Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Oncotarget ; 8(17): 28575-28587, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28212573

ABSTRACT

Small cell lung cancer (SCLC) is one of the most aggressive forms of cancer, with a 5-year survival <7%. A major barrier to progress is the absence of predictive biomarkers for chemotherapy and novel targeted agents such as PARP inhibitors. Using a high-throughput, integrated proteomic, transcriptomic, and genomic analysis of SCLC patient-derived xenografts (PDXs) and profiled cell lines, we identified biomarkers of drug sensitivity and determined their prevalence in patient tumors. In contrast to breast and ovarian cancer, PARP inhibitor response was not associated with mutations in homologous recombination (HR) genes (e.g., BRCA1/2) or HRD scores. Instead, we found several proteomic markers that predicted PDX response, including high levels of SLFN11 and E-cadherin and low ATM. SLFN11 and E-cadherin were also significantly associated with in vitro sensitivity to cisplatin and topoisomerase1/2 inhibitors (all commonly used in SCLC). Treatment with cisplatin or PARP inhibitors downregulated SLFN11 and E-cadherin, possibly explaining the rapid development of therapeutic resistance in SCLC. Supporting their functional role, silencing SLFN11 reduced in vitro sensitivity and drug-induced DNA damage; whereas ATM knockdown or pharmacologic inhibition enhanced sensitivity. Notably, SCLC with mesenchymal phenotypes (i.e., loss of E-cadherin and high epithelial-to-mesenchymal transition (EMT) signature scores) displayed striking alterations in expression of miR200 family and key SCLC genes (e.g., NEUROD1, ASCL1, ALDH1A1, MYCL1). Thus, SLFN11, EMT, and ATM mediate therapeutic response in SCLC and warrant further clinical investigation as predictive biomarkers.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Nuclear Proteins/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Ataxia Telangiectasia Mutated Proteins/metabolism , Biomarkers , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage , Drug Resistance, Neoplasm/genetics , Gene Expression , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism
2.
J Med Chem ; 59(1): 335-57, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26652717

ABSTRACT

We discovered and developed a novel series of tetrahydropyridophthlazinones as poly(ADP-ribose) polymerase (PARP) 1 and 2 inhibitors. Lead optimization led to the identification of (8S,9R)-47 (talazoparib; BMN 673; (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-(1-methyl-1H-1,2,4-triazol-5-yl)-2,7,8,9-tetrahydro-3H-pyrido[4,3,2-de]phthalazin-3-one). The novel stereospecific dual chiral-center-embedded structure of this compound has enabled extensive and unique binding interactions with PARP1/2 proteins. (8S,9R)-47 demonstrates excellent potency, inhibiting PARP1 and PARP2 enzyme activity with Ki = 1.2 and 0.87 nM, respectively. It inhibits PARP-mediated PARylation in a whole-cell assay with an EC50 of 2.51 nM and prevents proliferation of cancer cells carrying mutant BRCA1/2, with EC50 = 0.3 nM (MX-1) and 5 nM (Capan-1), respectively. (8S,9R)-47 is orally available, displaying favorable pharmacokinetic (PK) properties and remarkable antitumor efficacy in the BRCA1 mutant MX-1 breast cancer xenograft model following oral administration as a single-agent or in combination with chemotherapy agents such as temozolomide and cisplatin. (8S,9R)-47 has completed phase 1 clinical trial and is currently being studied in phase 2 and 3 clinical trials for the treatment of locally advanced and/or metastatic breast cancer with germline BRCA1/2 deleterious mutations.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Phthalazines/chemical synthesis , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/drug effects , Animals , Antineoplastic Agents/pharmacokinetics , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/genetics , BRCA2 Protein/antagonists & inhibitors , BRCA2 Protein/genetics , Cell Proliferation/drug effects , Clinical Trials, Phase I as Topic , Drug Discovery , Female , Germ-Line Mutation , Humans , Kinetics , Mice , Mice, Nude , Models, Molecular , Phthalazines/pharmacokinetics , Poly (ADP-Ribose) Polymerase-1 , Protein Binding , Rats , Stereoisomerism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 19(18): 5003-15, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23881923

ABSTRACT

PURPOSE: PARP1/2 inhibitors are a class of anticancer agents that target tumor-specific defects in DNA repair. Here, we describe BMN 673, a novel, highly potent PARP1/2 inhibitor with favorable metabolic stability, oral bioavailability, and pharmacokinetic properties. EXPERIMENTAL DESIGN: Potency and selectivity of BMN 673 was determined by biochemical assays. Anticancer activity either as a single-agent or in combination with other antitumor agents was evaluated both in vitro and in xenograft cancer models. RESULTS: BMN 673 is a potent PARP1/2 inhibitor (PARP1 IC50 = 0.57 nmol/L), but it does not inhibit other enzymes that we have tested. BMN 673 exhibits selective antitumor cytotoxicity and elicits DNA repair biomarkers at much lower concentrations than earlier generation PARP1/2 inhibitors (such as olaparib, rucaparib, and veliparib). In vitro, BMN 673 selectively targeted tumor cells with BRCA1, BRCA2, or PTEN gene defects with 20- to more than 200-fold greater potency than existing PARP1/2 inhibitors. BMN 673 is readily orally bioavailable, with more than 40% absolute oral bioavailability in rats when dosed in carboxylmethyl cellulose. Oral administration of BMN 673 elicited remarkable antitumor activity in vivo; xenografted tumors that carry defects in DNA repair due to BRCA mutations or PTEN deficiency were profoundly sensitive to oral BMN 673 treatment at well-tolerated doses in mice. Synergistic or additive antitumor effects were also found when BMN 673 was combined with temozolomide, SN38, or platinum drugs. CONCLUSION: BMN 673 is currently in early-phase clinical development and represents a promising PARP1/2 inhibitor with potentially advantageous features in its drug class.


Subject(s)
Breast Neoplasms/drug therapy , DNA Repair-Deficiency Disorders/drug therapy , Drug Resistance, Neoplasm/drug effects , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Female , Flow Cytometry , Humans , Mice , Mice, Nude , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/genetics , Rats , Tumor Cells, Cultured , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
4.
Cancer Res ; 64(19): 7099-109, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466206

ABSTRACT

The RAS/RAF signaling pathway is an important mediator of tumor cell proliferation and angiogenesis. The novel bi-aryl urea BAY 43-9006 is a potent inhibitor of Raf-1, a member of the RAF/MEK/ERK signaling pathway. Additional characterization showed that BAY 43-9006 suppresses both wild-type and V599E mutant BRAF activity in vitro. In addition, BAY 43-9006 demonstrated significant activity against several receptor tyrosine kinases involved in neovascularization and tumor progression, including vascular endothelial growth factor receptor (VEGFR)-2, VEGFR-3, platelet-derived growth factor receptor beta, Flt-3, and c-KIT. In cellular mechanistic assays, BAY 43-9006 demonstrated inhibition of the mitogen-activated protein kinase pathway in colon, pancreatic, and breast tumor cell lines expressing mutant KRAS or wild-type or mutant BRAF, whereas non-small-cell lung cancer cell lines expressing mutant KRAS were insensitive to inhibition of the mitogen-activated protein kinase pathway by BAY 43-9006. Potent inhibition of VEGFR-2, platelet-derived growth factor receptor beta, and VEGFR-3 cellular receptor autophosphorylation was also observed for BAY 43-9006. Once daily oral dosing of BAY 43-9006 demonstrated broad-spectrum antitumor activity in colon, breast, and non-small-cell lung cancer xenograft models. Immunohistochemistry demonstrated a close association between inhibition of tumor growth and inhibition of the extracellular signal-regulated kinases (ERKs) 1/2 phosphorylation in two of three xenograft models examined, consistent with inhibition of the RAF/MEK/ERK pathway in some but not all models. Additional analyses of microvessel density and microvessel area in the same tumor sections using antimurine CD31 antibodies demonstrated significant inhibition of neovascularization in all three of the xenograft models. These data demonstrate that BAY 43-9006 is a novel dual action RAF kinase and VEGFR inhibitor that targets tumor cell proliferation and tumor angiogenesis.


Subject(s)
Benzenesulfonates/pharmacology , MAP Kinase Kinase Kinase 1 , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/enzymology , Pyridines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Administration, Oral , Animals , Cell Line, Tumor , Disease Progression , Female , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Sorafenib , Xenograft Model Antitumor Assays
5.
Curr Opin Investig Drugs ; 4(12): 1436-41, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14763129

ABSTRACT

Recognition of the importance of the Raf pathway in the proliferation and survival of tumor cells recently increased with the discovery of activating BRAF mutations in human tumors. Therefore, in addition to a role in controlling tumors with Ras mutations and activated growth factor receptors, inhibitors of the Raf pathway may harbor therapeutic potential in tumors carrying a BRAF oncogene. A variety of agents have been discovered that interfere with the Raf pathway, including antisense oligonucleotides and small molecules. These inhibitors block the expression of Raf protein, block Ras/Raf interaction, block its kinase activity, or block the kinase activity of the Raf target protein mitogen-activated protein kinase kinase. Raf pathway inhibitors that are currently undergoing clinical evaluation show promising signs of anticancer efficacy with a very tolerable safety profile. Indeed, the Raf inhibitor BAY-43-9006 recently entered phase III clinical trials. Here, we review the current development status of potential Raf pathway therapeutics.


Subject(s)
Enzyme Inhibitors/therapeutic use , MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Animals , Benzenesulfonates/chemistry , Benzenesulfonates/pharmacology , Benzenesulfonates/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Signaling System/physiology , Neoplasms/enzymology , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pyridines/chemistry , Pyridines/pharmacology , Pyridines/therapeutic use , Sorafenib
6.
Curr Opin Investig Drugs ; 3(12): 1768-72, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12528315

ABSTRACT

Adenoviruses can be engineered to replicate selectively in tumor cells but inefficiently in normal cells. ONYX-015 (CI-1042, dl1520; Onyx Pharmaceuticals Inc), which replicates selectively in cells deficient in the p53 pathway, was the first such adenovirus to reach clinical testing. Multiple trials of ONYX-015 in over 300 cancer patients, and trials with other selectively replicating adenoviruses, have established the safety of this approach. Evidence of anticancer activity in patients is encouraging. Recently, the first clinical trial of a selectively replicating adenovirus carrying an inserted transgene was reported. Adenoviruses with improved efficiency of replication, technologies for use of the viruses as vectors for anticancer gene therapy, and various other approaches, provide promising directions to develop selectively replicating adenoviruses into systemic therapy for metastatic cancer.


Subject(s)
Adenoviridae/genetics , Clinical Trials as Topic/statistics & numerical data , Neoplasms/drug therapy , Technology, Pharmaceutical/methods , Animals , Humans , Neoplasms/genetics , Neoplasms/virology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL