Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 73: 128884, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35835377

ABSTRACT

11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1) has been identified as the primary enzyme responsible for the activation of hepatic cortisone to cortisol in specific peripheral tissues, resulting in the concomitant antagonism of insulin action within these tissues. Dysregulation of 11ß-HSD1, particularly in adipose tissues, has been associated with a variety of ailments including metabolic syndrome and type 2 diabetes mellitus. Therefore, inhibition of 11ß-HSD1 with a small nonsteroidal molecule is therapeutically desirable. Implementation of a scaffold-hopping approach revealed a 3-point pharmacophore for 11ß-HSD1 that was utilized to design a 2-spiroproline derivative as a steroid mimetic scaffold. Reiterative optimization provided valuable insight into the bioactive conformation of our novel scaffold and led to the discovery of several leads, such as compounds 39 and 51. Importantly, deleterious hERG inhibition and pregnane X receptor induction were mitigated by the introduction of a 4-hydroxyl group to the proline ring system.


Subject(s)
Diabetes Mellitus, Type 2 , Metabolic Syndrome , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrocortisone/metabolism
2.
Bioorg Med Chem Lett ; 69: 128782, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35537608

ABSTRACT

11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1) has been identified as the primary enzyme responsible for the activation of hepatic cortisone to cortisol in specific peripheral tissues resulting in the concomitant antagonism of insulin action within these tissues. Dysregulation of 11ß-HSD1, particularly in adipose tissues, has been associated with metabolic syndrome and type 2 diabetes mellitus. Therefore, inhibition of 11ß-HSD1 with a small nonsteroidal molecule is therapeutically desirable. Implementation of a scaffold-hopping approach revealed a three-point pharmacophore for 11ß-HSD1 that was utilized to design a steroid mimetic scaffold. Reiterative optimization provided valuable insight into the bioactive conformation of our novel scaffold and led to the discovery of INCB13739. Clinical evaluation of INCB13739 confirmed for the first time that tissue-specific inhibition of 11ß-HSD1 in patients with type 2 diabetes mellitus was efficacious in controlling glucose levels and reducing cardiovascular risk factors.


Subject(s)
Diabetes Mellitus, Type 2 , Metabolic Syndrome , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrocortisone/metabolism , Metabolic Syndrome/metabolism
3.
J Med Chem ; 64(15): 10666-10679, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34269576

ABSTRACT

Aberrant activation of FGFR has been linked to the pathogenesis of many tumor types. Selective inhibition of FGFR has emerged as a promising approach for cancer treatment. Herein, we describe the discovery of compound 38 (INCB054828, pemigatinib), a highly potent and selective inhibitor of FGFR1, FGFR2, and FGFR3 with excellent physiochemical properties and pharmacokinetic profiles. Pemigatinib has received accelerated approval from the U.S. Food and Drug Administration for the treatment of adults with previously treated, unresectable locally advanced or metastatic cholangiocarcinoma with a FGFR2 fusion or other rearrangement. Additional clinical trials are ongoing to evaluate pemigatinib in patients with FGFR alterations.


Subject(s)
Drug Discovery , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Molecular Structure , Morpholines/chemical synthesis , Morpholines/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Structure-Activity Relationship , United States , United States Food and Drug Administration
4.
Eur J Pharmacol ; 675(1-3): 47-56, 2012 Jan 30.
Article in English | MEDLINE | ID: mdl-22155710

ABSTRACT

The histamine H4 receptor mediates several histamine-induced cellular functions of leukocytes, including cell migration and cytokine production. Recent studies suggest that histamine signaling through the histamine H4 receptor can also have anti-pruritic and anti-nociceptive functions. 1-(7-(2-amino-6-(4-methylpiperazin-1-yl) pyrimidin-4-yl)-3, 4-dihdroisoquinolin-2(1H)-yl)-2-cyclopentylethanone (INCB38579) is a novel small molecule antagonist of the human and rodent histamine H4 receptors with at least 80-fold selectivity over the human histamine H1, H2 and H3 receptors, and has good pharmacokinetic properties in rats and mice. The compound is potent in inhibiting histamine binding to and signaling through the recombinant human, mouse and rat histamine H4 receptors and blocks the histamine-induced migration of human and mouse dendritic cells, as well as the cell shape change and migration of human eosinophils. INCB38579 and histamine may have separate but overlapping binding sites on the human histamine H4 receptor. This novel inhibitor is efficacious when evaluated in two previously established in vivo models for histamine H4 receptor activity (histamine-induced itch in mice and carrageenan-induced acute inflammatory pain in rats). When examined in formalin-induced pain models, INCB38579 significantly reduces the sustained inflammatory pain experienced by rats and mice. A good correlation between the protein binding adjusted potency from in vitro studies and its analgesic effect in vivo was observed. These results suggest that INCB38579 can serve as a useful tool for pharmacologic characterization of the histamine H4 receptor and further support the hypothesis that targeting the histamine H4 receptor may provide new therapeutic agents for various chronic inflammatory diseases, including inflammatory pain.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antipruritics/therapeutic use , Histamine Antagonists/therapeutic use , Isoquinolines/therapeutic use , Pyrimidines/therapeutic use , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/blood , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antipruritics/blood , Antipruritics/metabolism , Antipruritics/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Chemotactic Factors/blood , Chemotactic Factors/metabolism , Chemotactic Factors/pharmacology , Chemotactic Factors/therapeutic use , Female , HEK293 Cells , Histamine Antagonists/blood , Histamine Antagonists/metabolism , Histamine Antagonists/pharmacology , Humans , Immune System/cytology , Immune System/drug effects , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/physiopathology , Isoquinolines/blood , Isoquinolines/metabolism , Isoquinolines/pharmacology , Male , Mice , Mice, Inbred Strains , Pruritus/chemically induced , Pruritus/drug therapy , Pyrimidines/blood , Pyrimidines/metabolism , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/genetics , Receptors, Histamine/metabolism , Receptors, Histamine H4 , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism
5.
Clin Cancer Res ; 17(22): 7127-38, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21918175

ABSTRACT

PURPOSE: The c-MET receptor tyrosine kinase plays important roles in the formation, progression, and dissemination of human cancer and presents an attractive therapeutic target. This study describes the preclinical characterization of INCB28060, a novel inhibitor of c-MET kinase. EXPERIMENTAL DESIGN: Studies were conducted using a series of in vitro and in vivo biochemical and biological experiments. RESULTS: INCB28060 exhibits picomolar enzymatic potency and is highly specific for c-MET with more than 10,000-fold selectivity over a large panel of human kinases. This inhibitor potently blocks c-MET phosphorylation and activation of its key downstream effectors in c-MET-dependent tumor cell lines. As a result, INCB28060 potently inhibits c-MET-dependent tumor cell proliferation and migration and effectively induces apoptosis in vitro. Oral dosing of INCB28060 results in time- and dose-dependent inhibition of c-MET phosphorylation and tumor growth in c-MET-driven mouse tumor models, and the inhibitor is well tolerated at doses that achieve complete tumor inhibition. In a further exploration of potential interactions between c-MET and other signaling pathways, we found that activated c-MET positively regulates the activity of epidermal growth factor receptors (EGFR) and HER-3, as well as expression of their ligands. These effects are reversed with INCB28060 treatment. Finally, we confirmed that circulating hepatocyte growth factor levels are significantly elevated in patients with various cancers. CONCLUSIONS: Activated c-MET has pleiotropic effects on multiple cancer-promoting signaling pathways and may play a critical role in driving tumor cell growth and survival. INCB28060 is a potent and selective c-MET kinase inhibitor that may have therapeutic potential in cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , ErbB Receptors/metabolism , Neoplasms, Experimental/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptor Cross-Talk/drug effects , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation , Female , Glioblastoma/drug therapy , Humans , Imidazoles , Mice , Mice, Nude , Phosphorylation , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-3/metabolism , Triazines , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem Lett ; 19(13): 3525-30, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19457660

ABSTRACT

A serendipitous discovery that the metalloprotease binding profile of a novel class of 2-carboxamide-3-hydroxamic acid piperidines could be significantly attenuated by the modification of the unexplored P1 substituent enabled the design and synthesis of a novel 2-carboxamide-1-hydroxamic acid cyclohexyl scaffold core that exhibited excellent HER-2 potency and unprecedented MMP-selectivity that we believe would not have been possible via conventional P1' perturbations.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Antineoplastic Agents/chemical synthesis , Hydroxamic Acids/chemical synthesis , Membrane Proteins/metabolism , Receptor, ErbB-2/metabolism , ADAM10 Protein , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Design , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Matrix Metalloproteinase 2/metabolism , Protein Binding , Structure-Activity Relationship , Substrate Specificity
7.
Bioorg Med Chem Lett ; 18(1): 159-63, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18036818

ABSTRACT

In an effort to obtain a MMP selective and potent inhibitor of HER-2 sheddase (ADAM-10), the P1' group of a novel class of (6S,7S)-7-[(hydroxyamino)carbonyl]-6-carboxamide-5-azaspiro[2.5]octane-5-carboxylates was attenuated and the structure-activity relationships (SAR) will be discussed. In addition, it was discovered that unconventional perturbation of the P2' moiety could confer MMP selectivity, which was hypothesized to be a manifestation of the P2' group effecting global conformational changes.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Hydroxamic Acids/chemistry , Membrane Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Receptor, ErbB-2/antagonists & inhibitors , ADAM Proteins/metabolism , ADAM10 Protein , Amides/chemical synthesis , Amides/chemistry , Amides/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Aza Compounds/chemical synthesis , Aza Compounds/chemistry , Aza Compounds/pharmacology , Drug Design , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , Membrane Proteins/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Receptor, ErbB-2/metabolism , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Structure-Activity Relationship , Substrate Specificity
8.
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
9.
J Med Chem ; 50(4): 603-6, 2007 Feb 22.
Article in English | MEDLINE | ID: mdl-17256836

ABSTRACT

The design, synthesis, evaluation, and identification of a novel class of (6S,7S)-N-hydroxy-6-carboxamide-5-azaspiro[2.5]octane-7-carboxamides as the first potent and selective inhibitors of human epidermal growth factor receptor-2 (HER-2) sheddase is described. Several compounds were identified that possess excellent pharmacodynamic and pharmacokinetic properties and were shown to decrease tumor size, cleaved HER-2 extracellular domain plasma levels, and potentiate the effects of the humanized anti-HER-2 monoclonal antibody (trastuzumab) in vivo in a HER-2 overexpressing cancer murine xenograft model.


Subject(s)
Amides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Hydroxamic Acids/chemical synthesis , Piperidines/chemical synthesis , Receptor, ErbB-2/antagonists & inhibitors , Spiro Compounds/chemical synthesis , Administration, Oral , Amides/pharmacokinetics , Amides/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/pharmacology , Mice , Molecular Conformation , Piperidines/chemistry , Piperidines/pharmacology , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous , Trastuzumab
10.
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
11.
Cancer Biol Ther ; 5(6): 657-64, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16627989

ABSTRACT

Overexpression and activating mutations of ErbB family members have been implicated in the development and progression of a variety of tumor types. Cleavage of the HER2 receptor by an as yet unidentified ectodomain sheddase has been shown to liberate the HER2 extracellular domain (ECD) leaving a fragment with constitutive kinase activity that can provide ligand-independent growth and survival signals to the cell. This process is clinically relevant since HER2 ECD serum levels in metastatic breast cancer patients are associated with a poorer prognosis. Thus, inhibition of the HER2 sheddase may provide a novel therapeutic approach for breast cancer. We describe the use of transcriptional profiling, pharmacological and in vitro approaches to identify the major source of HER2 sheddase activity. Real-time PCR was used to identify those ADAM family members which were expressed in HER2 shedding cell lines. siRNAs that selectively inhibited ADAM10 expression reduced HER2 shedding. In addition, we profiled over 1000 small molecules for in vitro inhibition of a panel of ADAM and MMP proteins; a positive correlation was observed only between ADAM10 inhibition and reduction of HER2 ECD shedding in a cell based assay. Finally, in vitro studies demonstrate that in combination with low doses of Herceptin, selective ADAM10 inhibitors decrease proliferation in HER2 overexpressing cell lines while inhibitors, that do not inhibit ADAM10, have no impact. These results are consistent with ADAM10 being a major determinant of HER2 shedding, the inhibition of which, may provide a novel therapeutic approach for treating a variety of cancers with active HER2 signaling.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Breast Neoplasms/genetics , Membrane Proteins/metabolism , Receptor, ErbB-2/metabolism , ADAM10 Protein , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Base Sequence , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Polymerase Chain Reaction , RNA, Small Interfering/genetics , Trastuzumab
12.
J Med Chem ; 45(18): 4018-28, 2002 Aug 29.
Article in English | MEDLINE | ID: mdl-12190323

ABSTRACT

A small library consisting of two series of thymidine derivatives containing o-carboranylalkyl groups at the N-3 position was prepared. In both series, alkyl spacers of 2-7 methylene units were placed between the o-carborane cage and the thymidine scaffold. In one series, an additional dihydroxypropyl substituent was introduced at the second carbon atom of the carborane cage. In the series of N-3-substituted carboranyl thymidines without additional dihydroxypropyl substituent, three steps were required to obtain the target compounds in overall yields as high as 75%, while in the series of N-3-substituted carboranyl thymidines with additional dihydroxypropyl substituent, 9-10 steps were necessary with significantly lower overall yield. All target compounds were good substrates of human cytosolic thymidine kinase 1 while they were, if at all, poor substrates of the mitochondrial thymidine kinase 2. There was only a minor difference in phosphorylation rates between N-3-substituted carboranyl thymidines with additional dihydroxypropyl substituents with thymidine kinase 1 (range: 13-49% relative to thymidine) and their counterparts lacking this group (range: 11-57% relative to thymidine). Tether lengths of two and five methylene groups in both series gave the highest enzyme activities in the present study. A hypothesis for this result is presented.


Subject(s)
Boron Compounds/chemical synthesis , Thymidine Kinase/chemistry , Thymidine/analogs & derivatives , Thymidine/chemical synthesis , Boron Compounds/chemistry , Combinatorial Chemistry Techniques , Humans , Structure-Activity Relationship , Substrate Specificity , Thymidine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...