Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(16): 11177-11186, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35930799

ABSTRACT

Bromodomains are acetyllysine recognition domains present in a variety of human proteins. Bromodomains also bind small molecules that compete with acetyllysine, and therefore bromodomains have been targets for drug discovery efforts. Highly potent and selective ligands with good cellular permeability have been proposed as chemical probes for use in exploring the functions of many of the bromodomain proteins. We report here the discovery of a class of such inhibitors targeting the family VIII bromodomains of SMARCA2 (BRM) and SMARCA4 (BRG1), and PBRM1 (polybromo-1) bromodomain 5. We propose one example from this series, GNE-064, as a chemical probe for the bromodomains SMARCA2, SMARCA4, and PBRM1(5) with the potential for in vivo use.


Subject(s)
DNA Helicases , Transcription Factors , DNA-Binding Proteins , Humans , Nuclear Proteins , Protein Domains
2.
iScience ; 13: 488-497, 2019 Mar 29.
Article in English | MEDLINE | ID: mdl-30898619

ABSTRACT

Werner syndrome protein (WRN) is a RecQ enzyme involved in the maintenance of genome integrity. Germline loss-of-function mutations in WRN led to premature aging and predisposition to cancer. We evaluated synthetic lethal (SL) interactions between WRN and another human RecQ helicase, BLM, with DNA damage response genes in cancer cell lines. We found that WRN was SL with a DNA mismatch repair protein MutL homolog 1, loss of which is associated with high microsatellite instability (MSI-H). MSI-H cells exhibited increased double-stranded DNA breaks, altered cell cycles, and decreased viability in response to WRN knockdown, in contrast to microsatellite stable (MSS) lines, which tolerated depletion of WRN. Although WRN is the only human RecQ enzyme with a distinct exonuclease domain, only loss of helicase activity drives the MSI SL interaction. This SL interaction in MSI cancer cells positions WRN as a relevant therapeutic target in patients with MSI-H tumors.

3.
J Med Chem ; 61(6): 2227-2245, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29457982

ABSTRACT

Bruton's tyrosine kinase (Btk) is a nonreceptor cytoplasmic tyrosine kinase involved in B-cell and myeloid cell activation, downstream of B-cell and Fcγ receptors, respectively. Preclinical studies have indicated that inhibition of Btk activity might offer a potential therapy in autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus. Here we disclose the discovery and preclinical characterization of a potent, selective, and noncovalent Btk inhibitor currently in clinical development. GDC-0853 (29) suppresses B cell- and myeloid cell-mediated components of disease and demonstrates dose-dependent activity in an in vivo rat model of inflammatory arthritis. It demonstrates highly favorable safety, pharmacokinetic (PK), and pharmacodynamic (PD) profiles in preclinical and Phase 2 studies ongoing in patients with rheumatoid arthritis, lupus, and chronic spontaneous urticaria. On the basis of its potency, selectivity, long target residence time, and noncovalent mode of inhibition, 29 has the potential to be a best-in-class Btk inhibitor for a wide range of immunological indications.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Agammaglobulinaemia Tyrosine Kinase/drug effects , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/toxicity , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Dogs , Drug Discovery , Humans , Lupus Erythematosus, Systemic/drug therapy , Madin Darby Canine Kidney Cells , Models, Molecular , Molecular Structure , Piperazines/pharmacokinetics , Piperazines/toxicity , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/toxicity , Pyridones/pharmacokinetics , Pyridones/toxicity , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley
4.
ACS Chem Biol ; 11(10): 2897-2907, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27571029

ABSTRACT

The Bruton's tyrosine kinase (Btk) inhibitor ibrutinib has shown impressive clinical efficacy in a range of B-cell malignancies. However, acquired resistance has emerged, and second generation therapies are now being sought. Ibrutinib is a covalent, irreversible inhibitor that modifies Cys481 in the ATP binding site of Btk and renders the enzyme inactive, thereby blocking B-cell receptor signal transduction. Not surprisingly, Cys481 is the most commonly mutated Btk residue in cases of acquired resistance to ibrutinib. Mutations at other sites, including Thr474, a gatekeeper residue, have also been detected. Herein, we describe noncovalent Btk inhibitors that differ from covalent inhibitors like ibrutinib in that they do not interact with Cys481, they potently inhibit the ibrutinib-resistant Btk C481S mutant in vitro and in cells, and they are exquisitely selective for Btk. Noncovalent inhibitors such as GNE-431 also show excellent potency against the C481R, T474I, and T474M mutants. X-ray crystallographic analysis of Btk provides insight into the unique mode of binding of these inhibitors that explains their high selectivity for Btk and their retained activity against mutant forms of Btk. This class of noncovalent Btk inhibitors may provide a treatment option to patients, especially those who have acquired resistance to ibrutinib by mutation of Cys481 or Thr474.


Subject(s)
Cysteine/genetics , Mutation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Threonine/genetics , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Kinetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Piperidines , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use
5.
Mol Biol Cell ; 27(6): 878-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26976931
6.
Mol Cancer Ther ; 15(5): 1132-44, 2016 05.
Article in English | MEDLINE | ID: mdl-26939706

ABSTRACT

BCL-2 family proteins dictate survival of human multiple myeloma cells, making them attractive drug targets. Indeed, multiple myeloma cells are sensitive to antagonists that selectively target prosurvival proteins such as BCL-2/BCL-XL (ABT-737 and ABT-263/navitoclax) or BCL-2 only (ABT-199/GDC-0199/venetoclax). Resistance to these three drugs is mediated by expression of MCL-1. However, given the selectivity profile of venetoclax it is unclear whether coexpression of BCL-XL also affects antitumor responses to venetoclax in multiple myeloma. In multiple myeloma cell lines (n = 21), BCL-2 is expressed but sensitivity to venetoclax correlated with high BCL-2 and low BCL-XL or MCL-1 expression. Multiple myeloma cells that coexpress BCL-2 and BCL-XL were resistant to venetoclax but sensitive to a BCL-XL-selective inhibitor (A-1155463). Multiple myeloma xenograft models that coexpressed BCL-XL or MCL-1 with BCL-2 were also resistant to venetoclax. Resistance to venetoclax was mitigated by cotreatment with bortezomib in xenografts that coexpressed BCL-2 and MCL-1 due to upregulation of NOXA, a proapoptotic factor that neutralizes MCL-1. In contrast, xenografts that expressed BCL-XL, MCL-1, and BCL-2 were more sensitive to the combination of bortezomib with a BCL-XL selective inhibitor (A-1331852) but not with venetoclax cotreatment when compared with monotherapies. IHC of multiple myeloma patient bone marrow biopsies and aspirates (n = 95) revealed high levels of BCL-2 and BCL-XL in 62% and 43% of evaluable samples, respectively, while 34% were characterized as BCL-2(High)/BCL-XL (Low) In addition to MCL-1, our data suggest that BCL-XL may also be a potential resistance factor to venetoclax monotherapy and in combination with bortezomib. Mol Cancer Ther; 15(5); 1132-44. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Gene Expression Regulation, Neoplastic , Multiple Myeloma/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Sulfonamides/pharmacology , bcl-X Protein/genetics , Animals , Bcl-2-Like Protein 11/metabolism , Bortezomib/pharmacology , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Drug Therapy, Combination , Humans , Immunohistochemistry , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Xenograft Model Antitumor Assays
7.
Sci Transl Med ; 7(279): 279ra40, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25787766

ABSTRACT

The BCL-2/BCL-XL/BCL-W inhibitor ABT-263 (navitoclax) has shown promising clinical activity in lymphoid malignancies such as chronic lymphocytic leukemia. However, its efficacy in these settings is limited by thrombocytopenia caused by BCL-XL inhibition. This prompted the generation of the BCL-2-selective inhibitor venetoclax (ABT-199/GDC-0199), which demonstrates robust activity in these cancers but spares platelets. Navitoclax has also been shown to enhance the efficacy of docetaxel in preclinical models of solid tumors, but clinical use of this combination has been limited by neutropenia. We used venetoclax and the BCL-XL-selective inhibitors A-1155463 and A-1331852 to assess the relative contributions of inhibiting BCL-2 or BCL-XL to the efficacy and toxicity of the navitoclax-docetaxel combination. Selective BCL-2 inhibition suppressed granulopoiesis in vitro and in vivo, potentially accounting for the exacerbated neutropenia observed when navitoclax was combined with docetaxel clinically. By contrast, selectively inhibiting BCL-XL did not suppress granulopoiesis but was highly efficacious in combination with docetaxel when tested against a range of solid tumors. Therefore, BCL-XL-selective inhibitors have the potential to enhance the efficacy of docetaxel in solid tumors and avoid the exacerbation of neutropenia observed with navitoclax. These studies demonstrate the translational utility of this toolkit of selective BCL-2 family inhibitors and highlight their potential as improved cancer therapeutics.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Administration, Oral , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Benzothiazoles/chemistry , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cell Line, Tumor , Cell Survival , Docetaxel , Gene Expression Profiling , Granulocytes/metabolism , Humans , Isoquinolines/chemistry , Kinetics , Mice , Neoplasm Transplantation , Neoplasms/metabolism , Neutropenia/chemically induced , Neutrophils/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/therapeutic use , Taxoids/adverse effects , Thrombocytopenia/chemically induced , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/metabolism
8.
Biochem J ; 466(1): 45-54, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25423073

ABSTRACT

Evasion of cell death is one crucial capability acquired by tumour cells to ward-off anti-tumour therapies and represents a fundamental challenge to sustaining clinical efficacy for currently available agents. Inhibitor of apoptosis (IAP) proteins use their ubiquitin E3 ligase activity to promote cancer cell survival by mediating proliferative signalling and blocking cell death in response to diverse stimuli. Using immunoaffinity enrichment and MS, ubiquitination sites on thousands of proteins were profiled upon initiation of cell death by IAP antagonists in IAP antagonist-sensitive and -resistant breast cancer cell lines. Our analyses identified hundreds of proteins with elevated levels of ubiquitin-remnant [K-GG (Lys-Gly-Gly)] peptides upon activation of cell death by the IAP antagonist BV6. The majority of these were observed in BV6-sensitive, but not-resistant, cells. Among these were known pro-apoptotic regulators, including CYC (cytochrome c), RIP1 (receptor-interacting protein 1) and a selection of proteins known to reside in the mitochondria or regulate NF-κB (nuclear factor κB) signalling. Analysis of early time-points revealed that IAP antagonist treatment stimulated rapid ubiquitination of NF-κB signalling proteins, including TRAF2 [TNF (tumour necrosis factor) receptor-associated factor 2], HOIL-1 (haem-oxidized iron-regulatory protein 2 ubiquitin ligase-1), NEMO (NF-κB essential modifier), as well as c-IAP1 (cellular IAP1) auto-ubiquitination. Knockdown of several NF-κB pathway members reduced BV6-induced cell death and TNF production in sensitive cell lines. Importantly, RIP1 was found to be constitutively ubiquitinated in sensitive breast-cancer cell lines at higher basal level than in resistant cell lines. Together, these data show the diverse and temporally defined roles of protein ubiquitination following IAP-antagonist treatment and provide critical insights into predictive diagnostics that may enhance clinical efficacy.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Inhibitor of Apoptosis Proteins/genetics , Oligopeptides/pharmacology , Ubiquitin/genetics , Cell Line, Tumor , Cytochromes c/genetics , Cytochromes c/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression Profiling , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Transcription Factors , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
PLoS One ; 9(10): e109366, 2014.
Article in English | MEDLINE | ID: mdl-25285661

ABSTRACT

Inhibiting NAD biosynthesis by blocking the function of nicotinamide phosphoribosyl transferase (NAMPT) is an attractive therapeutic strategy for targeting tumor metabolism. However, the development of drug resistance commonly limits the efficacy of cancer therapeutics. This study identifies mutations in NAMPT that confer resistance to a novel NAMPT inhibitor, GNE-618, in cell culture and in vivo, thus demonstrating that the cytotoxicity of GNE-618 is on target. We determine the crystal structures of six NAMPT mutants in the apo form and in complex with various inhibitors and use cellular, biochemical and structural data to elucidate two resistance mechanisms. One is the surprising finding of allosteric modulation by mutation of residue Ser165, resulting in unwinding of an α-helix that binds the NAMPT substrate 5-phosphoribosyl-1-pyrophosphate (PRPP). The other mechanism is orthosteric blocking of inhibitor binding by mutations of Gly217. Furthermore, by evaluating a panel of diverse small molecule inhibitors, we unravel inhibitor structure activity relationships on the mutant enzymes. These results provide valuable insights into the design of next generation NAMPT inhibitors that offer improved therapeutic potential by evading certain mechanisms of resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Cytokines/antagonists & inhibitors , Cytokines/chemistry , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/chemistry , Catalytic Domain , Cell Line, Tumor , Cytokines/genetics , Humans , Models, Molecular , Mutation , Nicotinamide Phosphoribosyltransferase/genetics
10.
Neoplasia ; 15(10): 1151-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204194

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) is a critical metabolite that is required for a range of cellular reactions. A key enzyme in the NAD salvage pathway is nicotinamide phosphoribosyl transferase (NAMPT), and here, we describe GNE-618, an NAMPT inhibitor that depletes NAD and induces cell death in vitro and in vivo. While cells proficient for nicotinic acid phosphoribosyl transferase (NAPRT1) can be protected from NAMPT inhibition as they convert nicotinic acid (NA) to NAD independent of the salvage pathway, this protection only occurs if NA is added before NAD depletion. We also demonstrate that tumor cells are unable to generate NAD by de novo synthesis as they lack expression of key enzymes in this pathway, thus providing a mechanistic rationale for the reliance of tumor cells on the NAD salvage pathway. Identifying tumors that are sensitive to NAMPT inhibition is one potential way to enhance the therapeutic effectiveness of an NAMPT inhibitor, and here, we show that NAMPT, but not NAPRT1, mRNA and protein levels inversely correlate with sensitivity to GNE-618 across a panel of 53 non-small cell lung carcinoma cell lines. Finally, we demonstrate that GNE-618 reduced tumor growth in a patient-derived model, which is thought to more closely represent heterogeneous primary patient tumors. Thus, we show that dependence of tumor cells on the NAD salvage pathway renders them sensitive to GNE-618 in vitro and in vivo, and our data support further evaluation of the use of NAMPT mRNA and protein levels as predictors of overall sensitivity.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , NAD/metabolism , Pentosyltransferases/antagonists & inhibitors , Pyrazoles/pharmacology , Sulfones/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Death/drug effects , Cell Line, Tumor , Heterografts , Humans , Lung Neoplasms/drug therapy , Mice, Nude , Pentosyltransferases/genetics , Pyrazoles/therapeutic use , Sulfones/therapeutic use
11.
Clin Cancer Res ; 19(24): 6912-23, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24097869

ABSTRACT

PURPOSE: We sought to identify predictive biomarkers for a novel nicotinamide phosphoribosyltransferase (NAMPT) inhibitor. EXPERIMENTAL DESIGN: We use a NAMPT inhibitor, GNE-617, to evaluate nicotinic acid rescue status in a panel of more than 400 cancer cell lines. Using correlative analysis and RNA interference (RNAi), we identify a specific biomarker for nicotinic acid rescue status. We next determine the mechanism of regulation of expression of the biomarker. Finally, we develop immunohistochemical (IHC) and DNA methylation assays and evaluate cancer tissue for prevalence of the biomarker across indications. RESULTS: Nicotinate phosphoribosyltransferase (NAPRT1) is necessary for nicotinic acid rescue and its expression is the major determinant of rescue status. We demonstrate that NAPRT1 promoter methylation accounts for NAPRT1 deficiency in cancer cells, and NAPRT1 methylation is predictive of rescue status in cancer cell lines. Bisulfite next-generation sequencing mapping of the NAPRT1 promoter identified tumor-specific sites of NAPRT1 DNA methylation and enabled the development of a quantitative methylation-specific PCR (QMSP) assay suitable for use on archival formalin-fixed paraffin-embedded tumor tissue. CONCLUSIONS: Tumor-specific promoter hypermethylation of NAPRT1 inactivates one of two NAD salvage pathways, resulting in synthetic lethality with the coadministration of a NAMPT inhibitor. NAPRT1 expression is lost due to promoter hypermethylation in most cancer types evaluated at frequencies ranging from 5% to 65%. NAPRT1-specific immunohistochemical or DNA methylation assays can be used on archival formalin paraffin-embedded cancer tissue to identify patients likely to benefit from coadministration of a Nampt inhibitor and nicotinic acid.


Subject(s)
Cytokines/metabolism , Heterocyclic Compounds, 2-Ring/administration & dosage , Neoplasms/genetics , Niacin/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Pentosyltransferases/metabolism , Sulfones/administration & dosage , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cytokines/antagonists & inhibitors , Cytokines/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/pathology , Niacin/administration & dosage , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/genetics , Pentosyltransferases/antagonists & inhibitors , Pentosyltransferases/deficiency , Promoter Regions, Genetic
12.
Mol Cancer Ther ; 12(6): 853-64, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23475955

ABSTRACT

Although mitogen-activated protein (MAP)-extracellular signal-regulated kinase (ERK) kinase (MEK) inhibition is predicted to cause cell death by stabilization of the proapoptotic BH3-only protein BIM, the induction of apoptosis is often modest. To determine if addition of a Bcl-2 family inhibitor could increase the efficacy of a MEK inhibitor, we evaluated a panel of 53 non-small cell lung cancer and pancreatic cancer cell lines with the combination of navitoclax (ABT-263), a Bcl-2/Bcl-xL (BCL2/BCL2L1) antagonist, and a novel MAP kinase (MEK) inhibitor, G-963. The combination is synergistic in the majority of lines, with an enrichment of cell lines harboring KRAS mutations in the high synergy group. Cells exposed to G-963 arrest in G1 and a small fraction undergo apoptosis. The addition of navitoclax to G-963 does not alter the kinetics of cell-cycle arrest, but greatly increases the percentage of cells that undergo apoptosis. The G-963/navitoclax combination was more effective than either single agent in the KRAS mutant H2122 xenograft model; BIM stabilization and PARP cleavage were observed in tumors, consistent with the mechanism of action observed in cell culture. Addition of the phosphatidylinositol 3-kinase (PI3K, PIK3CA) inhibitor GDC-0941 to this treatment combination increases cell killing compared with double- or single-agent treatment. Taken together, these data suggest the efficacy of agents that target the MAPK and PI3K pathways can be improved by combination with a Bcl-2 family inhibitor.


Subject(s)
Lung Neoplasms/drug therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , bcl-X Protein/antagonists & inhibitors , Aniline Compounds/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Enzyme Inhibitors/administration & dosage , Genes, bcl-2/genetics , Humans , Indazoles/administration & dosage , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases/metabolism , Molecular Targeted Therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Sulfonamides/administration & dosage , bcl-X Protein/genetics
13.
Clin Cancer Res ; 18(14): 3901-11, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22586300

ABSTRACT

PURPOSE: Docetaxel is a front-line standard-of-care chemotherapeutic drug for the treatment of breast cancer. Phosphoinositide 3-kinases (PI3K) are lipid kinases that regulate breast tumor cell growth, migration, and survival. The current study was intended to determine whether GDC-0941, an orally bioavailable class I selective PI3K inhibitor, enhances the antitumor activity of docetaxel in human breast cancer models in vitro and in vivo. EXPERIMENTAL DESIGN: A panel of 25 breast tumor cell lines representing HER2+, luminal, and basal subtypes were treated with GDC-0941, docetaxel, or the combination of both drugs and assayed for cellular viability, modulation of PI3K pathway markers, and apoptosis induction. Drug combination effects on cellular viability were also assessed in nontransformed MCF10A human mammary epithelial cells. Human xenografts of breast cancer cell lines and patient-derived tumors were used to assess efficacy of GDC-0941 and docetaxel in vivo. RESULTS: Combination of GDC-0941 and docetaxel decreased the cellular viability of breast tumor cell lines in vitro but to variable degrees of drug synergy. Compared with nontransformed MCF10A cells, the addition of both drugs resulted in stronger synergistic effects in a subset of tumor cell lines that were not predicted by breast cancer subtype. In xenograft models, GDC-0941 enhanced the antitumor activity of docetaxel with maximum combination efficacy observed within 1 hour of administering both drugs. GDC-0941 increased the rate of apoptosis in cells arrested in mitosis upon cotreatment with docetaxel. CONCLUSION: GDC-0941 augments the efficacy of docetaxel by increasing drug-induced apoptosis in breast cancer models.


Subject(s)
Breast Neoplasms/drug therapy , Indazoles/administration & dosage , Phosphatidylinositol 3-Kinases , Sulfonamides/administration & dosage , Taxoids/administration & dosage , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Docetaxel , Drug Synergism , Female , Humans , Mice , Neoplasms, Experimental/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors
14.
Blood ; 119(24): 5807-16, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22538851

ABSTRACT

The BH3-mimetic ABT-737 and an orally bioavailable compound of the same class, navitoclax (ABT-263), have shown promising antitumor efficacy in preclinical and early clinical studies. Although both drugs avidly bind Bcl-2, Bcl-x(L), and Bcl-w in vitro, we find that Bcl-2 is the critical target in vivo, suggesting that patients with tumors overexpressing Bcl-2 will probably benefit. In human non-Hodgkin lymphomas, high expression of Bcl-2 but not Bcl-x(L) predicted sensitivity to ABT-263. Moreover, we show that increasing Bcl-2 sensitized normal and transformed lymphoid cells to ABT-737 by elevating proapoptotic Bim. In striking contrast, increasing Bcl-x(L) or Bcl-w conferred robust resistance to ABT-737, despite also increasing Bim. Cell-based protein redistribution assays unexpectedly revealed that ABT-737 disrupts Bcl-2/Bim complexes more readily than Bcl-x(L)/Bim or Bcl-w/Bim complexes. These results have profound implications for how BH3-mimetics induce apoptosis and how the use of these compounds can be optimized for treating lymphoid malignancies.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Biphenyl Compounds/pharmacology , Leukemia/drug therapy , Lymphoma/drug therapy , Molecular Targeted Therapy , Nitrophenols/pharmacology , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , Aniline Compounds/therapeutic use , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Biphenyl Compounds/therapeutic use , Cell Death/drug effects , Cytoprotection/drug effects , Drug Resistance, Neoplasm/drug effects , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia/genetics , Leukemia/pathology , Lymphoma/genetics , Lymphoma/pathology , Membrane Proteins/metabolism , Mice , Mutant Proteins/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Nitrophenols/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Binding/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/therapeutic use , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
15.
Mol Cancer Ther ; 11(4): 1026-35, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22302098

ABSTRACT

To examine the potential of combining Bcl-2 family inhibitors with chemotherapy in ovarian cancer, we evaluated a panel of 27 ovarian cancer cell lines for response to the combination of navitoclax (formerly ABT-263) and paclitaxel or gemcitabine. The majority of cell lines exhibited a greater than additive response to either combination, as determined by the Bliss independence model, and more than 50% of the ovarian cell lines exhibited strong synergy for the navitoclax/paclitaxel combination. To identify biomarkers for tumors likely to respond to this combination, we evaluated the protein levels of intrinsic apoptosis pathway components. Bcl-x(L) seems necessary, but not sufficient, for navitoclax/paclitaxel synergy in vitro, suggesting that exclusion of patients whose tumors have low or undetectable Bcl-x(L) would enrich for patients responsive to the combination. We evaluated Bcl-x(L) levels in ovarian cancer tumor tissue from 40 patients (20 taxane responsive and 20 with poor response to taxane) and found that patients with high Bcl-x(L) were less sensitive to taxane treatment (10 of 12) Bcl-x(L) positive patients, P = 0.014). These data support the use of navitoclax in combination with taxane-based therapy in ovarian cancer patients with high levels of Bcl-x(L).


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ovarian Neoplasms/drug therapy , Sulfonamides/pharmacology , bcl-X Protein/metabolism , Aniline Compounds/administration & dosage , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Synergism , Female , Humans , Immunohistochemistry , Ovarian Neoplasms/metabolism , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Sulfonamides/administration & dosage , Gemcitabine
16.
Nature ; 471(7336): 110-4, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368834

ABSTRACT

Microtubules have pivotal roles in fundamental cellular processes and are targets of antitubulin chemotherapeutics. Microtubule-targeted agents such as Taxol and vincristine are prescribed widely for various malignancies, including ovarian and breast adenocarcinomas, non-small-cell lung cancer, leukaemias and lymphomas. These agents arrest cells in mitosis and subsequently induce cell death through poorly defined mechanisms. The strategies that resistant tumour cells use to evade death induced by antitubulin agents are also unclear. Here we show that the pro-survival protein MCL1 (ref. 3) is a crucial regulator of apoptosis triggered by antitubulin chemotherapeutics. During mitotic arrest, MCL1 protein levels decline markedly, through a post-translational mechanism, potentiating cell death. Phosphorylation of MCL1 directs its interaction with the tumour-suppressor protein FBW7, which is the substrate-binding component of a ubiquitin ligase complex. The polyubiquitylation of MCL1 then targets it for proteasomal degradation. The degradation of MCL1 was blocked in patient-derived tumour cells that lacked FBW7 or had loss-of-function mutations in FBW7, conferring resistance to antitubulin agents and promoting chemotherapeutic-induced polyploidy. Additionally, primary tumour samples were enriched for FBW7 inactivation and elevated MCL1 levels, underscoring the prominent roles of these proteins in oncogenesis. Our findings suggest that profiling the FBW7 and MCL1 status of tumours, in terms of protein levels, messenger RNA levels and genetic status, could be useful to predict the response of patients to antitubulin chemotherapeutics.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tubulin Modulators/pharmacology , Tubulin/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Drug Resistance, Neoplasm , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Fibroblasts , Humans , Mice , Mitosis/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , Paclitaxel/pharmacology , Pharmacogenetics , Phosphorylation/drug effects , Polyploidy , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Vincristine/pharmacology
17.
Clin Cancer Res ; 17(6): 1394-404, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21220478

ABSTRACT

PURPOSE: To explore the potential of navitoclax in combination with taxane-based chemotherapy in the treatment of non-small cell lung cancer (NSCLC) by defining mechanism of synergy and identifying correlative biomarkers. EXPERIMENTAL DESIGN: We treated a panel of NSCLC lines with a dose matrix of paclitaxel and navitoclax (formerly ABT-263), an inhibitor of Bcl-2, Bcl-x(L), and Bcl-w (1), and evaluated synergy. We next used time-lapse microscopy to explore mechanism of synergy. Finally, we developed an immunohistochemical assay and assessed prevalence of Bcl-x(L) in NSCLC tumor tissues. RESULTS: All cell lines exhibit greater than additive response to the combination of navitoclax and a taxane. These results were extended to mouse xenograft tumor models, in which the combination is more efficacious than either single-agent docetaxel or navitoclax. Addition of navitoclax to paclitaxel decreases the time from mitotic entry to cell death and changes cell fate from mitotic slippage to death during mitotic arrest. The relative levels of Bcl-x(L) and Mcl-1 correlate with the extent of synergy, suggesting that cancers with elevated levels of Bcl-x(L) will be relatively resistant to taxane-based therapy but could benefit from the addition of navitoclax to taxane treatment. Finally, a significant percentage of NSCLC patient samples exhibit relatively high Bcl-x(L) levels. CONCLUSIONS: The addition of navitoclax to taxane-based chemotherapy in NSCLC has the potential to increase efficacy, particularly in patients whose tumors express high levels of Bcl-x(L).


Subject(s)
Aniline Compounds/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , Sulfonamides/pharmacology , Taxoids/chemistry , Aniline Compounds/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Cell Cycle , Cell Lineage , Cell Survival , Green Fluorescent Proteins/metabolism , Humans , Image Processing, Computer-Assisted , Mice , Mitosis , Neoplasm Transplantation , Paclitaxel/administration & dosage , Sulfonamides/administration & dosage , Time Factors , bcl-X Protein/antagonists & inhibitors
18.
Cancer Res ; 70(15): 6325-35, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20647325

ABSTRACT

Taxanes are very effective at causing mitotic arrest; however, there is variability among cancer cells in the apoptotic response to mitotic arrest. The variability in clinical efficacy of taxane-based therapy is likely a reflection of this variability in apoptotic response, thus elucidation of the molecular mechanism of the apoptotic response to mitotic stress could lead to improved clinical strategies. To identify genes whose expression influences the rate and extent of apoptosis after mitotic arrest, we screened a kinase-enriched small interfering RNA library for effects on caspase activation in response to maximally effective doses of paclitaxel, a PLK1 inhibitor, or cisplatin. Small interfering RNA oligonucleotides directed against an atypical protein kinase, TP53RK, caused the greatest increase in caspase-3/7 activation in response to antimitotic agents. Time-lapse microscopy revealed that cells entered mitosis with normal kinetics, but died after entry into mitosis in the presence of paclitaxel more rapidly when TP53RK was depleted. Because expression levels of TP53RK vary in cancers, TP53RK levels could provide a molecular marker to predict response to antimitotic agents. TP53RK inhibition may also sensitize cancers to taxanes.


Subject(s)
Apoptosis/physiology , Mitosis/physiology , Protein Kinases/physiology , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Apoptosis/drug effects , Cisplatin/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Screening Assays, Antitumor , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Mitosis/drug effects , Paclitaxel/pharmacology , Protein Kinases/deficiency , Protein Kinases/genetics , Protein Serine-Threonine Kinases , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection
19.
Proc Natl Acad Sci U S A ; 107(13): 5839-44, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20167803

ABSTRACT

Centromere-associated protein-E (CENP-E) is a kinetochore-associated mitotic kinesin that is thought to function as the key receptor responsible for mitotic checkpoint signal transduction after interaction with spindle microtubules. We have identified GSK923295, an allosteric inhibitor of CENP-E kinesin motor ATPase activity, and mapped the inhibitor binding site to a region similar to that bound by loop-5 inhibitors of the kinesin KSP/Eg5. Unlike these KSP inhibitors, which block release of ADP and destabilize motor-microtubule interaction, GSK923295 inhibited release of inorganic phosphate and stabilized CENP-E motor domain interaction with microtubules. Inhibition of CENP-E motor activity in cultured cells and tumor xenografts caused failure of metaphase chromosome alignment and induced mitotic arrest, indicating that tight binding of CENP-E to microtubules is insufficient to satisfy the mitotic checkpoint. Consistent with genetic studies in mice suggesting that decreased CENP-E function can have a tumor-suppressive effect, inhibition of CENP-E induced tumor cell apoptosis and tumor regression.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Sarcosine/analogs & derivatives , Allosteric Site , Animals , Antineoplastic Agents/chemistry , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Dogs , Drug Screening Assays, Antitumor , Humans , In Vitro Techniques , Kinesins/antagonists & inhibitors , Kinesins/chemistry , Kinesins/metabolism , Mice , Microtubules/metabolism , Mitosis/drug effects , Models, Molecular , Molecular Structure , Sarcosine/chemistry , Sarcosine/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...