Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 501
Filtrer
1.
Lancet Haematol ; 11(7): e487-e498, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38824932

RÉSUMÉ

BACKGROUND: Iadademstat is a potent, selective, oral inhibitor of both the enzymatic and scaffolding activities of the transcriptional repressor lysine-specific demethylase 1 (LSD1; also known as KDM1A) that showed promising early activity and safety in a phase 1 trial and strong preclinical synergy with azacitidine in acute myeloid leukaemia cell lines. Therefore, we aimed to investigate the combination of iadademstat and azacitidine for the treatment of adult patients with newly diagnosed acute myeloid leukaemia. METHODS: The open-label, phase 2a, dose-finding ALICE study was conducted at six hospitals in Spain and enrolled patients aged 18 years or older with newly diagnosed acute myeloid leukaemia not eligible for intensive chemotherapy and an ECOG performance status of 0-2. In the dose escalation portion of the trial, patients received a starting dose of iadademstat at 90 µg/m2 per day (with de-escalation to 60 µg/m2 per day and escalation up to 140 µg/m2 per day) orally, for 5 days on, 2 days off weekly, with azacitidine 75 mg/m2 subcutaneously, for seven of 28 days. The primary objectives were safety (analysed in the safety analysis set; all patients who received at least one dose of study treatment) and establishing the recommended phase 2 dose; secondary objectives included response rates in the efficacy analysis set (all patients who had at least one efficacy assessment). This study is registered on EudraCT (EudraCT 2018-000482-36) and has been completed. FINDINGS: Between Nov 12, 2018, and Sept 30, 2021, 36 patients with newly diagnosed acute myeloid leukaemia were enrolled; the median age was 76 (IQR 74-79) years, all patients were White, 18 (50%) were male, and 18 (50%) were female, and all had intermediate-risk or adverse-risk acute myeloid leukaemia. The median follow-up was 22 (IQR 16-31) months. The most frequent (≥10%) adverse events considered to be related to treatment were decreases in platelet (25 [69%]) and neutrophil (22 [61%]) counts (all grade 3-4) and anaemia (15 [42%]; of which ten [28%] were grade 3-4). Three patients had treatment-related serious adverse events (one fatal grade 5 intracranial haemorrhage, one grade 3 differentiation syndrome, and one grade 3 febrile neutropenia). Based on safety, pharmacokinetic and pharmacodynamic data, and efficacy, the recommended phase 2 dose of iadademstat was 90 µg/m2 per day with azacitidine. 22 (82%; 95% CI 62-94) of 27 patients in the efficacy analysis set had an objective response. 14 (52%) of 27 patients had complete remission or complete remission with incomplete haematological recovery; of these, ten of 11 evaluable for measurable residual disease achieved negativity. In the safety analysis set, 22 (61%) of 36 patients had an objective response. INTERPRETATION: The combination of iadademstat and azacitidine has a manageable safety profile and shows promising responses in patients with newly diagnosed acute myeloid leukaemia, including those with high-risk prognostic factors. FUNDING: Oryzon Genomics and Spain's Ministerio de Ciencia, Innovacion y Universidades (MICIU)-Agencia Estatal de Investigacion (AEI).


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Azacitidine , Leucémie aigüe myéloïde , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Azacitidine/usage thérapeutique , Azacitidine/administration et posologie , Azacitidine/effets indésirables , Mâle , Femelle , Sujet âgé , Adulte d'âge moyen , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Histone Demethylases/antagonistes et inhibiteurs , Adulte , Relation dose-effet des médicaments , Sujet âgé de 80 ans ou plus , Cyclohexanes , Diamines
2.
Chem Pharm Bull (Tokyo) ; 72(7): 630-637, 2024.
Article de Anglais | MEDLINE | ID: mdl-38945939

RÉSUMÉ

Alzheimer's disease (AD) is the leading cause of senile dementia, and the rapid increase in the frequency of AD cases has been attributed to population aging. However, current drugs have difficulty adequately suppressing symptoms and there is still a medical need for symptomatic agents. On the other hand, it has recently become clear that epigenetic dysfunctions are deeply involved in the development of cognitive impairments. Therefore, epigenetics-related proteins have attracted much attention as drug targets for AD. Early-developed epigenetic inhibitors were inappropriate for AD treatment because of their limited potential for oral administration, blood-brain barrier penetration, high target selectivity, and sufficient dose-limiting toxicity which are essential properties for small molecule drugs targeting chronic neurodegenerative diseases such as AD. In recent years, drug discovery studies have been actively performed to overcome such problems and several novel inhibitors targeting the epigenetics-related proteins are of interest as promising AD therapeutic agents. Here, we review the small molecule inhibitors of histone deacetylase (HDAC), lysine-specific demethylase 1 (LSD1) or bromodomains and extra-terminal domain (BET) protein, that enable memory function improvement in AD model mice.


Sujet(s)
Maladie d'Alzheimer , Épigenèse génétique , Inhibiteurs de désacétylase d'histone , Histone Demethylases , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Humains , Animaux , Épigenèse génétique/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/pharmacologie , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Histone deacetylases/métabolisme
3.
Molecules ; 29(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38893322

RÉSUMÉ

The Mediterranean diet well known for its beneficial health effects, including mood enhancement, is characterised by the relatively high consumption of extra virgin olive oil (EVOO), which is rich in bioactive phenolic compounds. Over 200 phenolic compounds have been associated with Olea europaea, and of these, only a relatively small fraction have been characterised. Utilising the OliveNetTM library, phenolic compounds were investigated as potential inhibitors of the epigenetic modifier lysine-specific demethylase 1 (LSD1). Furthermore, the compounds were screened for inhibition of the structurally similar monoamine oxidases (MAOs) which are directly implicated in the pathophysiology of depression. Molecular docking highlighted that olive phenolics interact with the active site of LSD1 and MAOs. Protein-peptide docking was also performed to evaluate the interaction of the histone H3 peptide with LSD1, in the presence of ligands bound to the substrate-binding cavity. To validate the in silico studies, the inhibitory activity of phenolic compounds was compared to the clinically approved inhibitor tranylcypromine. Our findings indicate that olive phenolics inhibit LSD1 and the MAOs in vitro. Using a cell culture model system with corticosteroid-stimulated human BJ fibroblast cells, the results demonstrate the attenuation of dexamethasone- and hydrocortisone-induced MAO activity by phenolic compounds. The findings were further corroborated using human embryonic stem cell (hESC)-derived neurons stimulated with all-trans retinoic acid. Overall, the results indicate the inhibition of flavin adenine dinucleotide (FAD)-dependent amine oxidases by olive phenolics. More generally, our findings further support at least a partial mechanism accounting for the antidepressant effects associated with EVOO and the Mediterranean diet.


Sujet(s)
Simulation de docking moléculaire , Inhibiteurs de la monoamine oxydase , Monoamine oxidase , Olea , Phénols , Humains , Inhibiteurs de la monoamine oxydase/pharmacologie , Inhibiteurs de la monoamine oxydase/composition chimique , Monoamine oxidase/métabolisme , Monoamine oxidase/composition chimique , Olea/composition chimique , Phénols/pharmacologie , Phénols/composition chimique , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Dépression/traitement médicamenteux , Huile d'olive/composition chimique , Simulation numérique
4.
J Chem Inf Model ; 64(12): 4773-4780, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38837697

RÉSUMÉ

Lysine-specific demethylase 1 (LSD1), a highly sophisticated epigenetic regulator, orchestrates a range of critical cellular processes, holding promising therapeutic potential for treating diverse diseases. However, the clinical research progress targeting LSD1 is very slow. After 20 years of research, only one small-molecule drug, BEA-17, targeting the degradation of LSD1 and CoREST has been approved by the U.S. Food and Drug Administration. The primary reason for this may be the lack of abundant structural data regarding its intricate functions. To gain a deeper understanding of its conformational dynamics and guide the drug design process, we conducted molecular dynamics simulations to explore the conformational states of LSD1 in the apo state and under the influence of cofactors of flavin adenine dinucleotide (FAD) and CoREST. Our results showed that, across all states, the substrate binding pocket exhibited high flexibility, whereas the FAD binding pocket remained more stable. These distinct dynamical properties are essential for LSD1's ability to bind various substrates while maintaining efficient demethylation activity. Both pockets can be enlarged by merging with adjacent pockets, although only the substrate binding pocket can shrink into smaller pockets. These new pocket shapes can inform inhibitor design, particularly for selectively FAD-competitive inhibitors of LSD1, given the presence of numerous FAD-dependent enzymes in the human body. More interestingly, in the absence of FAD binding, the united substrate and FAD binding pocket are partitioned by the conserved residue of Tyr761, offering valuable insights for the design of inhibitors that disrupt the crucial steric role of Tyr761 and the redox role of FAD. Additionally, we identified pockets that positively or negatively correlate with the substrate and FAD binding pockets, which can be exploited for the design of allosteric or concurrent inhibitors. Our results reveal the intricate dynamical properties of LSD1 as well as multiple novel conformational states, which deepen our understanding of its sophisticated functions and aid in the rational design of new inhibitors.


Sujet(s)
Conception de médicament , Antienzymes , Flavine adénine dinucléotide , Histone Demethylases , Simulation de dynamique moléculaire , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Histone Demethylases/composition chimique , Flavine adénine dinucléotide/métabolisme , Flavine adénine dinucléotide/composition chimique , Sites de fixation , Humains , Antienzymes/pharmacologie , Antienzymes/composition chimique , Antienzymes/métabolisme , Spécificité du substrat , Conformation des protéines , Liaison aux protéines
5.
J Cancer Res Clin Oncol ; 150(5): 253, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38748285

RÉSUMÉ

BACKGROUND: Lysine-specific demethylase 1 (LSD1) is highly expressed in a variety of malignant tumors, rendering it a crucial epigenetic target for anti-tumor therapy. Therefore, the inhibition of LSD1 activity has emerged as a promising innovative therapeutic approach for targeted cancer treatment. METHODS: In our study, we employed innovative structure-based drug design methods to meticulously select compounds from the ZINC15 database. Utilizing virtual docking, we evaluated docking scores and binding modes to identify potential inhibitors. To further validate our findings, we harnessed molecular dynamic simulations and conducted meticulous biochemical experiments to deeply analyze the binding interactions between the protein and compounds. RESULTS: Our results showcased that ZINC10039815 exhibits an exquisite binding mode with LSD1, fitting perfectly into the active pocket and forming robust interactions with multiple critical residues of the protein. CONCLUSIONS: With its significant inhibitory effect on LSD1 activity, ZINC10039815 emerges as a highly promising candidate for the development of novel LSD1 inhibitors.


Sujet(s)
Antienzymes , Histone Demethylases , Simulation de docking moléculaire , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Histone Demethylases/composition chimique , Humains , Antienzymes/pharmacologie , Antienzymes/composition chimique , Simulation de dynamique moléculaire , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Conception de médicament , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme
6.
Biochemistry ; 63(11): 1369-1375, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38742921

RÉSUMÉ

Lysine specific demethylase-1 (LSD1) serves as a regulator of transcription and represents a promising epigenetic target for anticancer treatment. LSD1 inhibitors are in clinical trials for the treatment of Ewing's sarcoma (EWS), acute myeloid leukemia, and small cell lung cancer, and the development of robust inhibitors requires accurate methods for probing demethylation, potency, and selectivity. Here, the inhibition kinetics on the H3K4me2 peptide and nucleosome substrates was examined, comparing the rates of demethylation in the presence of reversible [CC-90011 (PD) and SP-2577 (SD)] and irreversible [ORY-1001 (ID) and tranylcypromine (TCP)] inhibitors. Inhibitors were also subject to viability studies in three human cell lines and Western blot assays to monitor H3K4me2 nucleosome levels in EWS (TC-32) cells, enabling a correlation of drug potency, inhibition in vitro, and cell-based studies. For example, SP-2577, a drug in clinical trials for EWS, inhibits activity on small peptide substrates (Ki = 60 ± 20 nM) using an indirect coupled assay but does not inhibit demethylation on H3K4me2 peptides or nucleosomes using direct Western blot approaches. In addition, the drug has no effect on H3K4me2 levels in TC-32 cells. These data show that SP-2577 is not an LSD1 enzyme inhibitor, although the drug may function independent of demethylation due to its cytotoxic selectivity in TC-32 cells. Taken together, this work highlights the pitfalls of using coupled assays to ascribe a drug's mode of action, emphasizes the use of physiologically relevant substrates in epigenetic drug targeting strategies, and provides insight into the development of substrate-selective inhibitors of LSD1.


Sujet(s)
Antinéoplasiques , Histone Demethylases , Nucléosomes , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Humains , Nucléosomes/métabolisme , Nucléosomes/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antienzymes/pharmacologie , Antienzymes/composition chimique , Lignée cellulaire tumorale , Histone/métabolisme , Tranylcypromine/pharmacologie , Spécificité du substrat , Cinétique
7.
Nat Commun ; 15(1): 4327, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773088

RÉSUMÉ

The antitumor efficacy of adoptively transferred T cells is limited by their poor persistence, in part due to exhaustion, but the underlying mechanisms and potential interventions remain underexplored. Here, we show that targeting histone demethylase LSD1 by chemical inhibitors reshapes the epigenome of in vitro activated and expanded CD8+ T cells, and potentiates their antitumor efficacy. Upon T cell receptor activation and IL-2 signaling, a timely and transient inhibition of LSD1 suffices to improve the memory phenotype of mouse CD8+ T cells, associated with a better ability to produce multiple cytokines, resist exhaustion, and persist in both antigen-dependent and -independent manners after adoptive transfer. Consequently, OT1 cells primed with LSD1 inhibitors demonstrate an enhanced antitumor effect in OVA-expressing solid tumor models implanted in female mice, both as a standalone treatment and in combination with PD-1 blockade. Moreover, priming with LSD1 inhibitors promotes polyfunctionality of human CD8+ T cells, and increases the persistence and antitumor efficacy of human CD19-CAR T cells in both leukemia and solid tumor models. Thus, pharmacological inhibition of LSD1 could be exploited to improve adoptive T cell therapy.


Sujet(s)
Lymphocytes T CD8+ , Histone Demethylases , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Souris , Humains , Femelle , Souris de lignée C57BL , Immunothérapie adoptive/méthodes , Lignée cellulaire tumorale , Activation des lymphocytes/effets des médicaments et des substances chimiques , Transfert adoptif , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Interleukine-2/métabolisme , Antigènes CD19/métabolisme , Antigènes CD19/immunologie , Mémoire immunologique/effets des médicaments et des substances chimiques
8.
BMC Vet Res ; 20(1): 187, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38730463

RÉSUMÉ

BACKGROUND: Porcine epidemic diarrhea virus (PEDV), a type of coronavirus, is one of the main pathogens that can infect pigs of all ages. It causes diarrhea and acute death of newborn piglets, resulting in massive economic losses to the worldwide swine industry. While vaccination remains the primary approach in combating PEDV, it often fails to address all the challenges posed by the infection, particularly in light of the emergence of evolving mutant strains. Therefore, there is a critical need to identify potent antiviral drugs that can effectively safeguard pigs against PEDV infection. RESULTS: In this study, the antiviral efficacy of SP2509, a specific antagonist of Lysine-specific demethylase 1(LSD1), was evaluated in vitro. The RT-qPCR, Western blot, TCID50, and IFA showed that at a concentration of 1µmol/L, SP2509 significantly inhibited PEDV infection. Additionally, viral life cycle assays showed that SP2509 operates by impeding PEDV internalization and replication rather than attachment and release. Regarding mechanism, in Huh-7 cells, knockdowns LSD1 can suppress PEDV replication. This indicated that the inhibition effect of SP2509 on PEDV largely depends on the activity of its target protein, LSD1. CONCLUSION: Our results in vitro show that SP2509 can inhibit PEDV infection during the internalization and replication stage and revealed a role of LSD1 as a restriction factor for PEDV. These imply that LSD1 might be a target for interfering with the viral infection, and SP2509 could be developed as an effective anti-PEDV agent.


Sujet(s)
Antiviraux , Histone Demethylases , Virus de la diarrhée porcine épidémique , Réplication virale , Virus de la diarrhée porcine épidémique/effets des médicaments et des substances chimiques , Animaux , Antiviraux/pharmacologie , Réplication virale/effets des médicaments et des substances chimiques , Histone Demethylases/antagonistes et inhibiteurs , Suidae , Chlorocebus aethiops , Maladies des porcs/virologie , Maladies des porcs/traitement médicamenteux , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/traitement médicamenteux , Infections à coronavirus/virologie , Cellules Vero
9.
Mol Cancer ; 23(1): 109, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38769556

RÉSUMÉ

Breast cancer (BC) is the most frequent malignant cancer diagnosis and is a primary factor for cancer deaths in women. The clinical subtypes of BC include estrogen receptor (ER) positive, progesterone receptor (PR) positive, human epidermal growth factor receptor 2 (HER2) positive, and triple-negative BC (TNBC). Based on the stages and subtypes of BC, various treatment methods are available with variations in the rates of progression-free disease and overall survival of patients. However, the treatment of BC still faces challenges, particularly in terms of drug resistance and recurrence. The study of epigenetics has provided new ideas for treating BC. Targeting aberrant epigenetic factors with inhibitors represents a promising anticancer strategy. The KDM5 family includes four members, KDM5A, KDM5B, KDM5C, and KDMD, all of which are Jumonji C domain-containing histone H3K4me2/3 demethylases. KDM5 proteins have been extensively studied in BC, where they are involved in suppressing or promoting BC depending on their specific upstream and downstream pathways. Several KDM5 inhibitors have shown potent BC inhibitory activity in vitro and in vivo, but challenges still exist in developing KDM5 inhibitors. In this review, we introduce the subtypes of BC and their current therapeutic options, summarize KDM5 family context-specific functions in the pathobiology of BC, and discuss the outlook and pitfalls of KDM5 inhibitors in this disease.


Sujet(s)
Tumeurs du sein , Histone Demethylases , Thérapie moléculaire ciblée , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/thérapie , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Animaux , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Épigenèse génétique , Régulation de l'expression des gènes tumoraux , Jumonji Domain-Containing Histone Demethylases/métabolisme , Jumonji Domain-Containing Histone Demethylases/antagonistes et inhibiteurs , Jumonji Domain-Containing Histone Demethylases/génétique , Marqueurs biologiques tumoraux
10.
Bioorg Chem ; 147: 107336, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636431

RÉSUMÉ

In this series we report the structure-based design, synthesis and anticancer activity evaluation of a series of eighteen cyclopropylamine containing cyanopyrimidine derivatives. The computational predictions of ADMET properties revealed appropriate aqueous solubility, high GI absorption, no BBB permeability, no Lipinski rule violations, medium total clearance and no mutagenic, tumorigenic, irritant and reproductive toxic risks for most of the compounds. Compounds VIIb, VIIi and VIIm emerged as the most potent anticancer agents among all compounds evaluated against 60 cancer cell lines through the one-dose (10 µM) sulforhodamine B assay. Further, the multiple dose cell viability studies against cancer cell lines MOLT-4, A549 and HCT-116 revealed results consistent with the one-dose assay, besides sparing normal cell line HEK-293. The three potent compounds also displayed potent LSD1 inhibitory activity with IC50 values of 2.25, 1.80 and 6.08 µM. The n-propyl-thio/isopropyl-thio group bonded to the pyrimidine ring and unsubstituted/ electron donating group (at the para- position) attached to the phenyl ring resulted in enhanced anticancer activity. However, against leukemia cancer, the electron donating isopropyl group remarkably enhanced anti-cancer activity. Our findings provide important leads, which merit further optimization to result in better cancer therapeutics.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Relation dose-effet des médicaments , Conception de médicament , Tests de criblage d'agents antitumoraux , Histone Demethylases , Pyrimidines , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Pyrimidines/composition chimique , Pyrimidines/pharmacologie , Pyrimidines/synthèse chimique , Relation structure-activité , Structure moléculaire , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Lignée cellulaire tumorale , Simulation de dynamique moléculaire , Simulation de docking moléculaire , Survie cellulaire/effets des médicaments et des substances chimiques
11.
Nat Commun ; 15(1): 3563, 2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38670969

RÉSUMÉ

Synthetic glucocorticoids (GC), such as dexamethasone, are extensively used to treat chronic inflammation and autoimmune disorders. However, long-term treatments are limited by various side effects, including muscle atrophy. GC activities are mediated by the glucocorticoid receptor (GR), that regulates target gene expression in various tissues in association with cell-specific co-regulators. Here we show that GR and the lysine-specific demethylase 1 (LSD1) interact in myofibers of male mice, and that LSD1 connects GR-bound enhancers with NRF1-associated promoters to stimulate target gene expression. In addition, we unravel that LSD1 demethylase activity is required for triggering starvation- and dexamethasone-induced skeletal muscle proteolysis in collaboration with GR. Importantly, inhibition of LSD1 circumvents muscle wasting induced by pharmacological levels of dexamethasone, without affecting their anti-inflammatory activities. Thus, our findings provide mechanistic insights into the muscle-specific GC activities, and highlight the therapeutic potential of targeting GR co-regulators to limit corticotherapy-induced side effects.


Sujet(s)
Dexaméthasone , Glucocorticoïdes , Histone Demethylases , Muscles squelettiques , Amyotrophie , Récepteurs aux glucocorticoïdes , Animaux , Mâle , Histone Demethylases/métabolisme , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/génétique , Glucocorticoïdes/pharmacologie , Dexaméthasone/pharmacologie , Récepteurs aux glucocorticoïdes/métabolisme , Souris , Amyotrophie/induit chimiquement , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/traitement médicamenteux , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/anatomopathologie , Souris de lignée C57BL , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
12.
Comput Biol Chem ; 110: 108072, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636391

RÉSUMÉ

The methylation and demethylation of lysine and arginine side chains are fundamental processes in gene regulation and disease development. Histone lysine methylation, controlled by histone lysine methyltransferases (KMTs) and histone lysine demethylases (KDMs), plays a vital role in maintaining cellular homeostasis and has been implicated in diseases such as cancer and aging. This study focuses on two members of the lysine demethylase (KDM) family, KDM4E and KDM6B, which are significant in gene regulation and disease pathogenesis. KDM4E demonstrates selectivity for gene regulation, particularly concerning cancer, while KDM6B is implicated in inflammation and cancer. The study utilizes specific inhibitors, DA-24905 and GSK-J1, showcasing their exceptional selectivity for KDM4E and KDM6B, respectively. Employing an array of computational simulations, including sequence alignment, molecular docking, dynamics simulations, and free energy calculations, we conclude that although the binding cavities of KDM4E and KDM6B has high similarity, there are still some different crucial amino acid residues, indicating diverse binding forms between protein and ligands. Various interaction predominates when proteins are bound to different ligands, which also has significant effect on selective inhibition. These findings provide insights into potential therapeutic strategies for diseases by selectively targeting these KDM members.


Sujet(s)
Antienzymes , Jumonji Domain-Containing Histone Demethylases , Jumonji Domain-Containing Histone Demethylases/antagonistes et inhibiteurs , Jumonji Domain-Containing Histone Demethylases/métabolisme , Jumonji Domain-Containing Histone Demethylases/composition chimique , Humains , Antienzymes/composition chimique , Antienzymes/pharmacologie , Simulation de dynamique moléculaire , Découverte de médicament , Simulation de docking moléculaire , Structure moléculaire , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Histone Demethylases/composition chimique , Relation structure-activité
13.
Prostate ; 84(10): 909-921, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38619005

RÉSUMÉ

INTRODUCTION: Lysine-specific demethylase 1 (LSD1) is emerging as a critical mediator of tumor progression in metastatic castration-resistant prostate cancer (mCRPC). Neuroendocrine prostate cancer (NEPC) is increasingly recognized as an adaptive mechanism of resistance in mCRPC patients failing androgen receptor axis-targeted therapies. Safe and effective LSD1 inhibitors are necessary to determine antitumor response in prostate cancer models. For this reason, we characterize the LSD1 inhibitor bomedemstat to assess its clinical potential in NEPC as well as other mCRPC pathological subtypes. METHODS: Bomedemstat was characterized via crystallization, flavine adenine dinucleotide spectrophotometry, and enzyme kinetics. On-target effects were assessed in relevant prostate cancer cell models by measuring proliferation and H3K4 methylation using western blot analysis. In vivo, pharmacokinetic (PK) and pharmacodynamic (PD) profiles of bomedemstat are also described. RESULTS: Structural, biochemical, and PK/PD properties of bomedemstat, an irreversible, orally-bioavailable inhibitor of LSD1 are reported. Our data demonstrate bomedemstat has >2500-fold greater specificity for LSD1 over monoamine oxidase (MAO)-A and -B. Bomedemstat also demonstrates activity against several models of advanced CRPC, including NEPC patient-derived xenografts. Significant intra-tumoral accumulation of orally-administered bomedemstat is measured with micromolar levels achieved in vivo (1.2 ± 0.45 µM at the 7.5 mg/kg dose and 3.76 ± 0.43 µM at the 15 mg/kg dose). Daily oral dosing of bomedemstat at 40 mg/kg/day is well-tolerated, with on-target thrombocytopenia observed that is rapidly reversible following treatment cessation. CONCLUSIONS: Bomedemstat provides enhanced specificity against LSD1, as revealed by structural and biochemical data. PK/PD data display an overall safety profile with manageable side effects resulting from LSD1 inhibition using bomedemstat in preclinical models. Altogether, our results support clinical testing of bomedemstat in the setting of mCRPC.


Sujet(s)
Histone Demethylases , Tumeurs prostatiques résistantes à la castration , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Mâle , Humains , Animaux , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Souris , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/pharmacologie , Antienzymes/pharmacologie , Antienzymes/pharmacocinétique , Benzamides , Pipérazines , Triazoles
14.
Bioorg Med Chem ; 101: 117651, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38401457

RÉSUMÉ

Lysine-specific demethylase 1 (LSD1) is a histone lysine demethylase that is significantly overexpressed or dysregulated in different cancers and plays important roles in cell growth, invasion, migration, immune escape, angiogenesis, gene regulation, and transcription. Therefore, it is a superb target for the discovery of novel antitumor agents. However, because of their innate and acquired resistance and low selectivity, LSD1 inhibitors are associated with limited therapeutic efficacy and high toxicity. Furthermore, LSD1 inhibitors synergistically improve the efficacy of additional antitumor drugs, which encourages numerous medicinal chemists to innovate and develop new-generation LSD1-based dual-target agents. This review discusses the theoretical foundation of the design of LSD1-based dual-target agents and summarizes their possible applications in treating cancers.


Sujet(s)
Antinéoplasiques , Histone Demethylases , Tumeurs , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Antienzymes/pharmacologie , Antienzymes/usage thérapeutique , Régulation de l'expression des gènes , Histone Demethylases/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie
15.
Cancer Metastasis Rev ; 43(2): 795-821, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38227150

RÉSUMÉ

Modulation of histone methylation status is regarded as an important mechanism of epigenetic regulation and has substantial clinical potential for the therapy of diseases, including cancer and other disorders. The present study aimed to provide a comprehensive introduction to the enzymology of histone demethylases, as well as their cancerous roles, molecular mechanisms, therapeutic possibilities, and challenges for targeting them, in order to advance drug design for clinical therapy and highlight new insight into the mechanisms of these enzymes in cancer. A series of clinical trials have been performed to explore potential roles of histone demethylases in several cancer types. Numerous targeted inhibitors associated with immunotherapy, chemotherapy, radiotherapy, and targeted therapy have been used to exert anticancer functions. Future studies should evaluate the dynamic transformation of histone demethylases leading to carcinogenesis and explore individual therapy.


Sujet(s)
Histone Demethylases , Tumeurs , Humains , Tumeurs/génétique , Tumeurs/enzymologie , Tumeurs/anatomopathologie , Tumeurs/traitement médicamenteux , Histone Demethylases/métabolisme , Histone Demethylases/antagonistes et inhibiteurs , Animaux , Épigenèse génétique , Histone/métabolisme , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie
16.
J Cell Physiol ; 239(4): e31178, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38214211

RÉSUMÉ

Glioblastoma stem cells (GSCs) exert a crucial influence on glioblastoma (GBM) development, progression, resistance to therapy, and recurrence, making them an attractive target for drug discovery. UTX, a histone H3K27 demethylase, participates in regulating multiple cancer types. However, its functional role in GSCs remains insufficiently explored. This study aims to investigate the role and regulatory mechanism of UTX on GSCs. Analysis of TCGA data revealed heightened UTX expression in glioma, inversely correlating with overall survival. Inhibiting UTX suppressed GBM cell growth and induced apoptosis. Subsequently, we cultured primary GSCs from three patients, observing that UTX inhibition suppressed cell proliferation and induced apoptosis. RNA-seq was performed to analyze the gene expression changes after silencing UTX in GSCs. The results indicated that UTX-mediated genes were strongly correlated with GBM progression and regulatory tumor microenvironment. The transwell co-cultured experiment showed that silencing UTX in the transwell chamber GSCs inhibited the well plate cell proliferation. Protein-protein interaction analysis revealed that periostin (POSTN) played a role in the UTX-mediated transcriptional regulatory network. Replenishing POSTN reversed the effects of UTX inhibition on GSC proliferation and apoptosis. Our study demonstrated that UTX inhibition hindered POSTN expression by enhancing the H3K27me2/3 level, eventually resulting in inhibiting proliferation and promoting apoptosis of patient-derived GSCs. Our findings may provide a novel and effective strategy for the treatment of GBM.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Histone Demethylases , Cellules souches tumorales , Humains , Apoptose/génétique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Glioblastome/traitement médicamenteux , Glioblastome/génétique , Glioblastome/anatomopathologie , Cellules souches tumorales/anatomopathologie , , Microenvironnement tumoral , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme
17.
PLoS One ; 18(12): e0289860, 2023.
Article de Anglais | MEDLINE | ID: mdl-38134183

RÉSUMÉ

Elevated levels of Fetal Hemoglobin interfere with polymerization of sickle hemoglobin thereby reducing anemia, lessening the severity of symptoms, and increasing life span of patients with sickle cell disease. An affordable, small molecule drug that stimulates HbF expression in vivo would be ideally suited to treat the large numbers of SCD patients that exist worldwide. Our previous work showed that administration of the LSD1 (KDM1A) inhibitor RN-1 to normal baboons increased Fetal Hemoglobin (HbF) and was tolerated over a prolonged treatment period. HbF elevations were associated with changes in epigenetic modifications that included increased levels of H3K4 di-and tri-methyl lysine at the γ-globin promoter. While dramatic effects of the loss of LSD1 on hematopoietic differentiation have been observed in murine LSD1 gene deletion and silencing models, the effect of pharmacological inhibition of LSD1 in vivo on hematopoietic differentiation is unknown. The goal of these experiments was to investigate the in vivo mechanism of action of the LSD1 inhibitor RN-1 by determining its effect on γ-globin expression in highly purified subpopulations of bone marrow erythroid cells enriched for varying stages of erythroid differentiation isolated directly from baboons treated with RN-1 and also by investigating the effect of RN1 on the global transcriptome in a highly purified population of proerythroblasts. Our results show that RN-1 administered to baboons targets an early event during erythroid differentiation responsible for γ-globin repression and increases the expression of a limited number of genes including genes involved in erythroid differentiation such as GATA2, GFi-1B, and LYN.


Sujet(s)
Drépanocytose , Histone Demethylases , Animaux , Humains , Souris , Drépanocytose/génétique , Hémoglobine foetale/génétique , Globines gamma/génétique , Expression des gènes , Histone Demethylases/antagonistes et inhibiteurs , Papio anubis/génétique
18.
J Med Chem ; 66(7): 4275-4293, 2023 04 13.
Article de Anglais | MEDLINE | ID: mdl-37014989

RÉSUMÉ

Chemotherapy, targeted therapy, and immunotherapy are effective against most tumors, but drug resistance remains a barrier to successful treatment. Lysine-specific demethylase 1 (LSD1), a member of histone demethylation modifications, can regulate invasion, metastasis, apoptosis, and immune escape of tumor cells, which are associated with tumorigenesis and tumor progression. Recent studies suggest that LSD1 ablation regulates resensitivity of tumor cells to anticarcinogens containing immune checkpoint inhibitors (ICIs) via multiple upstream and downstream pathways. In this review, we describe the recent findings about LSD1 biology and its role in the development and progression of cancer drug resistance. Further, we summarize LSD1 inhibitors that have a reversal or resensitive effect on drug resistance and discuss the possibility of targeting LSD1 in combination with other agents to surmount resistance.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Histone Demethylases , Humains , Résistance aux substances , Résistance aux médicaments antinéoplasiques/génétique , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Immunothérapie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme
19.
Biochem Biophys Res Commun ; 647: 23-29, 2023 03 05.
Article de Anglais | MEDLINE | ID: mdl-36709669

RÉSUMÉ

The epigenetic regulation for gene expression determines cell plasticity. Oral squamous cell carcinoma (SCC) exhibits bidirectional cell plasticity, i.e. epithelial differentiation and epithelial to mesenchymal transition (EMT). The epigenetic regulator LSD1 is a histone H3-specific demethylase to which chemical inhibitors for its activity had been developed as an anti-cancer therapeutics. The bidirectional plasticity of the oral SCC cell line OM-1 had been characterized, but it remained unclear how chemical LSD1 inhibitors affect cell plasticity. Here we reported an adverse effect against cancer therapeutics, which was EMT induction in vitro by the chemical LSD1 inhibitor. The LSD1 inhibitor caused EMT-TF ZEB1 in OM-1 to undergo EMT. Furthermore, an additional EMT-TF Snail-dependent partial EMT phenotype in OM-1 progressed to complete EMT in conjunction with LSD1 inhibitor-dependent ZEB1 induction. The promotor activity of ZEB1 was up-regulated under LSD1 inhibition. The regulatory chromatin regions of ZEB1 accumulated histone H3 methylation under the chemical inhibition of LSD1. The LSD1 inhibitor also upregulates epithelial gene expression in vitro; however, the bidirectional effect of LSD1 inhibitor should be considered in cancer therapeutics.


Sujet(s)
Histone Demethylases , Tumeurs de la bouche , Carcinome épidermoïde de la tête et du cou , Humains , Lignée cellulaire tumorale , Épigenèse génétique , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Histone/métabolisme , Tumeurs de la bouche/traitement médicamenteux , Tumeurs de la bouche/génétique , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/génétique , Facteur de transcription Zeb1/génétique , Facteur de transcription Zeb1/métabolisme
20.
Eur J Med Chem ; 244: 114818, 2022 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-36223680

RÉSUMÉ

The epigenetic regulator lysine specific demethylase 1 (LSD1), a MYCN cofactor, cooperatively silences MYCN suppressor genes. Furthermore, LSD1 has been correlated with adverse effects in neuroblastic tumors by propagating an undifferentiated, malignant phenotype. We observed that high LSD1 mRNA expression in MYCN-expressing neuroblastoma (NB) correlated with poor prognosis, implicating LSD1 as an oncogenic accomplice in high-grade NB. Thus, LSD1 inhibition is a potential strategy for targeting treatment-resistant MYCN-expressing NB. Tranylcypromine-based covalent LSD1 inhibitors have demonstrated anti-tumor activity but are associated with undesirable off-target effects, such that only 2 non-covalent LSD1 inhibitors are in clinical trials. We now report 3 novel scaffolds for reversible LSD1 inhibition: 2-(arylsulfonamido)benzoic acid, N-(2-(1H-tetrazol-5-yl)phenyl)benzenesulfonamide and 2-(arylcarboxamido)benzoic acid analogues. The most active of these analogues, compound 48, exhibited potent and selective mixed reversible inhibition of LSD1 (IC50 = 0.58 µM) and significantly increased global H3K4me2 in NB cells. In addition, combination treatment with 48 and bortezomib in NB cells results in a synergistic effect.


Sujet(s)
Histone Demethylases , Neuroblastome , Humains , Lignée cellulaire tumorale , Histone Demethylases/antagonistes et inhibiteurs , Protéine du proto-oncogène N-Myc/génétique , Neuroblastome/traitement médicamenteux , Neuroblastome/métabolisme , Benzoates/pharmacologie , Benzoates/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...