Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
JCO Precis Oncol ; 8: e2300693, 2024 May.
Article in English | MEDLINE | ID: mdl-38754056

ABSTRACT

PURPOSE: To report the results of OPAL (ClinicalTrials.gov identifier: NCT03574779) cohort A, a single-arm substudy of niraparib plus dostarlimab and bevacizumab for the treatment of advanced, platinum-resistant ovarian cancer (PROC). METHODS: Participants with PROC who received 1-2 previous lines of therapy were treated with niraparib (200 or 300 mg once daily), dostarlimab (500 mg once every 3 weeks for four 21-day cycles, followed by 1,000 mg once every 6 weeks), and bevacizumab (15 mg/kg once every 3 weeks). The primary end point was investigator-assessed objective response rate (ORR) per RECIST v1.1. Safety was also assessed. Exploratory biomarker end points included evaluation of changes in the tumor molecular profile and microenvironment using baseline and on-treatment tumor samples. RESULTS: Of 41 enrolled participants (median age, 66.0 years [range, 37-83 years]), 9.8% had tumors that were BRCA-mutated, 19.5% were homologous recombination (HR)-deficient, and 17.1% were HR repair (HRR)-mutated. As of the cutoff date, all participants discontinued treatment. The ORR was 17.1% (80% CI, 9.8 to 27.0), including one complete response (2.4%); the disease control rate was 73.2% (80% CI, 62.3 to 82.2). Two participants withdrew before first postbaseline scan because of adverse events (AEs). Grade ≥3 treatment-emergent AEs were reported in 92.7% of participants, with the most common being hypertension (26.8%). Response was not correlated with BRCA, HRR, HR deficiency (HRD), or PD-L1 status. Changes suggesting immune activation were observed in on-treatment samples after triplet therapy. CONCLUSION: Results demonstrated modest activity of niraparib, dostarlimab, and bevacizumab in participants with PROC, many of whom had prognostic factors for poor treatment response. Most participants with response were bevacizumab-naïve. No association was found with HRD, BRCA, or PD-L1 status. AEs were consistent with previous monotherapy reports, except that hypertension was reported more frequently.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Bevacizumab , Drug Resistance, Neoplasm , Indazoles , Ovarian Neoplasms , Piperidines , Humans , Female , Middle Aged , Ovarian Neoplasms/drug therapy , Aged , Bevacizumab/therapeutic use , Adult , Indazoles/therapeutic use , Aged, 80 and over , Piperidines/therapeutic use , Piperidines/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Cohort Studies
2.
Trials ; 25(1): 301, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38702828

ABSTRACT

BACKGROUND: Maintenance therapy with niraparib, a poly(ADP-ribose) polymerase inhibitor, has been shown to extend progression-free survival in patients with newly diagnosed advanced ovarian cancer who responded to first-line platinum-based chemotherapy, regardless of biomarker status. However, there are limited data on niraparib's efficacy and safety in the neoadjuvant setting. The objective of Cohort C of the OPAL trial (OPAL-C) is to evaluate the efficacy, safety, and tolerability of neoadjuvant niraparib treatment compared with neoadjuvant platinum-taxane doublet chemotherapy in patients with newly diagnosed stage III/IV ovarian cancer with confirmed homologous recombination-deficient tumors. METHODS: OPAL is an ongoing global, multicenter, randomized, open-label, phase 2 trial. In OPAL-C, patients will be randomized 1:1 to receive three 21-day cycles of either neoadjuvant niraparib or platinum-taxane doublet neoadjuvant chemotherapy per standard of care. Patients with a complete or partial response per Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1) will then undergo interval debulking surgery; patients with stable disease may proceed to interval debulking surgery or alternative therapy at the investigator's discretion. Patients with disease progression will exit the study treatment and proceed to alternative therapy at the investigator's discretion. After interval debulking surgery, all patients will receive up to three 21-day cycles of platinum-taxane doublet chemotherapy followed by niraparib maintenance therapy for up to 36 months. Adult patients with newly diagnosed stage III/IV ovarian cancer eligible to receive neoadjuvant platinum-taxane doublet chemotherapy followed by interval debulking surgery may be enrolled. Patients must have tumors that are homologous recombination-deficient. The primary endpoint is the pre-interval debulking surgery unconfirmed overall response rate, defined as the investigator-assessed percentage of patients with unconfirmed complete or partial response on study treatment before interval debulking surgery per RECIST v1.1. DISCUSSION: OPAL-C explores the use of niraparib in the neoadjuvant setting as an alternative to neoadjuvant platinum-taxane doublet chemotherapy to improve postsurgical residual disease outcomes for patients with ovarian cancer with homologous recombination-deficient tumors. Positive findings from this approach could significantly impact preoperative ovarian cancer therapy, particularly for patients who are ineligible for primary debulking surgery. TRIAL REGISTRATION: ClinicalTrials.gov NCT03574779. Registered on February 28, 2022.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Indazoles , Neoadjuvant Therapy , Neoplasm Staging , Ovarian Neoplasms , Piperidines , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Neoadjuvant Therapy/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Piperidines/adverse effects , Piperidines/administration & dosage , Piperidines/therapeutic use , Indazoles/adverse effects , Indazoles/therapeutic use , Indazoles/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/adverse effects , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Multicenter Studies as Topic , Randomized Controlled Trials as Topic , Progression-Free Survival , Clinical Trials, Phase II as Topic , Homologous Recombination , Bridged-Ring Compounds/administration & dosage , Bridged-Ring Compounds/therapeutic use , Bridged-Ring Compounds/adverse effects , Piperazines/adverse effects , Piperazines/administration & dosage , Piperazines/therapeutic use , Time Factors
3.
Article in English | MEDLINE | ID: mdl-34527850

ABSTRACT

Poly (ADP-ribose) polymerase (PARP) inhibitors have emerged as promising therapy in cancers with homologous recombination repair deficiency. However, efficacy is limited by both intrinsic and acquired resistance. The Olaparib Combinations basket trial explored olaparib alone and in combination with other homologous recombination-directed targeted therapies. Here, we report the results of the arm in which olaparib was combined with the orally bioavailable ataxia telangiectasia and RAD3-related inhibitor ceralasertib in patients with relapsed or refractory cancers harboring DNA damage response and repair alterations, including patients with BRCA-mutated PARP inhibitor-resistant high-grade serous ovarian cancer (HGSOC). PATIENTS AND METHODS: Germline and somatic mutations had to be deleterious by COSMIC or ClinVar for eligibility. Olaparib was administered at 300 mg twice daily and ceralasertib at 160 mg daily on days 1-7 in 28-day cycles until progression or unacceptable toxicities. Primary end points were confirmed complete response (CR) or partial response (PR) rates and clinical benefit rate (CBR; CR + PR + stable disease [SD] at 16 weeks). RESULTS: Twenty-five patients were enrolled, with median four prior therapies. Five patients required dose reductions for myelosuppression. Overall response rate was 8.3% and CBR was 62.5% among the entire cohort. Two of five patients with tumor harboring ATM mutation achieved CR or SD ongoing at 24+ months, respectively (CBR 40%). Of seven patients with PARP inhibitor-resistant HGSOC, one achieved PR (-90%) and five had SD ranging 16-72 weeks (CBR 86%). CONCLUSION: Olaparib with ceralasertib demonstrated preliminary activity in ATM-mutated tumors and in PARP inhibitor-resistant BRCA1/2-mutated HGSOC. These data warrant additional studies to further confirm activity in these settings.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Indoles/administration & dosage , Morpholines/administration & dosage , Neoplasms/drug therapy , Phthalazines/administration & dosage , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , BRCA1 Protein/genetics , DNA Damage/drug effects , Female , Humans , Indoles/adverse effects , Male , Middle Aged , Morpholines/adverse effects , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/mortality , Phthalazines/adverse effects , Piperazines/adverse effects , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/adverse effects , Protein Kinase Inhibitors , Pyrimidines/adverse effects , Sulfonamides/adverse effects
4.
Clin Cancer Res ; 27(19): 5213-5224, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34301752

ABSTRACT

PURPOSE: This study reports the safety, tolerability, MTD, recommended phase II dose (RP2D), pharmacokinetic/pharmacodynamic profile, and preliminary antitumor activity of ceralasertib combined with carboplatin in patients with advanced solid tumors. It also examined exploratory predictive and pharmacodynamic biomarkers. PATIENTS AND METHODS: Eligible patients (n = 36) received a fixed dose of carboplatin (AUC5) with escalating doses of ceralasertib (20 mg twice daily to 60 mg once daily) in 21-day cycles. Sequential and concurrent combination dosing schedules were assessed. RESULTS: Two ceralasertib MTD dose schedules, 20 mg twice daily on days 4-13 and 40 mg once daily on days 1-2, were tolerated with carboplatin AUC5; the latter was declared the RP2D. The most common treatment-emergent adverse events (Common Terminology Criteria for Adverse Events grade ≥3) were anemia (39%), thrombocytopenia (36%), and neutropenia (25%). Dose-limiting toxicities of grade 4 thrombocytopenia (n = 2; including one grade 4 platelet count decreased) and a combination of grade 4 thrombocytopenia and grade 3 neutropenia occurred in 3 patients. Ceralasertib was quickly absorbed (tmax ∼1 hour), with a terminal plasma half-life of 8-11 hours. Upregulation of pRAD50, indicative of ataxia telangiectasia mutated (ATM) activation, was observed in tumor biopsies during ceralasertib treatment. Two patients with absent or low ATM or SLFN11 protein expression achieved confirmed RECIST v1.1 partial responses. Eighteen of 34 (53%) response-evaluable patients had RECIST v1.1 stable disease. CONCLUSIONS: The RP2D for ceralasertib plus carboplatin was established as ceralasertib 40 mg once daily on days 1-2 administered with carboplatin AUC5 every 3 weeks, with pharmacokinetic and pharmacodynamic studies confirming pharmacodynamic modulation and preliminary evidence of antitumor activity observed.


Subject(s)
Neoplasms , Neutropenia , Thrombocytopenia , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Ataxia Telangiectasia Mutated Proteins/genetics , Carboplatin , Humans , Indoles , Maximum Tolerated Dose , Morpholines , Neoplasms/etiology , Neutropenia/chemically induced , Nuclear Proteins , Protein Kinase Inhibitors/adverse effects , Pyrimidines , Sulfonamides , Sulfoxides , Thrombocytopenia/chemically induced
5.
Bioorg Med Chem Lett ; 26(4): 1314-8, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26786695

ABSTRACT

Inhibitors of the ATPase function of bacterial DNA gyrase, located in the GyrB subunit and its related ParE subunit in topoisomerase IV, have demonstrated antibacterial activity. In this study we describe an NMR fragment-based screening effort targeting Staphylococcus aureus GyrB that identified several attractive and novel starting points with good ligand efficiency. Fragment hits were further characterized using NMR binding studies against full-length S. aureus GyrB and Escherichia coli ParE. X-ray co-crystal structures of select fragment hits confirmed binding and suggested a path for medicinal chemistry optimization. The identification, characterization, and elaboration of one of these fragment series to a 0.265 µM inhibitor is described herein.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , DNA Gyrase/chemistry , Topoisomerase II Inhibitors/chemistry , Adenosine Triphosphatases/metabolism , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , DNA Gyrase/metabolism , DNA Topoisomerase IV/antagonists & inhibitors , DNA Topoisomerase IV/metabolism , Drug Design , Escherichia coli/metabolism , Ligands , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Protein Binding , Protein Structure, Tertiary , Staphylococcus aureus/enzymology , Topoisomerase II Inhibitors/metabolism
6.
J Med Chem ; 55(3): 1021-46, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22224594

ABSTRACT

Tissue transglutaminase 2 (TG2) is a multifunctional protein primarily known for its calcium-dependent enzymatic protein cross-linking activity via isopeptide bond formation between glutamine and lysine residues. TG2 overexpression and activity have been found to be associated with Huntington's disease (HD); specifically, TG2 is up-regulated in the brains of HD patients and in animal models of the disease. Interestingly, genetic deletion of TG2 in two different HD mouse models, R6/1 and R6/2, results in improved phenotypes including a reduction in neuronal death and prolonged survival. Starting with phenylacrylamide screening hit 7d, we describe the SAR of this series leading to potent and selective TG2 inhibitors. The suitability of the compounds as in vitro tools to elucidate the biology of TG2 was demonstrated through mode of inhibition studies, characterization of druglike properties, and inhibition profiles in a cell lysate assay.


Subject(s)
Acrylamides/chemical synthesis , GTP-Binding Proteins/antagonists & inhibitors , Huntington Disease/drug therapy , Sulfonamides/chemical synthesis , Transglutaminases/antagonists & inhibitors , Acrylamides/chemistry , Acrylamides/pharmacology , Animals , Caco-2 Cells , Cell Membrane Permeability , HEK293 Cells , Humans , In Vitro Techniques , Male , Mice , Microsomes, Liver/metabolism , Models, Molecular , Piperazines/chemical synthesis , Piperazines/chemistry , Piperazines/pharmacology , Protein Glutamine gamma Glutamyltransferase 2 , Pyridines/chemical synthesis , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology
7.
J Biol Chem ; 285(17): 12873-81, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20106978

ABSTRACT

Serine racemase is responsible for the synthesis of D-serine, an endogenous co-agonist for N-methyl-D-aspartate receptor-type glutamate receptors (NMDARs). This pyridoxal 5'-phosphate-dependent enzyme is involved both in the reversible conversion of L- to D-serine and serine catabolism by alpha,beta-elimination of water, thereby regulating D-serine levels. Because D-serine affects NMDAR signaling throughout the brain, serine racemase is a promising target for the treatment of disorders related to NMDAR dysfunction. To provide a molecular basis for rational drug design the x-ray crystal structures of human and rat serine racemase were determined at 1.5- and 2.1-A resolution, respectively, and in the presence and absence of the orthosteric inhibitor malonate. The structures revealed a fold typical of beta-family pyridoxal 5'-phosphate enzymes, with both a large domain and a flexible small domain associated into a symmetric dimer, and indicated a ligand-induced rearrangement of the small domain that organizes the active site for specific turnover of the substrate.


Subject(s)
Enzyme Inhibitors/chemistry , Malonates/chemistry , Protein Multimerization , Racemases and Epimerases/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Humans , Malonates/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Pyridoxal Phosphate/chemistry , Pyridoxal Phosphate/genetics , Pyridoxal Phosphate/metabolism , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/biosynthesis , Serine/genetics
8.
Protein Sci ; 19(1): 168-73, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19937655

ABSTRACT

The Mapkap kinases 2 and 3 (MK2 and MK3) have been implicated in intracellular signaling pathways leading to the production of the pro-inflammatory cytokine tumor necrosis factor alpha. MK2 has been pursued by the biopharmaceutical industry for many years for the development of a small molecule anti-inflammatory treatment and drug-like inhibitors have been described. The development of some of these compounds, however, has been slowed by the absence of a high-resolution crystal structure of MK2. Herein we present a high-resolution (1.9 A) crystal structure of the highly homologous MK3 in complex with a pharmaceutical lead compound. While all of the canonical features of Ser/Thr kinases in general and MK2 in particular are recapitulated in MK3, the detailed analysis of the binding interaction of the drug-like ligand within the adenine binding pocket allows relevant conclusions to be drawn for the further design of potent and selective drug candidates.


Subject(s)
Crystallography/methods , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Catalytic Domain , Humans , Intracellular Signaling Peptides and Proteins/genetics , Ligands , Models, Molecular , Protein Binding , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins
9.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 8): 872-4, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19622871

ABSTRACT

Protein crystallography has proven to be an effective method of obtaining high-resolution structures of protein-ligand complexes. However, in certain cases only apoprotein structures are readily available and the generation of crystal complexes is more problematic. Some crystallographic systems are not amenable to soaking of ligands owing to crystal-packing effects and many protein-ligand complexes do not crystallize under the same conditions as used for the apoprotein. Using crystals of human phosphodiesterase 10a (hPDE10a) as an example of such a challenging crystallographic system, the structure of the complex with papaverine was obtained to 2.8 A resolution using protein crystals cross-linked by glutaraldehyde prior to soaking of the ligand. Inspection of the electron-density maps suggested that the correct mode of binding was obtained in one of the two monomers in the asymmetric unit and inspection of crystal-packing contacts explained why cocrystallization experiments and soaking of crystals that were not cross-linked were unsuccessful.


Subject(s)
Multiprotein Complexes/chemistry , Papaverine/chemistry , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism , Crystallization , Crystallography, X-Ray , Glutaral/chemistry , Glutaral/metabolism , Humans , Ligands , Multiprotein Complexes/metabolism , Papaverine/metabolism , Phosphodiesterase Inhibitors/metabolism , Phosphoric Diester Hydrolases/metabolism , Protein Binding , Protein Conformation
10.
Bioorg Med Chem Lett ; 19(15): 4480-3, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19500983
11.
J Am Chem Soc ; 126(23): 7212-21, 2004 Jun 16.
Article in English | MEDLINE | ID: mdl-15186158

ABSTRACT

Incubation of farnesyl diphosphate (1) with Penicillium roqueforti aristolochene synthase yielded (+)-aristolochene (4), accompanied by minor quantities of the proposed intermediate (S)-(-)germacrene A (2) and the side-product (-)-valencene (5) in a 94:4:2 ratio. By contrast, the closely related aristolochene synthase from Aspergillus terreus cyclized farnesyl diphosphate only to (+)-aristolochene (4). Site-directed mutagenesis of amino acid residues in two highly conserved Mg(2+)-binding domains led in most cases to reductions in both k(cat) and k(cat)/K(m) as well as increases in the proportion of (S)-(-)germacrene A (2), with the E252Q mutant of the P. roqueforti aristolochene synthase producing only (-)-2. The P. roqueforti D115N, N244L, and S248A/E252D mutants were inactive, as was the A. terreus mutant E227Q. The P. roqueforti mutant Y92F displayed a 100-fold reduction in k(cat) that was offset by a 50-fold decrease in K(m), resulting in a relatively minor 2-fold decrease in catalytic efficiency, k(cat)/K(m). The finding that Y92F produced (+)-aristolochene (4) as 81% of the product, accompanied by 7% 5 and 12% 2, rules out Tyr-92 as the active site Lewis acid that is responsible for protonation of the germacrene A intermediate in the formation of aristolochene (4).


Subject(s)
Isomerases/chemistry , Isomerases/metabolism , Mutagenesis, Site-Directed/genetics , Penicillium/enzymology , Aspartic Acid/genetics , Aspartic Acid/metabolism , Aspergillus/enzymology , Aspergillus/genetics , Binding Sites , Catalysis , Gas Chromatography-Mass Spectrometry , Hydrocarbons/chemistry , Hydrogen-Ion Concentration , Isomerases/genetics , Kinetics , Magnesium/metabolism , Models, Molecular , Molecular Structure , Penicillium/genetics , Protein Structure, Tertiary , Sesquiterpenes, Germacrane/chemistry , Temperature , Tyrosine/genetics , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...