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










Publication year range
1.
J Med Chem ; 66(15): 10473-10496, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37427891

ABSTRACT

TYK2 is a key mediator of IL12, IL23, and type I interferon signaling, and these cytokines have been implicated in the pathogenesis of multiple inflammatory and autoimmune diseases such as psoriasis, rheumatoid arthritis, lupus, and inflammatory bowel diseases. Supported by compelling data from human genome-wide association studies and clinical results, TYK2 inhibition through small molecules is an attractive therapeutic strategy to treat these diseases. Herein, we report the discovery of a series of highly selective pseudokinase (Janus homology 2, JH2) domain inhibitors of TYK2 enzymatic activity. A computationally enabled design strategy, including the use of FEP+, was instrumental in identifying a pyrazolo-pyrimidine core. We highlight the utility of computational physics-based predictions used to optimize this series of molecules to identify the development candidate 30, a potent, exquisitely selective cellular TYK2 inhibitor that is currently in Phase 2 clinical trials for the treatment of psoriasis and psoriatic arthritis.


Subject(s)
Arthritis, Rheumatoid , Autoimmune Diseases , Psoriasis , Humans , TYK2 Kinase , Genome-Wide Association Study , Autoimmune Diseases/drug therapy , Psoriasis/drug therapy
2.
Front Immunol ; 13: 987298, 2022.
Article in English | MEDLINE | ID: mdl-36090975

ABSTRACT

A critical feature of cancer is the ability to induce immunosuppression and evade immune responses. Tumor-induced immunosuppression diminishes the effectiveness of endogenous immune responses and decreases the efficacy of cancer immunotherapy. In this study, we describe a new immunosuppressive pathway in which adenosine promotes Casitas B-lineage lymphoma b (Cbl-b)-mediated Notch1 degradation, causing suppression of CD8+ T-cells effector functions. Genetic knockout and pharmacological inhibition of Cbl-b prevents Notch1 degradation in response to adenosine and reactivates its signaling. Reactivation of Notch1 results in enhanced CD8+ T-cell effector functions, anti-cancer response and resistance to immunosuppression. Our work provides evidence that targeting the Cbl-b-Notch1 axis is a novel promising strategy for cancer immunotherapy.


Subject(s)
Lymphoma , Neoplasms , Adenosine , CD8-Positive T-Lymphocytes , Humans , Immunotherapy , Receptor, Notch1/genetics , Receptor, Notch1/metabolism
3.
Bioorg Med Chem Lett ; 73: 128891, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35842205

ABSTRACT

TYK2 is a member of the JAK family of kinases and a key mediator of IL-12, IL-23, and type I interferon signaling. These cytokines have been implicated in the pathogenesis of multiple inflammatory and autoimmune diseases such as psoriasis, rheumatoid arthritis, lupus, and inflammatory bowel diseases. Supported by compelling data from human genetic association studies, TYK2 inhibition is an attractive therapeutic strategy for these diseases. Herein, we report the discovery of a series of highly selective catalytic site TYK2 inhibitors designed using FEP+ and structurally enabled design starting from a virtual screen hit. We highlight the structure-based optimization to identify a lead candidate 30, a potent cellular TYK2 inhibitor with excellent selectivity, pharmacokinetic properties, and in vivo efficacy in a mouse psoriasis model.


Subject(s)
Psoriasis , TYK2 Kinase , Animals , Humans , Janus Kinases , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Psoriasis/drug therapy , Rodentia
4.
Diabetes Obes Metab ; 22(10): 1827-1836, 2020 10.
Article in English | MEDLINE | ID: mdl-32476252

ABSTRACT

AIM: Type 1 diabetes (T1D) is a chronic autoimmune disease leading to progressive loss of pancreatic beta cells. Interferon (IFN)-α plays a critical role in the crosstalk between pancreatic beta cells and the immune system in early insulitis. In human beta cells IFNα signals through JAK1 and TYK2, leading to endoplasmic reticulum stress, inflammation and HLA class I overexpression. IFNα, acting synergistically with IL-1ß, induces apoptosis. Polymorphisms in TYK2 that decrease its activity are associated with protection against T1D, and we hypothesized that pharmacological inhibitors that specifically target TYK2 could protect human beta cells against the deleterious effects of IFNα. MATERIALS AND METHODS: Two TYK2 inhibitors provided by Nimbus Lakshmi were tested in human insulin-producing EndoC-ßH1 cells and human islets to evaluate their effect on IFNα signalling, beta-cell function and susceptibility to viral infection using RT-qPCR, western blot, immunofluorescence, ELISA and nuclear dyes. RESULTS: The two TYK2 inhibitors tested prevented IFNα-induced human beta-cell gene expression in a dose-dependent manner. They also protected human islets against IFNα + IL-1ß-induced apoptosis. Importantly, these inhibitors did not modify beta-cell function or their survival following infection with the potential diabetogenic coxsackieviruses CVB1 and CVB5. CONCLUSIONS: The two TYK2 inhibitors tested inhibit the IFNα signalling pathway in human beta cells, decreasing its pro-inflammatory and pro-apoptotic effects without sensitizing the cells to viral infection. The preclinical findings could pave the way for future clinical trials with TYK2 inhibitors for the prevention and treatment of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Apoptosis , Cytoprotection , Diabetes Mellitus, Type 1/drug therapy , Endoplasmic Reticulum Stress , Humans , TYK2 Kinase/genetics
5.
Nature ; 568(7753): 566-570, 2019 04.
Article in English | MEDLINE | ID: mdl-30944472

ABSTRACT

ATP-citrate lyase (ACLY) is a central metabolic enzyme and catalyses the ATP-dependent conversion of citrate and coenzyme A (CoA) to oxaloacetate and acetyl-CoA1-5. The acetyl-CoA product is crucial for the metabolism of fatty acids6,7, the biosynthesis of cholesterol8, and the acetylation and prenylation of proteins9,10. There has been considerable interest in ACLY as a target for anti-cancer drugs, because many cancer cells depend on its activity for proliferation2,5,11. ACLY is also a target against dyslipidaemia and hepatic steatosis, with a compound currently in phase 3 clinical trials4,5. Many inhibitors of ACLY have been reported, but most of them have weak activity5. Here we report the development of a series of low nanomolar, small-molecule inhibitors of human ACLY. We have also determined the structure of the full-length human ACLY homo-tetramer in complex with one of these inhibitors (NDI-091143) by cryo-electron microscopy, which reveals an unexpected mechanism of inhibition. The compound is located in an allosteric, mostly hydrophobic cavity next to the citrate-binding site, and requires extensive conformational changes in the enzyme that indirectly disrupt citrate binding. The observed binding mode is supported by and explains the structure-activity relationships of these compounds. This allosteric site greatly enhances the 'druggability' of ACLY and represents an attractive target for the development of new ACLY inhibitors.


Subject(s)
ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , ATP Citrate (pro-S)-Lyase/ultrastructure , Benzyl Compounds/pharmacology , Cryoelectron Microscopy , Enzyme Inhibitors/pharmacology , ATP Citrate (pro-S)-Lyase/chemistry , ATP Citrate (pro-S)-Lyase/metabolism , Adenosine Diphosphate/metabolism , Allosteric Regulation/drug effects , Benzyl Compounds/chemistry , Binding Sites/drug effects , Citric Acid/metabolism , Enzyme Inhibitors/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Domains , Protein Multimerization , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 19(19): 5684-8, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19699639

ABSTRACT

We have identified a series of diphenylmethylene hydroxamic acids as novel and selective HDAC class IIa inhibitors. The original hit, N-hydroxy-2,2-diphenylacetamide (6), has sub-micromolar class IIa HDAC inhibitory activity, while the rigidified oxygen analogue, N-hydroxy-9H-xanthene-9-carboxamide (13), is slightly more selective for HDAC7 with an IC(50) of 0.05muM. Substitution of 6 allows for the modulation of selectivity and potency amongst the class IIa HDAC isotypes.


Subject(s)
Diphenylacetic Acids/chemistry , Enzyme Inhibitors/chemistry , Hydroxamic Acids/chemistry , Xanthenes/chemistry , Cell Line , Diphenylacetic Acids/chemical synthesis , Diphenylacetic Acids/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Structure-Activity Relationship , Xanthenes/chemical synthesis , Xanthenes/pharmacology
7.
Bioorg Med Chem Lett ; 19(3): 688-92, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19111466

ABSTRACT

In an effort to identify HDAC isoform selective inhibitors, we designed and synthesized novel, chiral 3,4-dihydroquinoxalin-2(1H)-one and piperazine-2,5-dione aryl hydroxamates showing selectivity (up to 40-fold) for human HDAC6 over other class I/IIa HDACs. The observed selectivity and potency (IC(50) values 10-200 nM against HDAC6) is markedly dependent on the absolute configuration of the chiral moiety, and suggests new possibilities for use of chiral compounds in selective HDAC isoform inhibition.


Subject(s)
Enzyme Inhibitors/chemistry , Histone Deacetylase Inhibitors , Histone Deacetylases/chemistry , Acetylation , Catalytic Domain , Chemistry, Pharmaceutical/methods , Drug Design , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Histones/chemistry , Humans , Hydroxamic Acids/pharmacology , Inhibitory Concentration 50 , Models, Chemical , Piperazine , Piperazines/chemistry , Protein Isoforms , Tubulin/chemistry
8.
Bioorg Med Chem Lett ; 19(3): 644-9, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19114304

ABSTRACT

Analogues of the clinical compound MGCD0103 (A) were designed and synthesized. These compounds inhibit recombinant human HDAC1 with IC(50) values in the sub-micromolar range. In human cancer cells growing in culture these compounds induce hyperacetylation of histones, cause expression of the tumor suppressor protein p21(WAF1/CIP1), and inhibit cellular proliferation. Lead molecule of the series, compound 25 is metabolically stable, possesses favorable pharmacokinetic characteristics and is orally active in vivo in different mouse tumor xenograft models.


Subject(s)
Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzamides/chemical synthesis , Cell Line, Tumor , Cell Proliferation , Chemistry, Pharmaceutical/methods , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drug Design , Enzyme Inhibitors/chemistry , Histone Deacetylase Inhibitors , Humans , Inhibitory Concentration 50 , Mice , Neoplasm Transplantation , Pyrimidines/chemical synthesis , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 19(2): 336-40, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19084395

ABSTRACT

The sulfamide moiety has been utilized to design novel HDAC inhibitors. The potency and selectivity of these inhibitors were influenced both by the nature of the scaffold, and the capping group. Linear long-chain-based analogs were primarily HDAC6-selective, while analogs based on the lysine scaffold resulted in potent HDAC1 and HDAC6 inhibitors.


Subject(s)
Amides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors
10.
J Med Chem ; 51(14): 4072-5, 2008 Jul 24.
Article in English | MEDLINE | ID: mdl-18570366

ABSTRACT

The design, synthesis, and biological evaluation of N-(2-aminophenyl)-4-[(4-pyridin-3-ylpyrimidin-2-ylamino)methyl]benzamide 8 (MGCD0103) is described. Compound 8 is an isotype-selective small molecule histone deacetylase (HDAC) inhibitor that selectively inhibits HDACs 1-3 and 11 at submicromolar concentrations in vitro. 8 blocks cancer cell proliferation and induces histone acetylation, p21 (cip/waf1) protein expression, cell-cycle arrest, and apoptosis. 8 is orally bioavailable, has significant antitumor activity in vivo, has entered clinical trials, and shows promise as an anticancer drug.


Subject(s)
Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Pyrimidines/pharmacology , Administration, Oral , Animals , Benzamides/administration & dosage , Benzamides/chemistry , Dogs , Drug Screening Assays, Antitumor , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Male , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 18(4): 1502-6, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18207391

ABSTRACT

The synthesis and biological evaluation of a variety of 4-(heteroarylaminomethyl)-N-(2-aminophenyl)-benzamides and their analogs is described. Some of these compounds were shown to inhibit HDAC1 with IC(50) values below the micromolar range, induce hyperacetylation of histones, upregulate expression of the tumor suppressor p21(WAF1/Cip1), and inhibit proliferation of human cancer cells. In addition, certain compounds of this class were active in several human tumor xenograft models in vivo.


Subject(s)
Aniline Compounds/chemical synthesis , Aniline Compounds/pharmacology , Benzamides/chemical synthesis , Benzamides/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Aniline Compounds/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzamides/chemistry , Breast/cytology , Breast/drug effects , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Enzyme Inhibitors/chemistry , Epithelial Cells/cytology , Epithelial Cells/drug effects , HCT116 Cells , Histone Deacetylase 1 , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacology , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 18(3): 1067-71, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18160287

ABSTRACT

Inhibition of histone deacetylases (HDAC) is emerging as a new strategy in human cancer therapy. The synthesis and biological evaluation of a variety of 4-(heteroarylaminomethyl)-N-(2-aminophenyl)-benzamides is presented herein. From the different series bearing a six-membered heteroaromatic ring studied, the s-triazine series showed the best HDAC1 enzyme and in vitro anti-proliferative activities with IC(50) values below micromolar range. Some of these compounds can also significantly reduce tumor growth in human tumor xenograft models in mice.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzamides/chemical synthesis , Benzamides/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Pyrimidines/pharmacology , Triazines/chemical synthesis , Triazines/pharmacology , Animals , Antineoplastic Agents/chemistry , Benzamides/chemistry , Disease Models, Animal , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Mice , Structure-Activity Relationship , Triazines/chemistry
13.
Bioorg Med Chem Lett ; 17(24): 6729-33, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17977726

ABSTRACT

A variety of N-(2-amino-phenyl)-4-(heteroarylmethyl)-benzamides were designed and synthesized. These compounds were shown to inhibit recombinant human HDAC1 with IC(50) values in the sub-micromolar range. In human cancer cells growing in culture these compounds induced hyperacetylation of histones, induced the expression of the tumor suppressor protein p21(WAF1/Cip1), and inhibited cellular proliferation. Certain compounds of this class also showed in vivo activity in various human tumor xenograft models in mice.


Subject(s)
Benzamides/chemistry , Benzamides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Amination , Animals , Binding Sites , Cell Line , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Methylation , Mice , Models, Molecular , Molecular Structure , Niacin/pharmacology , Structure-Activity Relationship , Urea/chemistry , Vasodilation/drug effects , ortho-Aminobenzoates/chemistry
14.
J Med Chem ; 50(23): 5543-6, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17941625

ABSTRACT

Significant effort is being made to understand the role of HDAC isotypes in human cancer and to develop antitumor agents with better therapeutic windows. A part of this endeavor was the exploration of the 14 A internal cavity adjacent to the enzyme catalytic site, which led to the design and synthesis of compound 4 with the unusual bis(aryl)-type pharmacophore. SAR studies around this lead resulted in optimization to potent, selective, nonhydroxamic acid HDAC inhibitors.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzamides/chemical synthesis , Histone Deacetylase Inhibitors , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Benzamides/chemistry , Benzamides/pharmacology , Catalytic Domain , Cell Line , Histone Deacetylase 1 , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Models, Molecular , NADH, NADPH Oxidoreductases/chemistry , NADH, NADPH Oxidoreductases/metabolism , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 16(15): 4048-52, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16713259

ABSTRACT

Inhibition of histone deacetylases (HDACs) is emerging as a new strategy in human cancer therapy. Novel 2-aminophenyl benzamides and acrylamides, that can inhibit human HDAC enzymes and induce hyperacetylation of histones in human cancer cells, have been designed and synthesized. These compounds selectively inhibit proliferation and cause cell cycle arrest in various human cancer cells but not in normal cells. The growth inhibition of 2-aminophenyl benzamides and acrylamides against human cancer cells in vitro is reversible and is dependent on the induction of histone acetylation. Compounds of this class can significantly reduce tumor growth in human tumor xenograft models.


Subject(s)
Acrylamides/pharmacology , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Acrylamides/chemistry , Benzamides/chemistry , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Humans , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 14(1): 283-7, 2004 Jan 05.
Article in English | MEDLINE | ID: mdl-14684344

ABSTRACT

A variety of omega-substituted alkanoic acid (2-amino-phenyl)-amides were designed and synthesized. These compounds were shown to inhibit recombinant human histone deacetylases (HDACs) with IC(50) values in the low micromolar range and induce hyperacetylation of histones in whole cells. They induced expression of p21WAF1/Cip1 and caused cell-cycle arrest in human cancer cells. Compounds in this class showed efficacy in human tumor xenograft models.


Subject(s)
Amides/chemistry , Amides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , Histone Deacetylases/metabolism , Humans
17.
J Med Chem ; 46(5): 820-30, 2003 Feb 27.
Article in English | MEDLINE | ID: mdl-12593661

ABSTRACT

A series of sulfonamide hydroxamic acids and anilides have been synthesized and studied as histone deacetylase (HDAC) inhibitors that can induce hyperacetylation of histones in human cancer cells. The inhibition of HDAC activity represents a novel approach for intervening in cell cycle regulation. The lead candidates were screened in a panel of human tumor and normal cell lines. They selectively inhibit proliferation, cause cell cycle blocks, and induce apoptosis in human cancer cells but not in normal cells. The structure-activity relationships, the antiproliferative activity, and the in vivo efficacy are described.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors , Hydroxamic Acids/chemical synthesis , Sulfonamides/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Transplantation, Heterologous
18.
Cancer Res ; 62(15): 4325-30, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12154036

ABSTRACT

Inhibition of histone deacetylases (HDACs) is emerging as a new strategy in human cancer therapy. We have designed and synthesized novel nonhydroxamate sulfonamide anilides that can inhibit human HDAC enzymes and can induce hyperacetylation of histones in human cancer cells. These compounds selectively inhibit proliferation and cause cell cycle blocks in various human cancer cells but not in normal cells. The growth inhibitory activity of sulfonamide anilides against human cancer cells in vitro is reversible and is dependent on the induction of histone acetylation. One of these compounds (Compound 2) can significantly reduce tumor growth of implanted human colon tumors in nude mice. Unlike another anilide-based HDAC inhibitor, MS-275, which decreases both red and white blood counts and reduces spleen weights in mice, Compound 2 does not exhibit noticeable toxicity. By using cDNA array analysis, we have identified downstream genes whose expression is altered by Compound 2 in human cancer cells. In correlation with its antitumor activity both in vitro and in vivo, Compound 2 induces expression of p21(WAF1/Cip1), gelsolin, and keratin 19, while down-regulating expression of cyclin A and cyclin B1 in human cancer cells in a dose-dependent manner. Our results suggest that sulfonamide anilides are novel HDAC inhibitors and may be useful as antiproliferative agents in cancer chemotherapy.


Subject(s)
Anilides/pharmacology , Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors , Sulfonamides/pharmacology , Acetylation/drug effects , Anilides/toxicity , Animals , Antineoplastic Agents/toxicity , Benzamides/pharmacology , Benzamides/toxicity , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin A/biosynthesis , Cyclin B/biosynthesis , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Female , G2 Phase/drug effects , Histones/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitosis/drug effects , Pyridines/pharmacology , Pyridines/toxicity , Sulfonamides/toxicity , Xenograft Model Antitumor Assays
19.
J Med Chem ; 45(13): 2877-85, 2002 Jun 20.
Article in English | MEDLINE | ID: mdl-12061890

ABSTRACT

A series of new, structurally simple trichostatin A (TSA)-like straight chain hydroxamates were prepared and evaluated for their ability to inhibit partially purified human histone deacetylase 1 (HDAC-1). Some of these compounds such as 8m, 8n, 12, and 15b exhibited potent HDAC inhibitory activity with low nanomolar IC(50) values, comparable to natural TSA. These compounds induce hyperacetylation of histones in T24 human cancer cells and significantly inhibit proliferation in various human cancer cells. They also induce expression of p21 and cause cell cycle blocks in human cancer cells. In this paper, we describe the synthesis of these new compounds as well as structure-activity relationship results from enzyme inhibition and alterations in cellular function.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors , Hydroxamic Acids/chemistry , Acetylation , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histones/drug effects , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , Recombinant Proteins/antagonists & inhibitors , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...