Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
ACS Med Chem Lett ; 15(2): 181-188, 2024 Feb 08.
Article En | MEDLINE | ID: mdl-38352830

We have designed and developed novel and selective TLR7 agonists that exhibited potent receptor activity in a cell-based reporter assay. In vitro, these agonists significantly induced secretion of cytokines IL-6, IL-1ß, IL-10, TNFa, IFNa, and IP-10 in human and mouse whole blood. Pharmacokinetic and pharmacodynamic studies in mice showed a significant secretion of IFNα and TNFα cytokines. When combined with aPD1 in a CT-26 tumor model, the lead compound showed strong synergistic antitumor activity with complete tumor regression in 8/10 mice dosed using the intravenous route. Structure-activity relationship studies enabled by structure-based designs of TLR7 agonists are disclosed.

2.
ACS Med Chem Lett ; 15(2): 189-196, 2024 Feb 08.
Article En | MEDLINE | ID: mdl-38352849

Small molecule toll-like receptor (TLR) 7 agonists have gathered considerable interest as promising therapeutic agents for applications in cancer immunotherapy. Herein, we describe the development and optimization of a series of novel TLR7 agonists through systematic structure-activity relationship studies focusing on modification of the phenylpiperidine side chain. Additional refinement of ADME properties culminated in the discovery of compound 14, which displayed nanomolar reporter assay activity and favorable drug-like properties. Compound 14 demonstrated excellent in vivo pharmacokinetic/pharmacodynamic profiles and synergistic antitumor activity when administered in combination with aPD1 antibody, suggesting opportunities of employing 14 in immuno-oncology therapies with immune checkpoint blockade agents.

3.
ACS Med Chem Lett ; 13(7): 1165-1171, 2022 Jul 14.
Article En | MEDLINE | ID: mdl-35859878

We describe the synthesis of triazole-containing carboline derivatives and their utility as bromodomain and extra-terminal (BET) inhibitors. A convergent synthetic route permitted the detailed investigation of deuteration and fluorination strategies to reduce clearance while maintaining a favorable in vitro profile. This work led to the identification of a potent BET inhibitor, 2-{8-fluoro-3-[4-(2H3)methyl-1-methyl-1H-1,2,3-triazol-5-yl]-5-[(S)-(oxan-4-yl)(phenyl)methyl]-5H-pyrido[3,2-b]indol-7-yl}propan-2-ol (15), which demonstrated reduced clearance and an improved pharmacokinetic (PK) profile across preclinical species. Importantly, no major metabolite was observed when 15 was incubated with human hepatocytes (hHEP) for 2 h. This study culminated with the evaluation of 15 in a mouse triple-negative breast cancer (TNBC) tumor model where it demonstrated robust efficacy at low doses.

4.
Bioorg Med Chem Lett ; 51: 128376, 2021 11 01.
Article En | MEDLINE | ID: mdl-34560263

We describe our efforts to introduce structural diversity to a previously described triazole-containing N1-carboline series of bromodomain and extra-terminal (BET) inhibitors. N9 carbolines were designed to retain favorable binding interactions that the N1-carbolines possess. A convergent synthetic route enabled modifications to reduce clearance, enhance physicochemical properties, and improve the overall in vitro profile. This work led to the identification of a potent BET inhibitor, (S)-2-{8-fluoro-5-[(3-fluoropyridin-2-yl)(oxan-4-yl)methyl]-7-[4-(2H3)methyl-1-methyl-1H-1,2,3-triazol-5-yl]-5H-pyrido[3,2-b]indol-3-yl}propan-2-ol (10), a compound with enhanced oral exposure in mice. Subsequent evaluation in a mouse triple-negative breast cancer tumor model revealed efficacy at 4 mg/kg of N9-carboline 10.


Antineoplastic Agents/pharmacology , Carbolines/pharmacology , Drug Development , Proteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Carbolines/administration & dosage , Carbolines/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Molecular Structure , Proteins/metabolism , Structure-Activity Relationship , Triple Negative Breast Neoplasms/pathology
5.
J Med Chem ; 64(19): 14247-14265, 2021 10 14.
Article En | MEDLINE | ID: mdl-34543572

Inhibition of the bromodomain and extra-terminal (BET) family of adaptor proteins is an attractive strategy for targeting transcriptional regulation of key oncogenes, such as c-MYC. Starting with the screening hit 1, a combination of structure-activity relationship and protein structure-guided drug design led to the discovery of a differently oriented carbazole 9 with favorable binding to the tryptophan, proline, and phenylalanine (WPF) shelf conserved in the BET family. Identification of an additional lipophilic pocket and functional group optimization to optimize pharmacokinetic (PK) properties culminated in the discovery of 18 (BMS-986158) with excellent potency in binding and functional assays. On the basis of its favorable PK profile and robust in vivo activity in a panel of hematologic and solid tumor models, BMS-986158 was selected as a candidate for clinical evaluation.


Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Drug Discovery , Phenylalanine/pharmacology , Proline/pharmacology , Tryptophan/pharmacology , Administration, Oral , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Carbazoles/administration & dosage , Carbazoles/chemistry , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Structure , Phenylalanine/administration & dosage , Phenylalanine/chemistry , Proline/administration & dosage , Proline/chemistry , Structure-Activity Relationship , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Tryptophan/administration & dosage , Tryptophan/chemistry
6.
Bioorg Med Chem Lett ; 44: 128108, 2021 07 15.
Article En | MEDLINE | ID: mdl-33991625

We describe our efforts to identify structurally diverse leads in the triazole-containing N1-carboline series of bromodomain and extra-terminal inhibitors. Replacement of the N5 "cap" phenyl moiety with various heteroaryls, coupled with additional modifications to the carboline core, provided analogs with similar potency, improved pharmacokinetic properties, and increased solubility compared to our backup lead, BMS-986225 (2). Rapid SAR exploration was enabled by a convergent, synthetic route. These efforts provided a potent BET inhibitor, 3-fluoropyridyl 12, that demonstrated robust efficacy in a multiple myeloma mouse tumor model at 1 mg/kg.


Antineoplastic Agents/pharmacology , Carbolines/pharmacology , Drug Development , Multiple Myeloma/diet therapy , Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carbolines/chemical synthesis , Carbolines/chemistry , Dose-Response Relationship, Drug , Humans , Mice , Molecular Structure , Multiple Myeloma/metabolism , Proteins/metabolism , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
7.
ACS Med Chem Lett ; 10(10): 1486-1491, 2019 Oct 10.
Article En | MEDLINE | ID: mdl-31620238

C-terminal Src kinase (CSK) functions as a negative regulator of T cell activation through inhibitory phosphorylation of LCK, so inhibitors of CSK are of interest as potential immuno-oncology agents. Screening of an internal kinase inhibitor collection identified pyridazinone lead 1, and a series of modifications led to optimized compound 13. Compound 13 showed potent activity in biochemical and cellular assays in vitro and demonstrated the ability to increase T cell proliferation induced by T cell receptor signaling. Compound 13 gave extended exposure in mice upon oral dosing and produced a functional response (decrease in LCK phosphorylation) in mouse spleens at 6 h post dose.

9.
ACS Med Chem Lett ; 6(5): 523-7, 2015 May 14.
Article En | MEDLINE | ID: mdl-26005526

Structure-activity relationships in a series of (2-oxo-1,4-benzodiazepin-3-yl)-succinamides identified highly potent inhibitors of γ-secretase mediated signaling of Notch1/2/3/4 receptors. On the basis of its robust in vivo efficacy at tolerated doses in Notch driven leukemia and solid tumor xenograft models, 12 (BMS-906024) was selected as a candidate for clinical evaluation.

10.
Bioorg Med Chem Lett ; 25(9): 1905-9, 2015 May 01.
Article En | MEDLINE | ID: mdl-25857941

This Letter describes synthesis, SAR, and biological activity of (2-oxo-1,4-benzodiazepin-3-yl)-succinamides as inhibitors of γ-secretase mediated signaling of Notch receptors. Optimization of this series led to the identification of BMS-871 (compound 30) which displayed robust in vivo efficacy in Notch-dependent leukemia and solid tumor xenograft models.


Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Benzodiazepinones/administration & dosage , Benzodiazepinones/pharmacology , Receptors, Notch/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Benzodiazepinones/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Receptors, Notch/metabolism , Structure-Activity Relationship
11.
Clin Cancer Res ; 17(12): 4031-41, 2011 Jun 15.
Article En | MEDLINE | ID: mdl-21531814

PURPOSE: The extensive involvement of the HER kinases in epithelial cancer suggests that kinase inhibitors targeting this receptor family have the potential for broad spectrum antitumor activity. BMS-690514 potently inhibits all three HER kinases, and the VEGF receptor kinases. This report summarizes data from biochemical and cellular pharmacology studies, as well as antitumor activity of BMS-690514. EXPERIMENTAL DESIGN: The potency and selectivity of BMS-690514 was evaluated by using an extensive array of enzymatic and binding assays, as well as cellular assays that measure proliferation and receptor signaling. Antitumor activity was evaluated by using multiple xenograft models that depend on HER kinase signaling. The antiangiogenic properties of BMS-690514 were assessed in a matrigel plug assay, and effect on tumor blood flow was measured by dynamic contrast-enhanced MRI. RESULTS: BMS-690514 is a potent and selective inhibitor of epidermal growth factor receptor (EGFR), HER2, and HER4, as well as the VEGF receptor kinases. It inhibits proliferation of tumor cells with potency that correlates with inhibition of receptor signaling, and induces apoptosis in lung tumor cells that have an activating mutation in EGFR. Antitumor activity was observed with BMS-690514 at multiple doses that are well tolerated in mice. There was evidence of suppression of tumor angiogenesis and endothelial function by BMS-690514, which may contribute to its efficacy. CONCLUSIONS: By combining inhibition of two receptor kinase families, BMS-690524 is a novel targeted agent that disrupts signaling in the tumor and its vasculature.


Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Neoplasms/enzymology , Piperidines/pharmacology , Pyrroles/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Triazines/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/blood supply , Neovascularization, Pathologic/enzymology , Receptor, ErbB-2/metabolism , Regional Blood Flow/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
Bioorg Med Chem Lett ; 21(2): 781-5, 2011 Jan 15.
Article En | MEDLINE | ID: mdl-21177105
14.
J Pharm Sci ; 99(8): 3579-93, 2010 Aug.
Article En | MEDLINE | ID: mdl-20166197

BMS-690514, a potent inhibitor of human epidermal growth factor receptor (HER) 1 (EGFR), 2, and 4, and vascular endothelial growth factor receptors (VEGFR) 1-3, is currently under investigation as an oral agent for the treatment of solid tumors. In vitro and in vivo studies were conducted to characterize the pharmacokinetics and metabolism. Through integration of in vitro and in vivo pharmacokinetic data and antitumor efficacy in nude mice, human pharmacokinetics and efficacious doses were projected for BMS-690514. The oral bioavailability of BMS-690514 was 78% in mice, approximately 100% in rats, 8% in monkeys, and 29% in dogs. The low oral bioavailability in monkeys could be attributed to high systemic clearance in that species, which was also consistent with predicted clearance using in vitro data from monkey liver microsomes. Permeability of BMS-690514 in Caco-2 cells was in the intermediate range with a moderate potential to be a P-gp substrate. Experiments using recombinant human CYP enzymes and human liver microsomes suggested that CYP2D6 and CYP3A4 are likely to play a key role in the metabolic clearance of BMS-690514; in addition, direct glucuronidation of BMS-690514 was also observed in human hepatocytes. BMS-690514 was able to cross the blood-brain barrier with a brain-to-plasma ratio of approximately 1. The preclinical ADME properties of BMS-690514 suggest good oral bioavailability in humans and metabolism by multiple pathways including oxidation and glucuronidation. Based on the efficacious AUC in nude mice and predicted human pharmacokinetics, the human efficacious QD dose is predicted to be in the range of 100-200 mg.


ErbB Receptors/antagonists & inhibitors , Piperidines/pharmacokinetics , Pyrroles/pharmacokinetics , Triazines/pharmacokinetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Algorithms , Animals , Blood Proteins/chemistry , Brain Chemistry , Caco-2 Cells , Cell Membrane Permeability , Chromatography, High Pressure Liquid , Computer Simulation , Dogs , Excipients , Hepatocytes/metabolism , Humans , In Vitro Techniques , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microsomes, Liver/metabolism , Permeability , Piperidines/analysis , Protein Binding , Pyrroles/analysis , Rats , Rats, Sprague-Dawley , Spectrophotometry, Ultraviolet , Tandem Mass Spectrometry , Triazines/analysis
15.
J Med Chem ; 52(21): 6527-30, 2009 Nov 12.
Article En | MEDLINE | ID: mdl-19821562

Structure-activity relationships in a series of 4-[1H-indazol-5-ylamino]pyrrolo[2,1-f][1,2,4]triazine-6-carbamates identified dual human epidermal growth factor receptor (HER)1/HER2 kinase inhibitors with excellent biochemical potency and kinase selectivity. On the basis of its favorable pharmacokinetic profile and robust in vivo activity in HER1 and HER2 driven tumor models, 13 (BMS-599626) was selected as a clinical candidate for treatment of solid tumors.


Antineoplastic Agents/chemical synthesis , Carbamates/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Triazines/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , Carbamates/pharmacokinetics , Carbamates/pharmacology , Cell Line, Tumor , Dogs , Drug Screening Assays, Antitumor , Humans , Macaca fascicularis , Mice , Neoplasm Transplantation , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous , Triazines/pharmacokinetics , Triazines/pharmacology
17.
Bioorg Med Chem Lett ; 17(17): 4947-54, 2007 Sep 01.
Article En | MEDLINE | ID: mdl-17606372

Pyrrolotriazine dual EGFR/HER2 kinase inhibitors with a 5-((4-aminopiperidin-1-yl)methyl) solubilizing group were found to be superior to analogs with previously reported C-5 solubilizing groups. New synthetic methodology was developed for the parallel synthesis of C-4 analogs with the new solubilizing group. Interesting new leads were evaluated in tumor xenograft models and the C-4 aminofluorobenzylindazole, 1c, was found to exhibit the best antitumor activity. It is hypothesized that this solubilizing group extends into the ribose-phosphate portion of the ATP binding pocket and enhances the binding affinity of the inhibitor.


Chemistry, Pharmaceutical/methods , ErbB Receptors/chemistry , Neoplasms/drug therapy , Piperidines/chemical synthesis , Pyrroles/chemical synthesis , Receptor, ErbB-2/chemistry , Triazines/chemical synthesis , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Insecta , Models, Chemical , Neoplasm Transplantation , Piperidines/chemistry , Piperidines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Triazines/chemistry , Triazines/pharmacology
20.
Clin Cancer Res ; 12(20 Pt 1): 6186-93, 2006 Oct 15.
Article En | MEDLINE | ID: mdl-17062696

PURPOSE: The studies described here are intended to characterize the ability of BMS-599626, a small-molecule inhibitor of the human epidermal growth factor receptor (HER) kinase family, to modulate signaling and growth of tumor cells that depend on HER1 and/or HER2. EXPERIMENTAL DESIGN: The potency and selectivity of BMS-599626 were assessed in biochemical assays using recombinant protein kinases, as well as in cell proliferation assays using tumor cell lines with varying degrees of dependence on HER1 or HER2 signaling. Modulation of receptor signaling was determined in cell assays by Western blot analyses of receptor autophosphorylation and downstream signaling. The ability of BMS-599626 to inhibit receptor heterodimer signaling in tumor cells was studied by receptor coimmunoprecipitation. Antitumor activity of BMS-599626 was evaluated using a number of different xenograft models that represent a spectrum of human tumors with HER1 or HER2 overexpression. RESULTS: BMS-599626 inhibited HER1 and HER2 with IC50 of 20 and 30 nmol/L, respectively, and was highly selective when tested against a broad panel of diverse protein kinases. Biochemical studies suggested that BMS-599626 inhibited HER1 and HER2 through distinct mechanisms. BMS-599626 abrogated HER1 and HER2 signaling and inhibited the proliferation of tumor cell lines that are dependent on these receptors, with IC50 in the range of 0.24 to 1 micromol/L. BMS-599626 was highly selective for tumor cells that depend on HER1/HER2 and had no effect on the proliferation of cell lines that do not express these receptors. In tumor cells that are capable of forming HER1/HER2 heterodimers, BMS-599626 inhibited heterodimerization and downstream signaling. BMS-599626 had antitumor activity in models that overexpress HER1 (GEO), as well as in models that have HER2 gene amplification (KPL4) or overexpression (Sal2), and there was good correlation between the inhibition of receptor signaling and antitumor activity. CONCLUSIONS: BMS-599626 is a highly selective and potent inhibitor of HER1 and HER2 kinases and inhibits tumor cell proliferation through modulation of receptor signaling. BMS-599626 inhibits HER1/HER2 receptor heterodimerization and provides an additional mechanism of inhibiting tumors in which receptor coexpression and heterodimerization play a major role in driving tumor growth. The preclinical data support the advancement of BMS-599626 into clinical development for the treatment of cancer.


Antineoplastic Agents/toxicity , Enzyme Inhibitors/toxicity , ErbB Receptors/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , CD8 Antigens/immunology , Cell Division/drug effects , Cell Line , Cell Line, Tumor , Dimerization , Humans
...