Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Gynecol Oncol ; 146(3): 484-490, 2017 09.
Article in English | MEDLINE | ID: mdl-28698009

ABSTRACT

OBJECTIVE: Indoleamine 2,3-dioxygenase-1 (IDO1) is a key regulator of immune tolerance in ovarian cancer. This study investigated efficacy and safety of the IDO1 enzyme inhibitor epacadostat versus tamoxifen in patients with biochemical-only recurrence (CA-125 elevation) following complete remission after first-line chemotherapy for advanced epithelial ovarian, primary peritoneal, or fallopian tube cancer. METHODS: In this open-label, phase 2 study (NCT01685255), patients were randomised 1:1 to epacadostat 600mg or tamoxifen 20mg twice daily for successive 28-day cycles and stratified by time since completion of first-line chemotherapy to first CA-125 elevation (3 to <12 or ≥12months). The primary endpoint was investigator-assessed progression-free survival (PFS; RECIST v1.1). Secondary endpoints included CA-125 response (Gynecologic Cancer InterGroup criteria), overall survival, safety, and tolerability. RESULTS: The study was terminated primarily due to slow accrual and lack of evidence of superiority. Median PFS was 3.75months for epacadostat (n=22) versus 5.56months for tamoxifen (n=20; HR, 1.34 [95% CI, 0.58-3.14]; P=0.54). Of evaluable patients, 1 (5.0%) epacadostat and 3 (15.8%) tamoxifen patients had confirmed CA-125 responses. The most common treatment-emergent adverse event was fatigue (epacadostat, 36.4%; tamoxifen, 40.0%). Immune-related adverse events, observed with epacadostat only, were primarily rash (18.2%) and pruritus (9.1%). Epacadostat pharmacokinetics/pharmacodynamics were consistent with its known mechanism of action. IDO1 expression was observed in 94% of archival tumour samples. CONCLUSIONS: This first report of immunotherapy evaluation in biochemical-only relapse ovarian cancer and of IDO1 inhibitor monotherapy in ovarian cancer found no significant difference in efficacy between epacadostat and tamoxifen. Epacadostat was generally well tolerated.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Fallopian Tube Neoplasms/drug therapy , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Oximes/therapeutic use , Peritoneal Neoplasms/drug therapy , Sulfonamides/therapeutic use , Tamoxifen/therapeutic use , Adult , Aged , Antineoplastic Agents, Hormonal/adverse effects , CA-125 Antigen/blood , Carcinoma, Ovarian Epithelial , Disease-Free Survival , Drug Eruptions/etiology , Early Termination of Clinical Trials , Exanthema/chemically induced , Fallopian Tube Neoplasms/blood , Fallopian Tube Neoplasms/chemistry , Fatigue/chemically induced , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/analysis , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Middle Aged , Neoplasms, Glandular and Epithelial/blood , Neoplasms, Glandular and Epithelial/chemistry , Ovarian Neoplasms/blood , Ovarian Neoplasms/chemistry , Oximes/adverse effects , Oximes/pharmacokinetics , Peritoneal Neoplasms/blood , Peritoneal Neoplasms/chemistry , Pruritus/chemically induced , Recurrence , Response Evaluation Criteria in Solid Tumors , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Survival Rate , Tamoxifen/adverse effects
2.
Blood ; 119(20): 4614-8, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22422826

ABSTRACT

We conducted a phase 2 study of ruxolitinib in patients with relapsed/refractory leukemias. Patients with acceptable performance status (0-2), adequate organ function, and no active infection, received ruxolitinib 25 mg orally twice a day for 4 weeks (1 cycle). Response was assessed after every 2 cycles of treatment, and patients who completed 2 cycles were allowed to continue treatment until disease progression. Dose escalation to 50 mg twice daily was permitted in patients demonstrating a benefit. Thirty-eight patients, with a median age of 69 years (range, 45-88), were treated. The median number of prior therapies was 2 (range, 1-6). Twelve patients had JAK2V617F mutation. Patients received a median of 2 cycles of therapy (range, 1-22). Three of 18 patients with postmyeloproliferative neoplasm (MPN) acute myeloid leukemia (AML) showed a significant response; 2 achieved complete remission (CR) and one achieved a CR with insufficient recovery of blood counts (CRi). The responding patients with palpable spleens also had significant reductions in spleen size. Overall, ruxolitinib was very well tolerated with only 4 patients having grade 3 or higher toxicity. Ruxolitinib has modest antileukemic activity as a single agent, particularly in patients with post-MPN AML. The study was registered at www.clinicaltrials.gov as NCT00674479.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Myeloproliferative Disorders/drug therapy , Pyrazoles/therapeutic use , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Janus Kinase 2/genetics , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Leukemia, Myeloid, Acute/pathology , Middle Aged , Mutation, Missense/physiology , Myeloproliferative Disorders/pathology , Nitriles , Phosphorylation/drug effects , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/adverse effects , Pyrimidines , Recurrence , STAT3 Transcription Factor/metabolism , Treatment Outcome
3.
Cancer Cell ; 10(1): 39-50, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16843264

ABSTRACT

We describe here the existence of a heregulin-HER3 autocrine loop, and the contribution of heregulin-dependent, HER2-mediated HER3 activation to gefitinib insensitivity in non-small cell lung cancer (NSCLC). ADAM17 protein, a major ErbB ligand sheddase, is upregulated in NSCLC and is required not only for heregulin-dependent HER3 signaling, but also for EGFR ligand-dependent signaling in NSCLC cell lines. A selective ADAM inhibitor, INCB3619, prevents the processing and activation of multiple ErbB ligands, including heregulin. In addition, INCB3619 inhibits gefitinib-resistant HER3 signaling and enhances gefitinib inhibition of EGFR signaling in NSCLC. These results show that ADAM inhibition affects multiple ErbB pathways in NSCLC and thus offers an excellent opportunity for pharmacological intervention, either alone or in combination with other drugs.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Piperidines/pharmacology , Receptor, ErbB-3/metabolism , Signal Transduction/drug effects , Spiro Compounds/pharmacology , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , ErbB Receptors/genetics , Female , Gefitinib , Gene Expression/genetics , Humans , Ligands , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Paclitaxel/pharmacology , Piperidines/therapeutic use , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Spiro Compounds/therapeutic use , Xenograft Model Antitumor Assays
4.
Blood ; 115(17): 3520-30, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20197554

ABSTRACT

Indoleamine 2,3-dioxygenase-1 (IDO1; IDO) mediates oxidative cleavage of tryptophan, an amino acid essential for cell proliferation and survival. IDO1 inhibition is proposed to have therapeutic potential in immunodeficiency-associated abnormalities, including cancer. Here, we describe INCB024360, a novel IDO1 inhibitor, and investigate its roles in regulating various immune cells and therapeutic potential as an anticancer agent. In cellular assays, INCB024360 selectively inhibits human IDO1 with IC(50) values of approximately 10nM, demonstrating little activity against other related enzymes such as IDO2 or tryptophan 2,3-dioxygenase (TDO). In coculture systems of human allogeneic lymphocytes with dendritic cells (DCs) or tumor cells, INCB024360 inhibition of IDO1 promotes T and natural killer (NK)-cell growth, increases IFN-gamma production, and reduces conversion to regulatory T (T(reg))-like cells. IDO1 induction triggers DC apoptosis, whereas INCB024360 reverses this and increases the number of CD86(high) DCs, potentially representing a novel mechanism by which IDO1 inhibition activates T cells. Furthermore, IDO1 regulation differs in DCs versus tumor cells. Consistent with its effects in vitro, administration of INCB024360 to tumor-bearing mice significantly inhibits tumor growth in a lymphocyte-dependent manner. Analysis of plasma kynurenine/tryptophan levels in patients with cancer affirms that the IDO pathway is activated in multiple tumor types. Collectively, the data suggest that selective inhibition of IDO1 may represent an attractive cancer therapeutic strategy via up-regulation of cellular immunity.


Subject(s)
Dendritic Cells/immunology , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Apoptosis/drug effects , Apoptosis/immunology , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , Coculture Techniques , Dendritic Cells/enzymology , Dose-Response Relationship, Drug , HeLa Cells , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/enzymology , T-Lymphocytes/enzymology , Tryptophan Oxygenase/immunology , Tryptophan Oxygenase/metabolism
5.
J Immunol ; 184(9): 5298-307, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20363976

ABSTRACT

Inhibiting signal transduction induced by inflammatory cytokines offers a new approach for the treatment of autoimmune diseases such as rheumatoid arthritis. Kinase inhibitors have shown promising oral disease-modifying antirheumatic drug potential with efficacy similar to anti-TNF biologics. Direct and indirect inhibition of the JAKs, with small molecule inhibitors like CP-690,550 and INCB018424 or neutralizing Abs, such as the anti-IL6 receptor Ab tocilizumab, have demonstrated rapid and sustained improvement in clinical measures of disease, consistent with their respective preclinical experiments. Therefore, it is of interest to identify optimized JAK inhibitors with unique profiles to maximize therapeutic opportunities. INCB028050 is a selective orally bioavailable JAK1/JAK2 inhibitor with nanomolar potency against JAK1 (5.9 nM) and JAK2 (5.7 nM). INCB028050 inhibits intracellular signaling of multiple proinflammatory cytokines including IL-6 and IL-23 at concentrations <50 nM. Significant efficacy, as assessed by improvements in clinical, histologic and radiographic signs of disease, was achieved in the rat adjuvant arthritis model with doses of INCB028050 providing partial and/or periodic inhibition of JAK1/JAK2 and no inhibition of JAK3. Diminution of inflammatory Th1 and Th17 associated cytokine mRNA levels was observed in the draining lymph nodes of treated rats. INCB028050 was also effective in multiple murine models of arthritis, with no evidence of suppression of humoral immunity or adverse hematologic effects. These data suggest that fractional inhibition of JAK1 and JAK2 is sufficient for significant activity in autoimmune disease models. Clinical evaluation of INCB028050 in RA is ongoing.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Experimental/enzymology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Animals , Arthritis, Experimental/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , Cell Line , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Female , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Janus Kinase 1/physiology , Janus Kinase 2/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Random Allocation , Rats , Rats, Inbred Lew , Signal Transduction/drug effects , Signal Transduction/immunology
6.
Bioorg Med Chem Lett ; 21(6): 1827-31, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21316220

ABSTRACT

This report describes the design and synthesis of a series of CCR2 antagonists incorporating novel non-aryl/heteroaryl RHS (right hand side) motifs. Previous SAR in the area has suggested an aryl/heteroaryl substituent as a necessary structural feature for binding to the CCR2 receptor. Herein we describe the SAR with regards to potency (binding to hCCR2), dofetilide activity and metabolic stability (in vitro HLM) for this series. The resulting outcome was the identification of compounds with excellent properties for the investigation of the role of CCR2 in disease.


Subject(s)
Drug Design , Receptors, CCR2/antagonists & inhibitors , Binding Sites , Models, Molecular , Structure-Activity Relationship
7.
Cancer Discov ; 11(2): 326-339, 2021 02.
Article in English | MEDLINE | ID: mdl-33218975

ABSTRACT

Pemigatinib, a selective FGFR1-3 inhibitor, has demonstrated antitumor activity in FIGHT-202, a phase II study in patients with cholangiocarcinoma harboring FGFR2 fusions/rearrangements, and has gained regulatory approval in the United States. Eligibility for FIGHT-202 was assessed using genomic profiling; here, these data were utilized to characterize the genomic landscape of cholangiocarcinoma and to uncover unique molecular features of patients harboring FGFR2 rearrangements. The results highlight the high percentage of patients with cholangiocarcinoma harboring potentially actionable genomic alterations and the diversity in gene partners that rearrange with FGFR2. Clinicogenomic analysis of pemigatinib-treated patients identified mechanisms of primary and acquired resistance. Genomic subsets of patients with other potentially actionable FGF/FGFR alterations were also identified. Our study provides a framework for molecularly guided clinical trials and underscores the importance of genomic profiling to enable a deeper understanding of the molecular basis for response and nonresponse to targeted therapy. SIGNIFICANCE: We utilized genomic profiling data from FIGHT-202 to gain insights into the genomic landscape of cholangiocarcinoma, to understand the molecular diversity of patients with FGFR2 fusions or rearrangements, and to interrogate the clinicogenomics of patients treated with pemigatinib. Our study highlights the utility of genomic profiling in clinical trials.This article is highlighted in the In This Issue feature, p. 211.


Subject(s)
Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Morpholines/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/genetics , Adult , Bile Duct Neoplasms/genetics , Cholangiocarcinoma/genetics , Cohort Studies , Drug Resistance, Neoplasm , Female , Gene Rearrangement , Humans , Male
8.
Biochem Biophys Res Commun ; 387(2): 251-5, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19576173

ABSTRACT

The chemokine receptor 2 (CCR2) directs migration of monocytes and has been proposed to be a drug target for chronic inflammatory diseases. INCB3344 was first published as a small molecule nanomolar inhibitor of rodent CCR2. Here, we show that INCB3344 can also bind human CCR2 (hCCR2) with high affinity, having a dissociation constant (K(d)) of approximately 5nM. The binding of the compound to the receptor is rapid and reversible. INCB3344 potently inhibits hCCR2 binding of monocyte chemoattractant protein-1 (MCP-1) and MCP-1-induced signaling and function in hCCR2-expressing cells, including ERK phosphorylation and chemotaxis, and is competitive against MCP-1 in vitro. INCB3344 also blocks MCP-1 binding to monocytes in human whole blood, with potency consistent with in vitro studies. The whole blood binding assay described here can be used for monitoring pharmacodynamic activity of CCR2 antagonists in both preclinical models and in the clinic.


Subject(s)
Pyrrolidines/pharmacology , Receptors, CCR2/antagonists & inhibitors , Biological Assay , Cells, Cultured , Chemokine CCL2/metabolism , Chemotaxis , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry/methods , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Phosphorylation , Pyrrolidines/metabolism
9.
J Immunother Cancer ; 7(1): 80, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894212

ABSTRACT

BACKGROUND: Epacadostat is a potent inhibitor of the immunosuppressive indoleamine 2,3-dioxygenase 1 (IDO1) enzyme. We present phase 1 results from a phase 1/2 clinical study of epacadostat in combination with ipilimumab, an anti-cytotoxic T-lymphocyte-associated protein 4 antibody, in advanced melanoma (NCT01604889). METHODS: Only the phase 1, open-label portion of the study was conducted, per the sponsor's decision to terminate the study early based on the changing melanoma treatment landscape favoring exploration of programmed cell death protein 1 (PD-1)/PD-ligand 1 inhibitor-based combination strategies. Such decision was not related to the safety of epacadostat plus ipilimumab. Patients received oral epacadostat (25, 50, 100, or 300 mg twice daily [BID]; 75 mg daily [50 mg AM, 25 mg PM]; or 50 mg BID intermittent [2 weeks on/1 week off]) plus intravenous ipilimumab 3 mg/kg every 3 weeks. RESULTS: Fifty patients received ≥1 dose of epacadostat. As of January 20, 2017, 2 patients completed treatment and 48 discontinued, primarily because of adverse events (AEs) and disease progression (n = 20 each). Dose-limiting toxicities occurred in 11 patients (n = 1 each with epacadostat 25 mg BID, 50 mg BID intermittent, 75 mg daily; n = 4 each with epacadostat 50 mg BID, 300 mg BID). The most common immune-related treatment-emergent AEs included rash (50%), alanine aminotransferase elevation (28%), pruritus (28%), aspartate aminotransferase elevation (24%), and hypothyroidism (10%). Among immunotherapy-naive patients (n = 39), the objective response rate was 26% by immune-related response criteria and 23% by Response Evaluation Criteria in Solid Tumors version 1.1. No objective response was seen in the 11 patients who received prior immunotherapy. Epacadostat exposure was dose proportional, with clinically significant IDO1 inhibition at doses ≥25 mg BID. CONCLUSIONS: When combined with ipilimumab, epacadostat ≤50 mg BID demonstrated clinical and pharmacologic activity and was generally well tolerated in patients with advanced melanoma. TRIAL REGISTRATION: ClinicalTrials.gov identifier, NCT01604889 . Registration date, May 9, 2012, retrospectively registered.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Ipilimumab/administration & dosage , Melanoma/drug therapy , Oximes/administration & dosage , Sulfonamides/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Drug Administration Schedule , Female , Humans , Ipilimumab/adverse effects , Male , Middle Aged , Neoplasm Metastasis , Oximes/adverse effects , Sulfonamides/adverse effects , Treatment Outcome
10.
Bioorg Med Chem Lett ; 18(6): 1958-62, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18282708

ABSTRACT

Potent and selective inhibitors of tumor necrosis factor-alpha converting enzyme (TACE) were discovered with several new heterocyclic P1' groups in conjunction with cyclic beta-amino hydroxamic acid scaffolds. Among them, the pyrazolopyridine provided the best overall profile when combined with tetrahydropyran beta-amino hydroxamic acid scaffold. Specifically, inhibitor 49 showed IC(50) value of 1 nM against porcine TACE and 170 nM in the suppression of LPS-induced TNF-alpha of human whole blood. Compound 49 also displayed excellent selectivity over a wide panel of MMPs as well as excellent oral bioavailability (F%>90%) in rat n-in-1 PK studies.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Benzofurans/chemistry , Imidazoles/chemistry , Indoles/chemistry , Protease Inhibitors/pharmacology , Pyrazoles/chemistry , Pyridines/chemistry , ADAM Proteins/metabolism , ADAM17 Protein , Administration, Oral , Animals , Biological Availability , Humans , Hydroxamic Acids/chemistry , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase Inhibitors , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacokinetics , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism
11.
Bioorg Med Chem Lett ; 18(4): 1288-92, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18234496

ABSTRACT

Two novel oxaspiro[4.4]nonane beta-benzamido hydroxamic scaffolds have been synthesized in enantio- and diasteriomerically pure form. These templates proved to be exceptional platforms that have led to the discovery of potent inhibitors of TACE that are active in a cellular assay measuring suppression of LPS-induced TNF-alpha. Furthermore, these inhibitors are selective against related MMPs, demonstrate permeability in a Caco-2 assay, and display good oral bioavailability.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Alkanes/chemistry , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Spiro Compounds/chemistry , ADAM17 Protein , Administration, Oral , Alkanes/chemical synthesis , Alkanes/pharmacokinetics , Alkanes/pharmacology , Animals , Biological Availability , Caco-2 Cells , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacokinetics , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/metabolism , Models, Molecular , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacokinetics , Rats , Rats, Sprague-Dawley , Spiro Compounds/chemical synthesis , Spiro Compounds/pharmacokinetics , Spiro Compounds/pharmacology
12.
Bioorg Med Chem Lett ; 18(5): 1577-82, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18242982

ABSTRACT

Novel ((2-substituted-1H-benzo[d]imidazol-1-yl)methyl)benzamides were found to be excellent P1' substituents in conjunction with unique constrained beta-amino hydroxamic acid scaffolds for the discovery of potent selective inhibitors of TNF-alpha Converting Enzyme (TACE). Optimized examples proved potent for TACE, exceptionally selective over a wide panel of MMP and ADAM proteases, potent in the suppression of LPS-induced TNF-alpha in human whole blood and orally bioavailable.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Benzamides/chemistry , Benzamides/pharmacology , ADAM17 Protein , Animals , Area Under Curve , Benzamides/blood , Benzamides/pharmacokinetics , Biological Availability , Dogs , Half-Life , Molecular Structure , Rats , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 18(2): 694-9, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18061445

ABSTRACT

Selective inhibitors of TNF-alpha Converting Enzyme (TACE) based on (1R,2S)-cyclopentyl, (3S,4S)-pyrrolidinyl, and (3R,4S)-tetrahydrofuranyl beta-benzamido hydroxamic acids have been synthesized and evaluated. This study has led to the discovery of novel inhibitors whose profiles include activity against TACE in an enzyme assay, potency in the suppression of LPS-stimulated TNF-alpha in human whole blood, selectivity against a panel of MMPs and oral bioavailability.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , ADAM17 Protein , Administration, Oral , Animals , Biological Availability , Chromatography, High Pressure Liquid , Enzyme Inhibitors/chemical synthesis , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacokinetics , Rats , Stereoisomerism
15.
Clin Cancer Res ; 13(6): 1892-902, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363546

ABSTRACT

PURPOSE: ErbB receptor signaling pathways are important regulators of cell fate, and their dysregulation, through (epi)genetic alterations, plays an etiologic role in multiple cancers. ErbB ligands are synthesized as membrane-bound precursors that are cleaved by members of the ADAM family of zinc-dependent metalloproteases. This processing, termed ectodomain shedding, is essential for the functional activation of ErbB ligands. Recent studies suggest that elevated levels of ErbB ligands may circumvent the effectiveness of ErbB-targeted therapeutics. Here, we describe the discovery and preclinical development of potent, selective inhibitors of ErbB ligand shedding. EXPERIMENTAL DESIGN: A series of biochemical and cell-based assays were established to identify selective inhibitors of ErbB ligand shedding. The therapeutic potential of these compounds was assessed in multiple in vivo models of cancer and matrix metalloprotease-related toxicity. RESULTS: INCB3619 was identified as a representative selective, potent, orally bioavailable small-molecule inhibitor of a subset of ADAM proteases that block shedding of ErbB ligands. Administration of INCB3619 to tumor-bearing mice reduced ErbB ligand shedding in vivo and inhibited ErbB pathway signaling (e.g., phosphorylation of Akt), tumor cell proliferation, and survival. Further, INCB3619 synergized with clinically relevant cancer therapeutics and showed no overt or compounding toxicities, including fibroplasia, the dose-limiting toxicity associated with broad-spectrum matrix metalloprotease inhibitors. CONCLUSIONS: Inhibition of ErbB ligand shedding offers a potentially novel and well-tolerated therapeutic strategy for the treatment of human cancers and is currently being evaluated in the clinic.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Oncogene Proteins v-erbB/metabolism , Signal Transduction/drug effects , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Neoplasms/metabolism , Neoplasms/pathology , Rats , Xenograft Model Antitumor Assays
16.
Clin Cancer Res ; 23(13): 3269-3276, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28053021

ABSTRACT

Purpose: Indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the degradation of tryptophan to N-formyl-kynurenine. Overexpressed in many solid malignancies, IDO1 can promote tumor escape from host immunosurveillance. This first-in-human phase I study investigated the maximum tolerated dose, safety, pharmacokinetics, pharmacodynamics, and antitumor activity of epacadostat (INCB024360), a potent and selective inhibitor of IDO1.Experimental Design: Fifty-two patients with advanced solid malignancies were treated with epacadostat [50 mg once daily or 50, 100, 300, 400, 500, 600, or 700 mg twice daily (BID)] in a dose-escalation 3 + 3 design and evaluated in 28-day cycles. Treatment was continued until disease progression or unacceptable toxicity.Results: One dose-limiting toxicity (DLT) occurred at the dose of 300 mg BID (grade 3, radiation pneumonitis); another DLT occurred at 400 mg BID (grade 3, fatigue). The most common adverse events in >20% of patients overall were fatigue, nausea, decreased appetite, vomiting, constipation, abdominal pain, diarrhea, dyspnea, back pain, and cough. Treatment produced significant dose-dependent reductions in plasma kynurenine levels and in the plasma kynurenine/tryptophan ratio at all doses and in all patients. Near maximal changes were observed at doses of ≥100 mg BID with >80% to 90% inhibition of IDO1 achieved throughout the dosing period. Although no objective responses were detected, stable disease lasting ≥16 weeks was observed in 7 of 52 patients.Conclusions: Epacadostat was generally well tolerated, effectively normalized kynurenine levels, and produced maximal inhibition of IDO1 activity at doses of ≥100 mg BID. Studies investigating epacadostat in combination with other immunomodulatory drugs are ongoing. Clin Cancer Res; 23(13); 3269-76. Ā©2017 AACR.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/pathology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Neoplasms/drug therapy , Oximes/administration & dosage , Sulfonamides/administration & dosage , Administration, Oral , Adult , Aged , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions/classification , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Male , Middle Aged , Neoplasm Staging , Neoplasms/genetics , Neoplasms/pathology
17.
Cancer Biol Ther ; 5(6): 648-56, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16627988

ABSTRACT

The HER-2 receptor tyrosine kinase is an important regulator of cell proliferation and survival, and it is a clinically validated target of therapeutic intervention for HER-2 positive breast cancer patients. Its extracellular domain (ECD) is frequently cleaved by protease(s) in HER-2 overexpressing breast cancer patients, rendering the remaining membrane-bound portion (p95) a constitutively activated kinase. The presence of both serum ECD and cellular p95 protein has been linked to poor clinical outcome as well as reduced effectiveness of some therapeutic treatments. We have identified a series of potent, selective small molecule inhibitors of ADAM proteases, exemplified here by INCB003619, and demonstrate that these inhibitors effectively block HER-2 cleavage in HER-2 overexpressing human breast cancer cell lines. Intriguingly, when used in combination, INCB003619 dramatically enhances the antiproliferative activity of suboptimal doses of the anti-HER-2 antibody, trastuzumab, in HER-2 overexpressing/shedding breast cancer cell lines, accompanied by reduced ERK and AKT phosphorylation. Furthermore, INCB003619, in combination with trastuzumab, augments the pro-apoptotic and antiproliferative effects of the chemotherapeutic agent paclitaxel. Consistent with these in vitro data, INCB003619 reduces serum ECD levels and enhances the antitumor effect of trastuzumab in a xenograft tumor model derived from the HER-2 overexpressing BT-474 breast cancer cell line. Collectively, these findings suggest that blocking HER-2 cleavage with selective ADAM inhibitors may represent a novel therapeutic approach for treating HER-2 overexpressing breast cancer patients.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Transplantation, Heterologous , Trastuzumab
18.
J Med Chem ; 48(6): 2194-211, 2005 Mar 24.
Article in English | MEDLINE | ID: mdl-15771462

ABSTRACT

Starting with our previously described(20) class of CC chemokine receptor-3 (CCR3) antagonist, we improved the potency by replacing the phenyl linker of 1 with a cyclohexyl linker and by replacing the 4-benzylpiperidine with a 3-benzylpiperidine. The resulting compound, 32, is a potent and selective antagonist of CCR3. SAR studies showed that the 3-acetylphenyl urea of 32 could be replaced with heterocyclic ureas or heterocyclic-substituted phenyl ureas and still maintain the potency (inhibition of eotaxin-induced chemotaxis) of this class of compounds in the low-picomolar range (IC(50) = 10-60 pM), representing some of the most potent CCR3 antagonists reported to date. The potency of 32 for mouse CCR3 (chemotaxis IC(50) = 41 nM) and its oral bioavailability in mice (20% F ) were adequate to assess the efficacy in animal models of allergic airway inflammation. Oral administration of 32 reduced eosinophil recruitment into the lungs in a dose-dependent manner in these animal models. On the basis of its overall potency, selectivity, efficacy, and safety profile, the benzenesulfonate salt of 32, designated DPC168, entered phase I clinical trials.


Subject(s)
Cyclohexanes/chemical synthesis , Phenylurea Compounds/chemical synthesis , Piperidines/chemical synthesis , Receptors, Chemokine/antagonists & inhibitors , Animals , Biological Availability , CHO Cells , Caco-2 Cells , Calcium/metabolism , Chemotaxis, Leukocyte/drug effects , Cricetinae , Cyclohexanes/chemistry , Cyclohexanes/pharmacology , Eosinophils/drug effects , Eosinophils/physiology , Female , Humans , Hypersensitivity/drug therapy , Hypersensitivity/immunology , In Vitro Techniques , Inflammation/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Permeability , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Receptors, CCR3 , Stereoisomerism , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemical synthesis , Urea/chemistry , Urea/pharmacology
19.
Matrix Biol ; 21(6): 499-511, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12392761

ABSTRACT

ADAM-TS5 (aggrecanase 2), one of two cartilage aggrecanases is a member of the ADAM protein family. Like ADAM-TS4 (aggrecanase 1) the enzyme cleaves cartilage aggrecan at the Glu(373)-Ala(374) bond, a marker of aggrecanase activity. In this study we have characterized the substrate specificity of ADAM-TS5 and compared it with that of ADAM-TS4. The recombinant human ADAM-TS5, like ADAM-TS4 cleaves aggrecan at Glu(1480)-Gly(1481), Glu(1667)-Gly(1668), Glu(1771)-Ala(1772) and Glu(1871)-Leu(1872) bonds more readily than at the Glu(373)-Ala(374) bond. In addition, ADAM-TS5 exhibited an additional site of cleavage in the region spanning residues Gly(1481) and Glu(1667), representing a unique cleavage of ADAM-TS5. ADAM-TS5 cleaved aggrecan approximately 2-fold slower than ADAM-TS4. Neither ADAM-TS5 nor ADAM-TS4 was able to cleave the extracellular matrix proteins fibronectin, thrombospondin, type I collagen, type II collagen, gelatin or general protein substrates such as casein and transferrin. Finally, the zymogen of stromelysin (MMP-3) was not activated by either ADAM-TS4 or ADAM-TS5.


Subject(s)
Metalloendopeptidases/metabolism , ADAM Proteins , ADAMTS4 Protein , ADAMTS5 Protein , Aggrecans , Animals , Cell Line , Drosophila/cytology , Extracellular Matrix Proteins/drug effects , Extracellular Matrix Proteins/metabolism , Humans , Lectins, C-Type , Metalloendopeptidases/pharmacology , Procollagen N-Endopeptidase , Proteoglycans/chemistry , Proteoglycans/drug effects , Proteoglycans/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Substrate Specificity , Time Factors
20.
J Med Chem ; 45(23): 4954-7, 2002 Nov 07.
Article in English | MEDLINE | ID: mdl-12408705

ABSTRACT

New gamma-lactam TACE inhibitors were designed from known MMP inhibitors. A homology model of TACE was built and examined to identify the S1' site as the key area for TACE selectivity over MMPs. Rational exploration of the P1'-S1' interactions resulted in the discovery of the 3,5-disubstituted benzyl ether as a TACE-selective P1' group. Further optimization led to the discovery of IK682 as a selective and orally bioavailable TACE inhibitor.


Subject(s)
Hydroxamic Acids/chemical synthesis , Lactams/chemical synthesis , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors/chemical synthesis , ADAM Proteins , ADAM17 Protein , Administration, Oral , Animals , Biological Availability , Dogs , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Lactams/chemistry , Lactams/pharmacology , Matrix Metalloproteinase 3/chemistry , Models, Molecular , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Swine
SELECTION OF CITATIONS
SEARCH DETAIL