialty ,Indoles ,Pyrrolidines ,Endothelin receptor antagonist ,Endothelin A Receptor Antagonists ,General Biochemistry, Genetics and Molecular Biology ,Fetal ,Fetal Development ,Rats, Sprague-Dawley ,Pharmacology, Toxicology and Pharmaceutics(all) ,Pregnancy ,Internal medicine ,Neonatal ,medicine ,Animals ,General Pharmacology, Toxicology and Pharmaceutics ,Maternal-Fetal Exchange ,Fetus ,Fetal Growth Retardation ,business.industry ,Biochemistry, Genetics and Molecular Biology(all) ,Antagonist ,Pregnancy Outcome ,General Medicine ,Azepines ,medicine.disease ,Rats ,Endocrinology ,Animals, Newborn ,Gestation ,Rat ,Female ,business ,Antagonism ,Endothelin receptor - Abstract
AimsEndothelin receptor A (ETA) antagonism normalizes fetal growth in several models of rodent fetal growth restriction (FGR). Our aims were to determine the levels of ETA antagonist in maternal and fetal plasma following chronic maternal administration, and to determine its impact on pregnancy outcome, survival and growth of rat pups.Main methodsTimed pregnant rats were treated with one of two endothelin receptor antagonists or vehicle, from gestation day 14–21 (term=22days). The antagonists and their respective doses were ABT-546 (20mg/kg/day) and FR139317 (12mg/kg/day). On day 21, in six rats per group, maternal and fetal plasma ABT-546 was assayed by HPLC. Five additional rats in each group delivered spontaneously and nursed their pups through postpartum day 7. Viability of newborns, oxygen saturation, litter sizes, and pup weights were recorded on postpartum days 1 and 7.Key findingsFetal antagonist levels reached only 2% of maternal levels (p
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.