21 results on '"Philip Lipari"'
Search Results
2. Supplementary Figure Legends 1-3, Table Legend 1 from A Fully Human Insulin-Like Growth Factor-I Receptor Antibody SCH 717454 (Robatumumab) Has Antitumor Activity as a Single Agent and in Combination with Cytotoxics in Pediatric Tumor Xenografts
- Author
-
Yan Wang, W. Robert Bishop, Jonathan A. Pachter, Ming Liu, Robert Ramos, Lianzhu Liang, Judith Hailey, Xiaoying Wang, Philip Lipari, and Yaolin Wang
- Abstract
Supplementary Figure Legends 1-3, Table Legend 1 from A Fully Human Insulin-Like Growth Factor-I Receptor Antibody SCH 717454 (Robatumumab) Has Antitumor Activity as a Single Agent and in Combination with Cytotoxics in Pediatric Tumor Xenografts
- Published
- 2023
3. Supplementary Figure 1 from A Fully Human Insulin-Like Growth Factor-I Receptor Antibody SCH 717454 (Robatumumab) Has Antitumor Activity as a Single Agent and in Combination with Cytotoxics in Pediatric Tumor Xenografts
- Author
-
Yan Wang, W. Robert Bishop, Jonathan A. Pachter, Ming Liu, Robert Ramos, Lianzhu Liang, Judith Hailey, Xiaoying Wang, Philip Lipari, and Yaolin Wang
- Abstract
Supplementary Figure 1 from A Fully Human Insulin-Like Growth Factor-I Receptor Antibody SCH 717454 (Robatumumab) Has Antitumor Activity as a Single Agent and in Combination with Cytotoxics in Pediatric Tumor Xenografts
- Published
- 2023
4. Data from A Fully Human Insulin-Like Growth Factor-I Receptor Antibody SCH 717454 (Robatumumab) Has Antitumor Activity as a Single Agent and in Combination with Cytotoxics in Pediatric Tumor Xenografts
- Author
-
Yan Wang, W. Robert Bishop, Jonathan A. Pachter, Ming Liu, Robert Ramos, Lianzhu Liang, Judith Hailey, Xiaoying Wang, Philip Lipari, and Yaolin Wang
- Abstract
The insulin-like growth factor-I receptor (IGF-IR) and its ligands (IGF-I and IGF-II) have been implicated in the growth, survival, and metastasis of a broad range of malignancies including pediatric tumors. Blocking the IGF-IR action is a potential cancer treatment. A fully human neutralizing monoclonal antibody, SCH 717454 (19D12, robatumumab), specific to IGF-IR, has shown potent antitumor effects in ovarian cancer in vitro and in vivo. In this study, SCH 717454 was evaluated in several pediatric solid tumors including neuroblastoma, osteosarcoma, and rhabdomyosarcoma. SCH 717454 is shown here to downregulate IGF-IR as well as inhibit IGF-IR and insulin receptor substrate-1 phosphorylation in pediatric tumor cells. IGF-IR and insulin receptor substrate-1 phosphorylation in the tumor cells. In vivo, SCH 717454 exhibits activity as a single agent and significantly inhibited growth of neuroblastoma, osteosarcoma, and rhabdomyosarcoma tumor xenografts. Combination of SCH 717454 with cisplatin or cyclophosphamide enhanced both the degree and the duration of the in vivo antitumor activity compared with single-agent treatments. Furthermore, SCH 717454 treatment markedly reduced Ki-67 expression and blood vessel formation in tumor xenografts, showing that the in vivo activity is derived from its inhibition of tumor cell proliferation and angiogenesis activity. Mol Cancer Ther; 9(2); 410–8
- Published
- 2023
5. Supplementary Figure 3 from A Fully Human Insulin-Like Growth Factor-I Receptor Antibody SCH 717454 (Robatumumab) Has Antitumor Activity as a Single Agent and in Combination with Cytotoxics in Pediatric Tumor Xenografts
- Author
-
Yan Wang, W. Robert Bishop, Jonathan A. Pachter, Ming Liu, Robert Ramos, Lianzhu Liang, Judith Hailey, Xiaoying Wang, Philip Lipari, and Yaolin Wang
- Abstract
Supplementary Figure 3 from A Fully Human Insulin-Like Growth Factor-I Receptor Antibody SCH 717454 (Robatumumab) Has Antitumor Activity as a Single Agent and in Combination with Cytotoxics in Pediatric Tumor Xenografts
- Published
- 2023
6. Supplementary Figure 2 from A Fully Human Insulin-Like Growth Factor-I Receptor Antibody SCH 717454 (Robatumumab) Has Antitumor Activity as a Single Agent and in Combination with Cytotoxics in Pediatric Tumor Xenografts
- Author
-
Yan Wang, W. Robert Bishop, Jonathan A. Pachter, Ming Liu, Robert Ramos, Lianzhu Liang, Judith Hailey, Xiaoying Wang, Philip Lipari, and Yaolin Wang
- Abstract
Supplementary Figure 2 from A Fully Human Insulin-Like Growth Factor-I Receptor Antibody SCH 717454 (Robatumumab) Has Antitumor Activity as a Single Agent and in Combination with Cytotoxics in Pediatric Tumor Xenografts
- Published
- 2023
7. Discovery of Novel 3,3-Disubstituted Piperidines as Orally Bioavailable, Potent, and Efficacious HDM2-p53 Inhibitors
- Author
-
Yaolin Wang, Yao Ma, Philip Lipari, Stephane L. Bogen, Cynthia Seidel-Dugan, Yinghui Lin, Asra Mirza, Craig R. Gibeau, Doll Ronald J, Corey Strickland, Giovanna Scapin, Winifred W. Prosise, Diane Rindgen, Paul Reichert, Xiaoying Wang, Ming Liu, Brian R. Lahue, Daniel J. Hicklin, Gerald W. Shipps, Tian Yuan, Elise Seigel, Suxing Liu, W. Robert Bishop, Weidong Pan, Amin A. Nomeir, and Latha G. Nair
- Subjects
0301 basic medicine ,Trifluoromethyl ,Stereochemistry ,Organic Chemistry ,Biochemistry ,Bioavailability ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,Drug Discovery ,Tumor regression ,Moiety ,Piperidine ,Human cancer - Abstract
A new subseries of substituted piperidines as p53-HDM2 inhibitors exemplified by 21 has been developed from the initial lead 1. Research focused on optimization of a crucial HDM2 Trp23-ligand interaction led to the identification of 2-(trifluoromethyl)thiophene as the preferred moiety. Further investigation of the Leu26 pocket resulted in potent, novel substituted piperidine inhibitors of the HDM2-p53 interaction that demonstrated tumor regression in several human cancer xenograft models in mice. The structure of HDM2 in complex with inhibitors 3, 10, and 21 is described.
- Published
- 2016
8. Toxin-Mediated Paracellular Transport of Antitoxin Antibodies Facilitates Protection against Clostridium difficile Infection
- Author
-
Zuo Zhang, Xinhua Chen, Ciaran P. Kelly, S. Kramer, Amy M. Flattery, Alex G. Therien, Shi-Juan Chen, Lorraine D. Hernandez, Fred Racine, H. Cape, Philip Lipari, and Jon D. Polishook
- Subjects
Male ,Bacterial Toxins ,Immunology ,Fc receptor ,Receptors, Fc ,Microbiology ,Immunoglobulin G ,Enterotoxins ,Organ Culture Techniques ,Bacterial Proteins ,Animals ,Transcellular ,Mice, Knockout ,Mesocricetus ,biology ,Clostridioides difficile ,Histocompatibility Antigens Class I ,Immunization, Passive ,Bacterial Infections ,Antibodies, Bacterial ,Antibodies, Neutralizing ,Author Corrections ,Gut Epithelium ,Mice, Inbred C57BL ,Disease Models, Animal ,Infectious Diseases ,Bezlotoxumab ,Paracellular transport ,Clostridium Infections ,biology.protein ,Female ,Parasitology ,Antitoxins ,Antibody ,Antitoxin - Abstract
The exotoxins TcdA and TcdB are the major virulence factors of Clostridium difficile . Circulating neutralizing antitoxin antibodies are protective in C. difficile infection (CDI), as demonstrated, in part, by the protective effects of actoxumab and bezlotoxumab, which bind to and neutralize TcdA and TcdB, respectively. The question of how systemic IgG antibodies neutralize toxins in the gut lumen remains unresolved, although it has been suggested that the Fc receptor FcRn may be involved in active antibody transport across the gut epithelium. In this study, we demonstrated that genetic ablation of FcRn and excess irrelevant human IgG have no impact on actoxumab-bezlotoxumab-mediated protection in murine and hamster models of CDI, suggesting that Fc-dependent transport of antibodies across the gut wall is not required for efficacy. Tissue distribution studies in hamsters suggest, rather, that the transport of antibodies depends on toxin-induced damage to the gut lining. In an in vitro two-dimensional culture system that mimics the architecture of the intestinal mucosal epithelium, toxins on the apical side of epithelial cell monolayers are neutralized by basolateral antibodies, and antibody transport across the cell layer is dramatically increased upon addition of toxin to the apical side. Similar data were obtained with F(ab′) 2 fragments, which lack an Fc domain, consistent with FcRn-independent paracellular, rather than transcellular, transport of antibodies. Kinetic studies show that initial damage caused by apical toxin is required for efficient neutralization by basolateral antibodies. These data may represent a general mechanism of humoral response-mediated protection against enteric pathogens.
- Published
- 2014
9. Inhibition of insulin-like growth factor-I receptor (IGF-IR) signaling and tumor cell growth by a fully human neutralizing anti–IGF-IR antibody
- Author
-
Peter Brams, Yaolin Wang, Deba Saha, Xiaoying Wang, Michael Malkowski, Guanghua Li, Denise Williams, Yan Wang, Lei Xie, Leonard G. Presta, Judith Hailey, Philip Lipari, Robert A. Ramos, Susan Cannon-Carlson, W. Robert Bishop, Robert Greenberg, Jonathan A. Pachter, Wai Lam W. Ling, and Robert L. Shields
- Subjects
Cancer Research ,Down-Regulation ,Antineoplastic Agents ,Protein Serine-Threonine Kinases ,Biology ,Receptor tyrosine kinase ,Receptor, IGF Type 1 ,Mice ,Growth factor receptor ,Cell surface receptor ,Cell Line, Tumor ,Neoplasms ,Proto-Oncogene Proteins ,Animals ,Humans ,Insulin-Like Growth Factor I ,Phosphorylation ,Protein kinase B ,Insulin-like growth factor 1 receptor ,Antibodies, Monoclonal ,Phosphoproteins ,Xenograft Model Antitumor Assays ,Insulin receptor ,Oncology ,Insulin Receptor Substrate Proteins ,biology.protein ,Cancer research ,Dimerization ,Proto-Oncogene Proteins c-akt ,Tyrosine kinase ,Platelet-derived growth factor receptor ,Signal Transduction - Abstract
Insulin-like growth factor-I receptor (IGF-IR) plays an important role in tumor cell growth and survival. On ligand stimulation, IGF-IR, a receptor tyrosine kinase, phosphorylates tyrosine residues on two major substrates, IRS-1 and Shc, which subsequently signal through the Ras/mitogen-activated protein kinase and phosphatidylinositol 3-kinase/AKT pathways. Here, we describe the characterization of a fully human anti–IGF-IR monoclonal antibody 19D12 that inhibits IGF binding and autophosphorylation of both IGF-IR/IGF-IR homodimers and IGF-IR/insulin receptor heterodimers. 19D12 does not recognize insulin receptor homodimers. In addition to inhibiting IGF-IR autophosphorylation, 19D12 also inhibits IRS-1 phosphorylation and activation of the major downstream signaling molecules AKT and extracellular signal-regulated kinase 1/2. Furthermore, the antibody down-regulates the total IGF-IR protein level and can exhibit antibody-dependent cellular cytotoxicity activity against a non–small cell adenocarcinoma cell line in vitro in the presence of isolated human natural killer cells. 19D12 binds tightly to the receptor, with an affinity of 3.8 pmol/L as measured by KinExA. In cell culture, 19D12 inhibits proliferation and soft agar growth of various tumor cell lines. In vivo, 19D12 inhibits the tumor growth of a very aggressive human ovarian tumor xenograft model A2780. These data support the development of this anti–IGF-IR monoclonal antibody as a promising anticancer agent.
- Published
- 2005
10. Kibdelomycin Is a Potent and Selective Agent against Toxigenic Clostridium difficile
- Author
-
Lynn Miesel, Amy M. Flattery, Jon D. Polishook, James R. Osmolski, Jing Lan, Lianzhu Liang, Fangbiao Li, Philip Lipari, Dale N. Gerding, Sheo B. Singh, David W. Hecht, Jenny Liu, and David B. Olsen
- Subjects
Drug ,Male ,media_common.quotation_subject ,Hamster ,Microbial Sensitivity Tests ,Biology ,Microbiology ,Mice ,Cricetinae ,Kibdelomycin ,medicine ,Animals ,Pharmacology (medical) ,Experimental Therapeutics ,Colitis ,media_common ,Pharmacology ,chemistry.chemical_classification ,Clostridioides difficile ,Clostridium difficile ,medicine.disease ,Anti-Bacterial Agents ,Diarrhea ,Infectious Diseases ,Enzyme ,chemistry ,Clostridium Infections ,medicine.symptom ,Anaerobic exercise - Abstract
Clostridium difficile is the causative agent of C. difficile -associated diarrhea (CDAD), with increased risk in elderly populations. Kibdelomycin, a novel natural-product inhibitor of type II topoisomerase enzymes, was evaluated for activity against C. difficile and gastrointestinal anaerobic organisms. Toxigenic C. difficile isolates ( n = 168) from U.S. hospitals and anaerobic Gram-positive and Gram-negative organisms ( n = 598) from Chicago-area hospitals were tested. Kibdelomycin showed potent activity against toxigenic C. difficile (MIC 90 = 0.25 μg/ml) and most Gram-positive aerobic organisms but had little activity against Bacteroides species (MIC 50 > 32 μg/ml; n = 270). Potent anti- C. difficile activity was also observed in the hamster model of C. difficile colitis. Dosing at 1.6 mg/kg (twice-daily oral dose) resulted in protection from a lethal infection and a 2-log reduction in C. difficile cecal counts. A 6.25-mg/kg twice-daily oral dose completely eliminated detectable C. difficile counts in cecal contents. A single 6.25-mg/kg oral dose showed that cecal contents were exposed to the drug at >2 μM (eightfold higher than the MIC), with no significant plasma exposure. These findings support further exploration of kibdelomycin for development of an anti- C. difficile agent.
- Published
- 2014
11. Adenovirus-mediated p53 gene therapy has greater efficacy when combined with chemotherapy against human head and neck, ovarian, prostate, and breast cancer
- Author
-
Maya Gurnani, Janet Dell, Loretta L. Nielsen, Bin Shi, and Philip Lipari
- Subjects
Male ,Cancer Research ,Tumor suppressor gene ,medicine.medical_treatment ,Genetic enhancement ,Antineoplastic Agents ,Breast Neoplasms ,Mice, SCID ,Toxicology ,Adenoviridae ,Mice ,Breast cancer ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Doxorubicin ,Ovarian Neoplasms ,Pharmacology ,Cisplatin ,Mice, Inbred ICR ,Chemotherapy ,business.industry ,Prostatic Neoplasms ,Cancer ,Genetic Therapy ,Genes, p53 ,medicine.disease ,Combined Modality Therapy ,Oncology ,Head and Neck Neoplasms ,Immunology ,Cancer research ,Female ,Ovarian cancer ,business ,DNA Damage ,medicine.drug - Abstract
Purpose: Adenovirus-mediated p53 gene therapy for cancer is currently undergoing phase I/II clinical trials. The drug used in our clinical trials (p53 Ad; ACN53; SCH58500) consists of a replication-deficient, type 5 adenovirus vector expressing human wildtype p53 tumor suppressor under the control of the cytomegalovirus promoter. In preclinical models, p53 Ad has therapeutic efficacy against a wide range of human tumor types containing nonfunctional p53, both in vitro and in vivo. Results from early clinical trials using p53 gene therapy by itself support optimism for the future of this therapeutic approach. However, it is likely that many phase II/III trials will incorporate an arm comparing traditional chemotherapy against chemotherapy combined with p53 gene therapy. Therefore, it is important to study possible interactions between p53 Ad and chemotherapeutic drugs in preclinical models before starting the clinical trials. Methods: Proliferation of tumor cells was quantitated after incubation with various combinations of p53 Ad and chemotherapeutic drugs. Human tumor xenografts in scid mice were dosed with intraperitoneal or intratumoral p53 Ad with or without chemotherapeutic drugs and the tumor burden after therapy monitored. Results: p53 Ad combined with cisplatin, doxorubicin, 5-fluorouracil, methotrexate, or etoposide inhibited cell proliferation more effectively than chemotherapy alone in SCC-9 head and neck, SCC-15 head and neck, SCC-25 head and neck, SK-OV-3 ovarian, DU-145 prostate, MDA-MB-468 breast, and MDA-MB-231 breast tumor cells. No obvious dependence on dosing schedule was observed. Greater anticancer efficacy was also demonstrated in four human tumor xenograft models in vivo. Of particular significance, there was enhanced efficacy using the three drug combination of p53 Ad, cisplatin, and paclitaxel in an ovarian cancer model. Conclusion: These results support the combination of p53 gene therapy with chemotherapy in clinical trials.
- Published
- 1999
12. Inhibitors of Farnesyl Protein Transferase. 4-Amido, 4-Carbamoyl, and 4-Carboxamido Derivatives of 1-(8-Chloro-6,11-dihydro-5H-benzo[5,6]- cyclohepta[1,2-b]pyridin-11-yl)piperazine and 1-(3-Bromo-8-chloro-6,11- dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-yl)piperazine
- Author
-
Alan K. Mallams, Randall R. Rossman, Ronald J. Doll, Viyyoor M. Girijavallabhan, Ashit K. Ganguly, Joanne Petrin, Lynn Wang, Robert Patton, W. Robert Bishop, Donna M. Carr, Paul Kirschmeier, Joseph J. Catino, Matthew S. Bryant, Kwang-Jong Chen, Walter A. Korfmacher, Cymbelene Nardo, Shiyong Wang, Amin A. Nomeir, Chin-Chung Lin, Zujun Li, Jianping Chen, Suining Lee, Janet Dell, Philip Lipari, Michael Malkowski, Bodan Yaremko, Ivan King, and Ming Liu
- Subjects
chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,biology ,Farnesyl Protein Transferase ,Stereochemistry ,Chemical synthesis ,Nitrone ,Piperazine ,chemistry.chemical_compound ,Enzyme ,chemistry ,Enzyme inhibitor ,Drug Discovery ,biology.protein ,Molecular Medicine ,Moiety - Abstract
The synthesis of a variety of novel 4-amido, 4-carbamoyl and 4-carboxamido derivatives of 1-(8-chloro-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-yl)piperazine to explore the SAR of of this series of FPT inhibitors is described. This resulted in the synthesis of the 4- and 3-pyridylacetyl analogues 45a and 50a, respectively, both of which were orally active but were found to be rapidly metabolized in vivo. Identification of the principal metabolites led to the synthesis of a variety of new compounds that would be less readily metabolized, the most interesting of which were the 3- and 4-pyridylacetyl N-oxides 80a and 83a. Novel replacements for the pyridylacetyl moiety were also sought, and this resulted in the discovery of the 4-N-methyl and 4-N-carboxamidopiperidinylacetyl derivatives 135a and 160a, respectively. All of these derivatives exhibited greatly improved pharmacokinetics. The synthesis of the corresponding 3-bromo analogues resulted in the discovery of the 4-pyridylacetyl N-oxides 83b...
- Published
- 1998
13. Correction for Zhang et al., Toxin-Mediated Paracellular Transport of Antitoxin Antibodies Facilitates Protection against Clostridium difficile Infection
- Author
-
Alex G. Therien, S. Kramer, Philip Lipari, Xinhua Chen, Zuo Zhang, Lorraine D. Hernandez, H. Cape, Jon D. Polishook, Shi-Juan Chen, Ciaran P. Kelly, Amy M. Flattery, and Fred Racine
- Subjects
biology ,Toxin ,Immunology ,Clostridium difficile ,medicine.disease_cause ,Microbiology ,Virology ,Infectious Diseases ,Paracellular transport ,biology.protein ,medicine ,Parasitology ,Antitoxin ,Antibody - Abstract
Volume 83, no. 1, p. [405–416][1], 2015. Page 408: Figure 1 should appear as shown below. ![FIG 1][2] FIG 1 [1]: /lookup/doi/10.1128/IAI.02550-14 [2]: pending:yes
- Published
- 2015
14. A fully human insulin-like growth factor-I receptor antibody SCH 717454 (Robatumumab) has antitumor activity as a single agent and in combination with cytotoxics in pediatric tumor xenografts
- Author
-
Judith Hailey, Robert A. Ramos, Yan Wang, W. Robert Bishop, Xiaoying Wang, Yaolin Wang, Ming Liu, Jonathan A. Pachter, Philip Lipari, and Lianzhu Liang
- Subjects
Cancer Research ,Angiogenesis ,Mice, Nude ,Antineoplastic Agents ,Pharmacology ,Metastasis ,Receptor, IGF Type 1 ,Mice ,In vivo ,Neuroblastoma ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Insulin-Like Growth Factor I ,Phosphorylation ,Rhabdomyosarcoma ,biology ,Neovascularization, Pathologic ,Antibodies, Monoclonal ,medicine.disease ,Insulin receptor ,Ki-67 Antigen ,Oncology ,biology.protein ,Osteosarcoma ,Female ,Ovarian cancer ,Neoplasm Transplantation - Abstract
The insulin-like growth factor-I receptor (IGF-IR) and its ligands (IGF-I and IGF-II) have been implicated in the growth, survival, and metastasis of a broad range of malignancies including pediatric tumors. Blocking the IGF-IR action is a potential cancer treatment. A fully human neutralizing monoclonal antibody, SCH 717454 (19D12, robatumumab), specific to IGF-IR, has shown potent antitumor effects in ovarian cancer in vitro and in vivo. In this study, SCH 717454 was evaluated in several pediatric solid tumors including neuroblastoma, osteosarcoma, and rhabdomyosarcoma. SCH 717454 is shown here to downregulate IGF-IR as well as inhibit IGF-IR and insulin receptor substrate-1 phosphorylation in pediatric tumor cells. IGF-IR and insulin receptor substrate-1 phosphorylation in the tumor cells. In vivo, SCH 717454 exhibits activity as a single agent and significantly inhibited growth of neuroblastoma, osteosarcoma, and rhabdomyosarcoma tumor xenografts. Combination of SCH 717454 with cisplatin or cyclophosphamide enhanced both the degree and the duration of the in vivo antitumor activity compared with single-agent treatments. Furthermore, SCH 717454 treatment markedly reduced Ki-67 expression and blood vessel formation in tumor xenografts, showing that the in vivo activity is derived from its inhibition of tumor cell proliferation and angiogenesis activity. Mol Cancer Ther; 9(2); 410–8
- Published
- 2010
15. Fluorescence polarization assay and inhibitor design for MDM2/p53 interaction
- Author
-
Rumin, Zhang, Todd, Mayhood, Philip, Lipari, Yaolin, Wang, James, Durkin, Rosalinda, Syto, Jennifer, Gesell, Charles, McNemar, and William, Windsor
- Subjects
Drug Design ,Neoplasms ,Proto-Oncogene Proteins ,Cell Cycle ,Humans ,Nuclear Proteins ,Antineoplastic Agents ,Fluorescence Polarization ,Proto-Oncogene Proteins c-mdm2 ,Enzyme Inhibitors ,Tumor Suppressor Protein p53 ,Peptides ,Protein Binding - Abstract
MDM2 is an important negative regulator of the tumor suppressor protein p53 which regulates the expression of many genes including MDM2. The delicate balance of this autoregulatory loop is crucial for the maintenance of the genome and control of the cell cycle and apoptosis. MDM2 hyperactivity, due to amplification/overexpression or mutational inactivation of the ARF locus, inhibits the function of wild-type p53 and can lead to the development of a wide variety of cancers. Thus, the development of anti-MDM2 therapies may restore normal p53 function in tumor cells and induce growth suppression and apoptosis. We report here a novel high-throughput fluorescence polarization binding assay and its application in rank ordering small-molecule inhibitors that block the binding of MDM2 to a p53-derived fluorescent peptide.
- Published
- 2004
16. Effects of SCH 59228, an orally bioavailable farnesyl protein transferase inhibitor, on the growth of oncogene-transformed fibroblasts and a human colon carcinoma xenograft in nude mice
- Author
-
Matthew Bryant, Dell J, Ming Liu, Amin A. Nomeir, Bohdan Yaremko, Randall R. Rossman, Paul Kirschmeier, Doll Ronald J, Chin-Chung Lin, Walter A. Korfmacher, Joseph J. Catino, N. Prioli, M. Malkowski, Philip Lipari, Walter R. Bishop, V. Girijavallabhan, Z. Li, Alan K. Mallams, Jianping Chen, and Suininhg Lee
- Subjects
Cancer Research ,Farnesyl Protein Transferase ,medicine.medical_treatment ,Biological Availability ,Mice, Nude ,Antineoplastic Agents ,Biology ,Toxicology ,Transfection ,Piperazines ,Cyclic N-Oxides ,chemistry.chemical_compound ,Mice ,Prenylation ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Enzyme Inhibitors ,Cell Line, Transformed ,Pharmacology ,Farnesyl-diphosphate farnesyltransferase ,Genes, mos ,COS cells ,Alkyl and Aryl Transferases ,Oncogene ,Growth factor ,Fibroblasts ,Molecular biology ,Genes, ras ,Oncology ,chemistry ,Biochemistry ,Colonic Neoplasms ,Signal transduction ,Growth inhibition ,Cell Division ,Neoplasm Transplantation - Abstract
The products of the Ha-, Ki-, and N-ras proto-oncogenes comprise a family of 21 kDa guanine nucleotide-binding proteins which play a crucial role in growth factor signal transduction and in the control of cellular proliferation and differentiation. Activating mutations in the ras oncogenes occur in a wide variety of human tumors. Ras proteins undergo a series of posttranslational processing events. The first modification is addition of the 15-carbon isoprene, farnesyl, to a Cys residue near the carboxy-terminus of Ras. Prenylation allows the Ras oncoprotein to localize to the plasma membrane where it can initiate downstream signalling events leading to cellular transformation. Inhibitors of the enzyme which catalyzes this step, farnesyl protein transferase (FPT), are a potential class of novel anticancer drugs which interfere with Ras function. SCH 59228 is a tricyclic FPT inhibitor which inhibits the farnesylation of purified Ha-Ras with an IC50 of 95 nM and blocks the processing of Ha-Ras in Cos cells with an IC50 of 0.6 microM. SCH 59228 has favorable pharmacokinetic properties upon oral dosing in nude mice. The in vivo efficacy of SCH 59228 was evaluated using a panel of tumor models grown in nude mice. These included several rodent fibroblast lines expressing mutationally-activated (val12) forms of the Ha-Ras oncogene. In some cases, these proteins contain their native C-terminal sequence (CVLS) which directs farnesylation. In one model, the C-terminal sequence was altered to CVLL, making the expressed protein a substrate for a distinct prenyl transferase, geranylgeranyl protein transferase-1. When dosed orally at 10 and 50 mg/kg (four times a day, 7 days a week) SCH 59228 significantly inhibited tumor growth of cells expressing farnesylated Ha-Ras in a dose-dependent manner; over 90% growth inhibition was observed at the 50 mg/kg dose. Tumor growth of cells expressing the geranylgeranylated form of Ha-Ras was less potently inhibited. Growth of tumors derived from a rodent fibroblast line expressing activated Ki-Ras containing its native C-terminal sequence (CVIM), which preferentially directs farnesylation, was also inhibited by SCH 59228. Inhibition in the Ki-Ras model was less than that observed in the Ha-Ras model. In contrast, tumors derived from cells transformed with the mos oncogene were not significantly inhibited even at the highest dose level. SCH 59228 also significantly and dose-dependently inhibited the growth of human colon adenocarcinoma DLD-1 xenografts (which express activated Ki-ras). These results indicate that SCH 59228 possesses in vivo antitumor activity upon oral dosing in tumor models expressing activated ras oncogenes. This is the first report of oral antitumor activity with an FPT inhibitor. These results are discussed in light of recent observations on alternative prenylation of some Ras isoforms.
- Published
- 1999
17. Structure-activity relationship of 3-substituted N-(pyridinylacetyl)-4- (8-chloro-5,6-dihydro-11H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-ylidene )- piperidine inhibitors of farnesyl-protein transferase: design and synthesis of in vivo active antitumor compounds
- Author
-
Bohdan Yaremko, Ashit K. Ganguly, D. F. Rane, M. Malkowski, Robert Patton, Chin-Chung Lin, Walter R. Bishop, Bancha Vibulbhan, Jianping Chen, Amin A. Nomeir, Philip Lipari, Doll Ronald J, Matthew Bryant, Joanne M. Petrin, Chen Kj, Dell J, Ming Liu, Z. Li, F. G. Njoroge, Girijavallabhan, I. King, Suining Lee, and Joseph J. Catino
- Subjects
Magnetic Resonance Spectroscopy ,Farnesyl Protein Transferase ,medicine.drug_class ,Stereochemistry ,Pyridines ,Substituent ,Protein Prenylation ,Mice, Nude ,Carboxamide ,Antineoplastic Agents ,Transfection ,Chemical synthesis ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Piperidines ,Drug Discovery ,medicine ,Structure–activity relationship ,Phenyl group ,Animals ,Enzyme Inhibitors ,Alkyl and Aryl Transferases ,biology ,Molecular Structure ,3T3 Cells ,Neoplasms, Experimental ,chemistry ,Enzyme inhibitor ,Drug Design ,COS Cells ,biology.protein ,ras Proteins ,Molecular Medicine ,Piperidine - Abstract
Novel tricyclic Ras farnesyl-protein transferase (FPT) inhibitors are described. A comprehensive structure-activity relationship (SAR) study of compounds arising from substitution at the 3-position of the tricyclic pyridine ring system has been explored. In the case of halogens, the chloro, bromo, and iodo analogues 19, 22, and 28 were found to be equipotent. However, the fluoro analogue 17 was an order of magnitude less active. Whereas a small alkyl substituent such as a methyl group resulted in a very potent FPT inhibitor (SCH 56580), introduction of bulky substituents such as tert-butyl, compound 33, or a phenyl group, compound 29, resulted in inactive FPT inhibitors. Polar groups at the 3-position such as amino 5, alkylamino 6, and hydroxyl 12 were less active. Whereas compound SCH 44342 did not show appreciable in vivo antitumor activity, the 3-bromo-substituted pyridyl N-oxide amide analogue 38 was a potent FPT inhibitor that reduced tumor growth by 81% when administered q.i.d. at 50 mpk and 52% at 10 mpk. These compounds are nonpeptidic and do not contain sulfhydryl groups. They selectively inhibit FPT and not geranylgeranyl-protein transferase-1 (GGPT-1). They also inhibit H-Ras processing in COS monkey kidney cells and soft agar growth of Ras-transformed cells.
- Published
- 1998
18. Abstract 2785: Discovery of a novel HDM2 inhibitor with potent in vivo anti-tumor activity
- Author
-
Yaolin Wang, Philip Lipari, Asra Mirza, Yinghui Lin, Rumin Zhang, Stephane Bogen, Yao Ma, Daniel J. Hicklin, Cynthia Seidel-Dugan, Suxing Liu, Xiaoying Wang, Ronald Doll, Gerald Shipps, and Walter Robert Bishop
- Subjects
Cancer Research ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Pharmacology ,medicine.disease ,In vitro ,chemistry.chemical_compound ,Oncology ,chemistry ,Cell culture ,In vivo ,Cancer cell ,Medicine ,Osteosarcoma ,Growth inhibition ,business ,Ovarian cancer - Abstract
HDM2 is a major negative regulator of the p53 tumor suppressor pathway. Aberrant HDM2 overexpression and gene amplification contributed to accelerated cancer development and growth. Several small molecule inhibitors of HDM2-p53 protein-protein interaction have been reported in recent years with anti-tumor activities in tumor xenograft models. Here we describe a novel and potent small molecule inhibitor of HDM-p53 inhibitor that binds selectively to HDM2 with high affinity compared to HDM4. Treatment of cancer cells with this HDM2 inhibitor results in activation of p53 pathway as demonstrated by study of pharmacodynamic biomarkers both in cell culture and in tumor xenograft in vivo. More importantly, cancer cells response to this HDM2 inhibitor is mechanism based and dependent on the presence of functional p53 status as shown in a profiling of a broad-panel of cancer cell lines. This inhibitor is very potent against cancer cell growth with IC50 below 200 nM for most cell lines tested in vitro. It is orally bioavailable and has single agent activity that results tumor regression in SJSA-1 osteosarcoma model or growth inhibition in A549 NSCLC and A2780 ovarian cancer xenograft models. In addition, combination of this HDM2 inhibitor with various chemotherapy agents results in added or synergistic anti-tumor response both in vitro and in vivo in several human cancer xenograft models with limited bone marrow toxicity at the efficacious dose. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2785. doi:1538-7445.AM2012-2785
- Published
- 2012
19. Abstract 1536: Characterization of tumor cell lines selected for resistance to the HDM2 antagonist Nutlin-3a
- Author
-
David James Devlin, Suxing Liu, Philip Lipari, Yaolin Wang, Diane Levitane, Lei Chen, Xiaoying Wang, Jason S. Simon, Daniel J. Hicklin, Cynthia Seidel-Dugan, and Walter Robert Bishop
- Subjects
Senescence ,Cancer Research ,Programmed cell death ,DNA damage ,Nutlin ,Biology ,In vitro ,law.invention ,chemistry.chemical_compound ,Oncology ,chemistry ,law ,In vivo ,Immunology ,Cancer research ,Suppressor ,Gene - Abstract
The tumor suppressor p53 is activated in response to various cellular stress signals including DNA damage and oncogene activation. Activated p53 regulates the expression of multiple downstream target genes involved in cell cycle control, cell death and senescence. Dysfunction of the p53 pathway is the most frequent alteration in human cancers. HDM2 protein binds directly to p53 and acts as a major negative regulator of the p53 function. Nutlin-3a is an antagonist of the HDM2-p53 protein-protein interaction and restores p53 pathway function in tumors with wild-type p53. Nutlin-3a demonstrates anti-tumor activity both in vitro and in vivo. Emergence of resistance is frequently observed with many anti-cancer therapeutics. The goal of the current studies is to understand the mechanism of resistance to Nutlin-3a. Cancer cell lines were treated with Nutlin-3a for an extended period and resistant clones were isolated. We found using molecular profiling studies that the emergence of Nutlin resistance is primarily due to the inactivation of the p53 signaling pathway through either mutation of the p53 gene or loss of p53 protein expression. These Nutlin-resistant cells are also resistant to others antagonists of the HDM2-p53 protein-protein interaction. These results suggest that, following chronic treatment, resistance to HDM2 antagonists may occur in the clinic. It will be important to combine HDM2 antagonists with other anti-cancer drugs early on during the treatment cycle in order to minimize the emergence of resistance. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1536.
- Published
- 2010
20. Abstract 4534: A novel and potent small molecule antagonist of HDM2, SCH 1450206, activates the p53 pathway with mechanism-based activity
- Author
-
Yinghui Lin, Cynthia Seidel-Dugan, Daniel J. Hicklin, Yao Ma, Suxing Liu, Asra Mirza, W. Robert Bishop, Doll Ronald J, Philip Lipari, Gerald W. Shipps, Yaolin Wang, Xiaoying Wang, and Stephane L. Bogen
- Subjects
Cancer Research ,Cell cycle checkpoint ,Cell growth ,Antagonist ,Cancer ,Biology ,Pharmacology ,medicine.disease ,Small hairpin RNA ,Oncology ,Mechanism of action ,Apoptosis ,Cell culture ,Cancer research ,medicine ,medicine.symptom - Abstract
p53 is an attractive therapeutic target in oncology since aberrant regulation of p53 expression is associated with cancer development and progression. HDM2 is a primary negative regulator of p53 function in cells. The blockade of the p53-HDM2 interaction presents an attractive approach for development of drugs against tumors expressing wild-type p53. Here we report a novel and potent small molecule antagonist of HDM2, SCH 1450206. It binds selectively to the HDM2 protein with high affinity, with weak affinity to HDMX protein. SCH 1450206 induced dose- and time-dependent stabilization of p53 protein and its downstream targets in several human cancer cell lines with wild-type p53, but not in cell with p53 gene deletion. SCH 1450206-induced stabilization of p53 led to distinct changes in expression of p53 target genes regulating apoptosis and cell cycle checkpoints. Inhibition of cell proliferation and induction of apoptosis were surveyed in a panel of human tumor cell lines following SCH 1450206 treatment. Inactivation of p53 by shRNA in cell lines expressing wt-p53 abolished the inhibition of cell proliferation and appearance of senescence in response to SCH 1450206, suggesting the cellular activity of SCH 1450206 is p53 specific and mechanism-based. Taken together, our results demonstrated the mechanism of action of SCH 1450206, a novel HDM2 antagonist that can activate the p53 pathway, and offers a potential anti-tumor agent targeting tumors expressing wild-type p53. Note: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4534.
- Published
- 2010
21. Abstract 4533: In vivo activation of the p53 pathway leading to tumor regression by a novel and potent HDM2 antagonist SCH 1450206
- Author
-
Asra Mirza, Ming Liu, Xiaoying Wang, Stephane L. Bogen, Yao Ma, Philip Lipari, Walter Robert Bishop, Cynthia Seidel-Dugan, Yinghui Liu, Doll Ronald J, Gerald W. Shipps, Yaolin Wang, Daniel J. Hicklin, Suxing Liu, and Abdul-Samad Ponery
- Subjects
Cancer Research ,Chemistry ,Antagonist ,Cancer ,Pharmacology ,medicine.disease ,Small molecule ,In vitro ,Oncology ,Oral administration ,In vivo ,Toxicity ,medicine ,Osteosarcoma - Abstract
The HDM2-p53 protein-protein interaction is well characterized through X-ray crystallography. Disrupting this HDM2-p53 protein-protein interaction by a small molecule would therefore release the p53 from the negative inhibition of HDM2 and restore its anti-tumor activities. Peptides, antisense oligonucleotides and small molecules have been identified which disrupt this interaction and result in the stabilization of p53 protein and activation of its downstream targets. We have recently discovered a novel, potent HDM2 antagonist (SCH 1450206) with mechanism-based activity both in vitro and in vivo. Oral administration of SCH 1450206 as a single agent resulted in tumor regression in the SJSA-1 osteosarcoma xenograft model without any observable toxicity. Analysis of the pharmacodynamic markers demonstrated that the anti-tumor activity of SCH 1450206 correlated with the robust activation of p53 pathway in vivo. In addition to its single agent anti-tumor activity, combination of HDM2 antagonist SCH 1450206 with various cytotoxics resulted in further tumor growth inhibition in various human cancer xenograft models. The activation of p53 pathway in vivo targets preferentially to tumor tissues compared to high proliferating and radio sensitive organs of the mouse at the efficacious dose. The lack of single agent toxicity at the efficacious dose and schedule would potentially allow the combination of this type of HDM2 antagonist with other anti-cancer agents at full dose in the clinic. Note: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4533.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.