Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Med Chem Lett ; 11(5): 754-759, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32435381

ABSTRACT

Lysine-specific demethylase 1 (LSD1 or KDM1A) is a FAD-dependent enzyme that acts as a transcription corepressor or coactivator by regulating the methylation status of histone H3 lysines K4 and K9, respectively. KDM1A represents an attractive target for cancer therapy. While, in the past, the main medicinal chemistry strategy toward KDM1A inhibition was based on the optimization of ligands that irreversibly bind the FAD cofactor within the enzyme catalytic site, we and others have also identified reversible inhibitors. Herein we reported the discovery of 5-imidazolylthieno[3,2-b]pyrroles, a new series of KDM1A inhibitors endowed with picomolar inhibitory potency, active in cells and efficacious after oral administration in murine leukemia models.

2.
J Med Chem ; 59(4): 1501-17, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26702542

ABSTRACT

We report the stereoselective synthesis and biological activity of a novel series of tranylcypromine (TCPA) derivatives (14a-k, 15, 16), potent inhibitors of KDM1A. The new compounds strongly inhibit the clonogenic potential of acute leukemia cell lines. In particular three molecules (14d, 14e, and 14g) showing selectivity versus MAO A and remarkably inhibiting colony formation in THP-1 human leukemia cells, were assessed in mouse for their preliminary pharmacokinetic. 14d and 14e were further tested in vivo in a murine acute promyelocytic leukemia model, resulting 14d the most effective. Its two enantiomers were synthesized: the (1S,2R) enantiomer 15 showed higher activity than its (1R,2S) analogue 16, in both biochemical and cellular assays. Compound 15 exhibited in vivo efficacy after oral administration, determining a 62% increased survival in mouse leukemia model with evidence of KDM1A inhibition. The biological profile of compound 15 supports its further investigation as a cancer therapeutic.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Histone Demethylases/antagonists & inhibitors , Leukemia, Promyelocytic, Acute/drug therapy , Tranylcypromine/chemistry , Tranylcypromine/therapeutic use , Administration, Oral , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Histone Demethylases/metabolism , Humans , Leukemia, Promyelocytic, Acute/metabolism , Structure-Activity Relationship , Tranylcypromine/administration & dosage , Tranylcypromine/pharmacology
3.
Nat Commun ; 5: 5637, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25472497

ABSTRACT

Mcl-1 is a unique Bcl-2 family member that plays crucial roles in apoptosis. Apoptosis-unrelated functions of Mcl-1 are however emerging, further justifying its tight regulation. Here we unravel a novel mechanism of Mcl-1 regulation mediated by the haplo-insufficient tumour suppressor Beclin 1. Beclin 1 negatively modulates Mcl-1 stability in a reciprocal manner whereby depletion of one leads to the stabilization of the other. This co-regulation is independent of autophagy and of their physical interaction. Both Beclin 1 and Mcl-1 are deubiquitinated and thus stabilized by binding to a common deubiquitinase, USP9X. Beclin 1 and Mcl-1 negatively modulate the proteasomal degradation of each other through competitive displacement of USP9X. The analysis of patient-derived melanoma cells and tissue samples shows that the levels of Beclin 1 decrease, while Mcl-1 levels subsequently increase during melanoma progression in a significant inter-dependent manner. The identified inverse co-regulation of Beclin 1 and Mcl-1 represents a mechanism of functional counteraction in cancer.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carcinogenesis , Melanoma/metabolism , Membrane Proteins/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Skin Neoplasms/metabolism , Animals , Autophagy , Beclin-1 , HEK293 Cells , Haploinsufficiency , HeLa Cells , Humans , Melanoma/secondary , Mice , Neoplasm Transplantation , Skin Neoplasms/pathology , Tumor Cells, Cultured , Ubiquitin Thiolesterase/metabolism
4.
ChemMedChem ; 9(7): 1574-85, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24753447

ABSTRACT

Heat-shock protein 90 (Hsp90) is a molecular chaperone involved in the stabilization of key oncogenic signaling proteins, and therefore, inhibition of Hsp90 represents a new strategy in cancer therapy. 2-Amino-7-[4-fluoro-2-(3-pyridyl)phenyl]-4-methyl-7,8-dihydro-6H-quinazolin-5-one oxime is a racemic Hsp90 inhibitor that targets the N-terminal adenosine triphosphatase site. We developed a method to resolve the enantiomers and evaluated their inhibitory activity on Hsp90 and the consequent antitumor effects. The (S) stereoisomer emerged as a potent Hsp90 inhibitor in biochemical and cellular assays. In addition, this enantiomer exhibited high oral bioavailability in mice and excellent antitumor activity in two different human cancer xenograft models.


Subject(s)
Antineoplastic Agents/chemistry , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Oximes/chemistry , Quinazolinones/chemistry , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Cell Line, Tumor , Female , HCT116 Cells , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Nude , Microsomes/metabolism , Molecular Docking Simulation , Neoplasms/drug therapy , Oximes/pharmacology , Oximes/therapeutic use , Protein Binding/drug effects , Protein Structure, Tertiary , Stereoisomerism , Xenograft Model Antitumor Assays
5.
Blood ; 121(17): 3459-68, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23440245

ABSTRACT

Aberrant recruitment of histone deacetylases (HDACs) by the oncogenic fusion protein PML-RAR is involved in the pathogenesis of acute promyelocytic leukemia (APL). PML-RAR, however, is not sufficient to induce disease in mice but requires additional oncogenic lesions during the preleukemic phase. Here, we show that knock-down of Hdac1 and Hdac2 dramatically accelerates leukemogenesis in transgenic preleukemic mice. These events are not restricted to APL because lymphomagenesis driven by deletion of p53 or, to a lesser extent, by c-myc overexpression, was also accelerated by Hdac1 knock-down. In the preleukemic phase of APL, Hdac1 counteracts the activity of PML-RAR in (1) blocking differentiation; (2) impairing genomic stability; and (3) increasing self-renewal in hematopoietic progenitors, as all of these events are affected by the reduction in Hdac1 levels. This led to an expansion of a subpopulation of PML-RAR-expressing cells that is the major source of leukemic stem cells in the full leukemic stage. Remarkably, short-term treatment of preleukemic mice with an HDAC inhibitor accelerated leukemogenesis. In contrast, knock-down of Hdac1 in APL mice led to enhanced survival duration of the leukemic animals. Thus, Hdac1 has a dual role in tumorigenesis: oncosuppressive in the early stages, and oncogenic in established tumor cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Leukemia, Promyelocytic, Acute/etiology , Leukemia, Promyelocytic, Acute/prevention & control , Tumor Suppressor Protein p53/physiology , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/pathology , Female , Flow Cytometry , Genomic Instability , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Leukemia, Promyelocytic, Acute/mortality , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Valproic Acid/pharmacology
6.
ChemMedChem ; 7(4): 709-21, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22354629

ABSTRACT

A series of spiro[chromane-2,4'-piperidine] derivatives based on a previously published lead benzyl spirocycle 1 and bearing various N-aryl and N-alkylaryl substituents on the piperidine ring were prepared as novel histone deacetylase (HDAC) inhibitors. The compounds were evaluated for their abilities to inhibit nuclear HDACs, their in vitro antiproliferative activities, and in vitro ADME profiles. Based on these activities, 4-fluorobenzyl and 2-phenylethyl spirocycles were selected for further characterization. In vivo pharmacokinetic (PK) studies showed that both compounds exhibit an overall lower clearance rate, an increased half-life, and higher AUCs after intravenous and oral administration than spiropiperidine 1 under the conditions used. The improved PK behavior of these two compounds also correlated with superior in vivo antitumor activity in an HCT-116 xenograft model.


Subject(s)
Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Piperidines/chemistry , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Blood Proteins/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochrome P-450 Enzyme Inhibitors , Drug Evaluation, Preclinical , Half-Life , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacokinetics , Injections, Intravenous , Metabolic Clearance Rate , Mice , Mice, Nude , Molecular Structure , Structure-Activity Relationship , Xenograft Model Antitumor Assays
7.
J Med Chem ; 54(8): 3051-64, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21417419

ABSTRACT

New spiro[chromane-2,4'-piperidine] and spiro[benzofuran-2,4'-piperidine] hydroxamic acid derivatives as HDAC inhibitors have been identified by combining privileged structures with a hydroxamic acid moiety as zinc binding group. The compounds were evaluated for their ability to inhibit nuclear extract HDACs and for their in vitro antiproliferative activity on different tumor cell lines. This work resulted in the discovery of spirocycle 30d that shows good oral bioavailability and tumor growth inhibition in an HCT-116 murine xenograft model.


Subject(s)
Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Administration, Oral , Animals , Biological Availability , Cell Line, Tumor , Drug Evaluation, Preclinical , Histone Deacetylase Inhibitors/chemical synthesis , Humans , Hydroxamic Acids/chemical synthesis , Magnetic Resonance Spectroscopy , Mice , Molecular Structure , Spectrometry, Mass, Electrospray Ionization
8.
Nat Cell Biol ; 13(3): 292-302, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21336312

ABSTRACT

Two major mechanisms have been causally implicated in the establishment of cellular senescence: the activation of the DNA damage response (DDR) pathway and the formation of senescence-associated heterochromatic foci (SAHF). Here we show that in human fibroblasts resistant to premature p16(INK4a) induction, SAHF are preferentially formed following oncogene activation but are not detected during replicative cellular senescence or on exposure to a variety of senescence-inducing stimuli. Oncogene-induced SAHF formation depends on DNA replication and ATR (ataxia telangiectasia and Rad3-related). Inactivation of ATM (ataxia telangiectasia mutated) or p53 allows the proliferation of oncogene-expressing cells that retain increased heterochromatin induction. In human cancers, levels of heterochromatin markers are higher than in normal tissues, and are independent of the proliferative index or stage of the tumours. Pharmacological and genetic perturbation of heterochromatin in oncogene-expressing cells increase DDR signalling and lead to apoptosis. In vivo, a histone deacetylase inhibitor (HDACi) causes heterochromatin relaxation, increased DDR, apoptosis and tumour regression. These results indicate that heterochromatin induced by oncogenic stress restrains DDR and suggest that the use of chromatin-modifying drugs in cancer therapies may benefit from the study of chromatin and DDR status of tumours.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage , Heterochromatin/genetics , Neoplasms/metabolism , Oncogenes , Animals , Apoptosis , Cell Line, Tumor , Chromatin/metabolism , DNA Replication , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Microscopy, Fluorescence/methods , Neoplasm Transplantation , Plasmids/metabolism , RNA, Small Interfering/metabolism
9.
Proc Natl Acad Sci U S A ; 107(50): 21535-40, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21106756

ABSTRACT

Epigenetic alterations in the pattern of DNA and histone modifications play a crucial role in cancer development. Analysis of patient samples, however, is hampered by technical limitations in the study of chromatin structure from pathology archives that usually consist of heavily fixed, paraffin-embedded material. Here, we present a methodology [pathology tissue-ChIP (PAT-ChIP)] to extract and immunoprecipitate chromatin from paraffin-embedded patient samples up to several years old. In a pairwise comparison with canonical ChIP, PAT-ChIP showed a high reproducibility of results for several histone marks and an identical ability to detect dynamic changes in chromatin structure upon pharmacological treatment. Finally, we showed that PAT-ChIP can be coupled with high-throughput sequencing (PAT-ChIP-Seq) for the genome-wide analysis of distinct chromatin modifications. PAT-ChIP therefore represents a versatile procedure and diagnostic tool for the analysis of epigenetic alterations in cancer and potentially other diseases.


Subject(s)
Chromatin Immunoprecipitation/methods , Epigenomics/methods , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods , Animals , Humans , Neoplasms/genetics , Neoplasms/pathology , Protein Processing, Post-Translational , Tissue Fixation/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...