Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.991
Filtrer
1.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(3): 679-684, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-38926953

RÉSUMÉ

OBJECTIVE: To investigate the efficacy of decitabine combined with preexcitation regimen in the treatment of newly diagnosed acute myeloid leukemia (AML) patients who have not been relieved by the first standard induction chemotherapy and its influence on the relative content of regulatory T lymphocytes (Tregs). METHODS: The clinical data of 102 newly diagnosed AML patients (except acute promyelocytic leukemia) who did not relieve after initial standard induction chemotherapy in Shaanxi Provincial People's Hospital from March 2013 to March 2019 were retrospectively analyzed. Fifty-one patients who accepted pre-excitation regimen were divided into regular group, while another 51 patients treated with decitabine combined with pre-excitation regimen were divided into combination group. The efficacy, incidence of toxic and side effects, Core Scale of Quality of Life (QLQ-C30) score before and after treatment, T lymphocyte subsets (CD3+, CD4+, CD4+/CD8+, Tregs) and 3-year overall survival (OS) rate were compared between the two groups. RESULTS: The total effective rate of combination group was 80.39%, which was significantly higher than 62.75% of regular group (P < 0.05). After treatment, the QLQ-C30 score of combination group was 60.27±6.96, which was significantly lower than 65.73±7.96 of regular group (P < 0.001). There was no statistical difference in the incidence of toxic and side effects between the two groups (P >0.05). After treatment, the levels of CD3+, CD4+, CD4+/CD8+ in the combination group were higher than those in the regular group (all P < 0.001), while Treg was lower (P < 0.001). The 3-year OS rate in the combination group was 72.55%, which was significantly higher than 52.94% in the regular group (P < 0.001). CONCLUSION: Decitabine combined with preexcitation regimen has a significant effect on AML patients who have not been alleviated by standard induction chemotherapy in the first course of treatment. It can reduce anti-tumor immune suppression and improve immune function by regulating the relative content of Tregs, thus prolongs survival time and improves life quality of patients without increasing adverse reactions.


Sujet(s)
Décitabine , Chimiothérapie d'induction , Leucémie aigüe myéloïde , Humains , Décitabine/administration et posologie , Études rétrospectives , Leucémie aigüe myéloïde/traitement médicamenteux , Lymphocytes T régulateurs , Qualité de vie , Mâle , Femelle , Résultat thérapeutique , Taux de survie
2.
Oncotarget ; 15: 361-373, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38829622

RÉSUMÉ

Histone deacetylase inhibitors (HDACi) can modulate the acetylation status of proteins, influencing the genomic instability exhibited by cancer cells. Poly (ADP ribose) polymerase (PARP) inhibitors (PARPi) have a direct effect on protein poly (ADP-ribosyl)ation, which is important for DNA repair. Decitabine is a nucleoside cytidine analogue, which when phosphorylated gets incorporated into the growing DNA strand, inhibiting methylation and inducing DNA damage by inactivating and trapping DNA methyltransferase on the DNA, thereby activating transcriptionally silenced DNA loci. We explored various combinations of HDACi and PARPi +/- decitabine (hypomethylating agent) in pancreatic cancer cell lines BxPC-3 and PL45 (wild-type BRCA1 and BRCA2) and Capan-1 (mutated BRCA2). The combination of HDACi (panobinostat or vorinostat) with PARPi (talazoparib or olaparib) resulted in synergistic cytotoxicity in all cell lines tested. The addition of decitabine further increased the synergistic cytotoxicity noted with HDACi and PARPi, triggering apoptosis (evidenced by increased cleavage of caspase 3 and PARP1). The 3-drug combination treatments (vorinostat, talazoparib, and decitabine; vorinostat, olaparib, and decitabine; panobinostat, talazoparib, and decitabine; panobinostat, olaparib, and decitabine) induced more DNA damage (increased phosphorylation of histone 2AX) than the individual drugs and impaired the DNA repair pathways (decreased levels of ATM, BRCA1, and ATRX proteins). The 3-drug combinations also altered the epigenetic regulation of gene expression (NuRD complex subunits, reduced levels). This is the first study to demonstrate synergistic interactions between the aforementioned agents in pancreatic cancer cell lines and provides preclinical data to design individualized therapeutic approaches with the potential to improve pancreatic cancer treatment outcomes.


Sujet(s)
Azacitidine , Décitabine , Synergie des médicaments , Inhibiteurs de désacétylase d'histone , Tumeurs du pancréas , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Décitabine/pharmacologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Lignée cellulaire tumorale , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Azacitidine/pharmacologie , Azacitidine/analogues et dérivés , Apoptose/effets des médicaments et des substances chimiques , Protocoles de polychimiothérapie antinéoplasique/pharmacologie
3.
Cell Rep Med ; 5(6): 101592, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38843841

RÉSUMÉ

Environmental lipids are essential for fueling tumor energetics, but whether these exogenous lipids transported into cancer cells facilitate immune escape remains unclear. Here, we find that CD36, a transporter for exogenous lipids, promotes acute myeloid leukemia (AML) immune evasion. We show that, separately from its established role in lipid oxidation, CD36 on AML cells senses oxidized low-density lipoprotein (OxLDL) to prime the TLR4-LYN-MYD88-nuclear factor κB (NF-κB) pathway, and exogenous palmitate transfer via CD36 further potentiates this innate immune pathway by supporting ZDHHC6-mediated MYD88 palmitoylation. Subsequently, NF-κB drives the expression of immunosuppressive genes that inhibit anti-tumor T cell responses. Notably, high-fat-diet or hypomethylating agent decitabine treatment boosts the immunosuppressive potential of AML cells by hijacking CD36-dependent innate immune signaling, leading to a dampened therapeutic effect. This work is of translational interest because lipid restriction by US Food and Drug Administration (FDA)-approved lipid-lowering statin drugs improves the efficacy of decitabine therapy by weakening leukemic CD36-mediated immunosuppression.


Sujet(s)
Antigènes CD36 , Décitabine , Leucémie aigüe myéloïde , Métabolisme lipidique , Lipoprotéines LDL , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/immunologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Décitabine/pharmacologie , Décitabine/usage thérapeutique , Lipoprotéines LDL/métabolisme , Animaux , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire tumorale , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/métabolisme , Acyltransferases/génétique , Immunité innée/effets des médicaments et des substances chimiques , Souris de lignée C57BL
4.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38906675

RÉSUMÉ

Decitabine and azacytidine are considered as epigenetic drugs that induce DNA methyltransferase (DNMT)-DNA crosslinks, resulting in DNA hypomethylation and damage. Although they are already applied against myeloid cancers, important aspects of their mode of action remain unknown, highly limiting their clinical potential. Using a combinatorial approach, we reveal that the efficacy profile of both compounds primarily depends on the level of induced DNA damage. Under low DNMT activity, only decitabine has a substantial impact. Conversely, when DNMT activity is high, toxicity and cellular response to both compounds are dramatically increased, but do not primarily depend on DNA hypomethylation or RNA-associated processes. By investigating proteome dynamics on chromatin, we show that decitabine induces a strictly DNMT-dependent multifaceted DNA damage response based on chromatin recruitment, but not expression-level changes of repair-associated proteins. The choice of DNA repair pathway hereby depends on the severity of decitabine-induced DNA lesions. Although under moderate DNMT activity, mismatch (MMR), base excision (BER), and Fanconi anaemia-dependent DNA repair combined with homologous recombination are activated in response to decitabine, high DNMT activity and therefore immense replication stress induce activation of MMR and BER followed by non-homologous end joining.


Sujet(s)
Azacitidine , Altération de l'ADN , Méthylation de l'ADN , Réparation de l'ADN , Décitabine , Décitabine/pharmacologie , Altération de l'ADN/effets des médicaments et des substances chimiques , Humains , Réparation de l'ADN/effets des médicaments et des substances chimiques , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Azacitidine/analogues et dérivés , Azacitidine/pharmacologie , Antimétabolites antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , DNA (cytosine-5-)-methyltransferase/métabolisme , Chromatine/métabolisme , Chromatine/effets des médicaments et des substances chimiques , DNA modification methylases/métabolisme
5.
Viruses ; 16(5)2024 05 08.
Article de Anglais | MEDLINE | ID: mdl-38793627

RÉSUMÉ

Equid herpesvirus 4 (EHV-4) is a common respiratory pathogen in horses. It sporadically induces abortion or neonatal death. Although its contribution in neurological disorders is not clearly demonstrated, there is a strong suspicion of its involvement. Despite preventive treatments using vaccines against EHV-1/EHV-4, the resurgence of alpha-EHV infection still constitutes an important threat to the horse industry. Yet very few studies have been conducted on the search for antiviral molecules against EHV-4. A screening of 42 antiviral compounds was performed in vitro on equine fibroblast cells infected with the EHV-4 405/76 reference strain (VR2230). The formation of cytopathic effects was monitored by real-time cell analysis (RTCA), and the viral load was quantified by quantitative PCR. Aciclovir, the most widely used antiviral against alpha-herpesviruses in vivo, does not appear to be effective against EHV-4 in vitro. Potential antiviral activities were confirmed for eight molecules (idoxuridine, vidarabine, pritelivir, cidofovir, valganciclovir, ganciclovir, aphidicolin, and decitabine). Decitabine demonstrates the highest efficacy against EHV-4 in vitro. Transcriptomic analysis revealed the up-regulation of various genes implicated in interferon (IFN) response, suggesting that decitabine triggers the immune antiviral pathway.


Sujet(s)
Antiviraux , Décitabine , Herpèsvirus équin de type 4 , Immunité innée , Animaux , Antiviraux/pharmacologie , Equus caballus , Décitabine/pharmacologie , Immunité innée/effets des médicaments et des substances chimiques , Herpèsvirus équin de type 4/effets des médicaments et des substances chimiques , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/virologie , Infections à Herpesviridae/traitement médicamenteux , Infections à Herpesviridae/virologie , Infections à Herpesviridae/médecine vétérinaire , Infections à Herpesviridae/immunologie , Maladies des chevaux/virologie , Maladies des chevaux/traitement médicamenteux , Maladies des chevaux/immunologie , Charge virale/effets des médicaments et des substances chimiques , Lignée cellulaire , Réplication virale/effets des médicaments et des substances chimiques , Évaluation préclinique de médicament
6.
Med Oncol ; 41(7): 165, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38819590

RÉSUMÉ

Myeloid-derived suppressor cells (MDSCs) play a crucial role in promoting melanoma metastasis. Reprogramming MDSCs into mature M1 macrophages has emerged as a strategy to inhibit metastasis. Decitabine (Dec) is known to eradicate MDSCs and suppress tumor growth. In this study, we provide evidence that Dec not only reduces the MDSC population by inducing apoptosis, arresting cell cycle, and impairing recruitment, but also suppresses their immunosuppressive function by downregulating related genes and facilitating differentiation into M1 macrophages. Transcriptomic analysis of Dec-treated MDSCs revealed a marked downregulation of immunosuppressive genes including S100a9, S100a8, Vegf, Cxcr2, and Nos2. Meanwhile, M1 macrophage-associated genes involved in immune activation were upregulated, such as Ddx58, Isg15, Tap1, Ccl5, Cxcl9, and Cxcl10. Further bioinformatic analysis indicated that Dec promotes MDSC-to-M1 macrophage differentiation and activates innate immune pathways including NOD-like signaling to enhance anti-tumor immunity. Time-course studies implied that Dec upregulates myeloid transcription factor Irf7 to initiate MDSC differentiation and orchestrate the anti-tumor immune response. Collectively, our study unveils a novel dual-functional mechanism of Dec as both a cytotoxic agent reducing MDSCs and an inducer of their differentiation into M1 macrophages, thereby alleviating immunosuppression. This highlights Dec's potential for clinical melanoma metastasis suppression.


Sujet(s)
Décitabine , Mélanome , Cellules myéloïdes suppressives , Cellules myéloïdes suppressives/effets des médicaments et des substances chimiques , Cellules myéloïdes suppressives/immunologie , Cellules myéloïdes suppressives/métabolisme , Animaux , Décitabine/pharmacologie , Souris , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Mélanome/immunologie , Humains , Souris de lignée C57BL , Différenciation cellulaire/effets des médicaments et des substances chimiques , Antimétabolites antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Tolérance immunitaire/effets des médicaments et des substances chimiques
7.
Eur J Med Chem ; 272: 116447, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38714044

RÉSUMÉ

Histone deacetylase 6 (HDAC6) is an emerging drug target to treat oncological and non-oncological conditions. Since highly selective HDAC6 inhibitors display limited anticancer activity when used as single agent, they usually require combination therapies with other chemotherapeutics. In this work, we synthesized a mini library of analogues of the preferential HDAC6 inhibitor HPOB in only two steps via an Ugi four-component reaction as the key step. Biochemical HDAC inhibition and cell viability assays led to the identification of 1g (highest antileukemic activity) and 2b (highest HDAC6 inhibition) as hit compounds. In subsequent combination screens, both 1g and especially 2b showed synergy with DNA methyltransferase inhibitor decitabine in acute myeloid leukemia (AML). Our findings highlight the potential of combining HDAC6 inhibitors with DNA methyltransferase inhibitors as a strategy to improve AML treatment outcomes.


Sujet(s)
Antinéoplasiques , Décitabine , Tests de criblage d'agents antitumoraux , Synergie des médicaments , Histone deacetylase 6 , Inhibiteurs de désacétylase d'histone , Leucémie aigüe myéloïde , Humains , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/synthèse chimique , Histone deacetylase 6/antagonistes et inhibiteurs , Histone deacetylase 6/métabolisme , Décitabine/pharmacologie , Décitabine/composition chimique , Relation structure-activité , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/métabolisme , Structure moléculaire , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Lignée cellulaire tumorale , Peptoïdes/composition chimique , Peptoïdes/pharmacologie , Peptoïdes/synthèse chimique , Aminopyridines , Benzamides
8.
Sci Rep ; 14(1): 11595, 2024 05 21.
Article de Anglais | MEDLINE | ID: mdl-38773164

RÉSUMÉ

Despite growing evidence implicating the calcium-activated chloride channel anoctamin1 (ANO1) in cancer metastasis, its direct impact on the metastatic potential of prostate cancer and the possible significance of epigenetic alteration in this process are not fully understood. Here, we show that ANO1 is minimally expressed in LNCap and DU145 prostate cancer cell lines with low metastatic potential but overexpressed in high metastatic PC3 prostate cancer cell line. The treatment of LNCap and DU145 cells with DNMT inhibitor 5-aza-2'-deoxycytidine (5-Aza-CdR) potentiates ANO1 expression, suggesting that DNA methylation is one of the mechanisms controlling ANO1 expression. Consistent with this notion, hypermethylation was detected at the CpG island of ANO1 promoter region in LNCap and DU145 cells, and 5-Aza-CdR treatment resulted in a drastic demethylation at promoter CpG methylation sites. Upon 5-Aza-CdR treatment, metastatic indexes, such as cell motility, invasion, and metastasis-related gene expression, were significantly altered in LNCap and DU145 cells. These 5-Aza-CdR-induced metastatic hallmarks were, however, almost completely ablated by stable knockdown of ANO1. These in vitro discoveries were further supported by our in vivo observation that ANO1 expression in xenograft mouse models enhances the metastatic dissemination of prostate cancer cells into tibial bone and the development of osteolytic lesions. Collectively, our results help elucidate the critical role of ANO1 expression in prostate cancer bone metastases, which is epigenetically modulated by promoter CpG methylation.


Sujet(s)
Anoctamine-1 , Tumeurs osseuses , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , Protéines tumorales , Régions promotrices (génétique) , Tumeurs de la prostate , Mâle , Anoctamine-1/métabolisme , Anoctamine-1/génétique , Humains , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Animaux , Lignée cellulaire tumorale , Tumeurs osseuses/secondaire , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Souris , Ilots CpG , Décitabine/pharmacologie , Mouvement cellulaire/génétique , Épigenèse génétique , Azacitidine/pharmacologie
9.
Methods Cell Biol ; 186: 131-150, 2024.
Article de Anglais | MEDLINE | ID: mdl-38705597

RÉSUMÉ

Hypomethylating therapies using decitabine or azacitidine are actively investigated to treat acute myeloid leukemia, myelodysplastic syndromes, as maintenance therapy after allogenic stem cell transplant and hemoglobinopathies. The therapeutic mechanism is to de-repress genes that have been turned off through oncogenesis or development via methylation. The therapy can be non-cytotoxic at low dosage, sparing healthy stem cells and operating on committed precursors. Because the methods of determining maximum tolerated dose are not well suited to this paradigm, and because the mechanism of action, which is depletion of DNA methylase 1 (DNMT1), is complex and dependent on passing through a cell cycle, a pharmacodynamic assay that measures DNMT1 can inform clinical trials aimed at establishing and improving therapy. Herein, we provide an assay that measures DNMT1 relative levels in circulating T cells of peripheral blood.


Sujet(s)
Azacitidine , DNA (Cytosine-5-)-methyltransferase 1 , Méthylation de l'ADN , Décitabine , Azacitidine/pharmacologie , Humains , Décitabine/pharmacologie , Méthylation de l'ADN/effets des médicaments et des substances chimiques , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/génétique , Antimétabolites antinéoplasiques/pharmacologie , Antimétabolites antinéoplasiques/usage thérapeutique , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , DNA (cytosine-5-)-methyltransferase/métabolisme , DNA (cytosine-5-)-methyltransferase/génétique , Syndromes myélodysplasiques/traitement médicamenteux , Syndromes myélodysplasiques/métabolisme
10.
Anticancer Res ; 44(6): 2459-2470, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38821592

RÉSUMÉ

BACKGROUND/AIM: Gastric cancer, with its high global incidence and mortality rates, poses a significant challenge due to the rapid decline in patient survival upon metastasis. Understanding and combating metastasis are crucial in improving outcomes. The metastasis suppressor gene CD82 has demonstrated efficacy in inhibiting metastasis across various carcinomas but is frequently down-regulated. However, its role and regulatory mechanisms in gastric cancer remain elusive. MATERIALS AND METHODS: Utilizing public data, we assessed patient survival in relation to CD82 expression. CD82 expression in gastric cancer cell lines was evaluated via western blotting, and its impact on cell mobility was assessed through wound healing and Transwell assays. The demethylation of CD82 was induced using 5-aza-deoxycytidine, while methylation levels were detected via methylation-specific PCR. RESULTS: Low CD82 expression correlated with poor prognosis in patients, and down-regulation and over-expression of CD82 significantly affected cell mobility. Treatment with 5-aza-deoxycytidine restored CD82 expression in low-expressing cell lines, highlighting its methylation-dependent regulation. CONCLUSION: CD82 serves as a pivotal regulator of cell mobility in gastric cancer by suppressing metastasis. Its expression is attenuated in gastric cancer cells through promoter hypermethylation.


Sujet(s)
Mouvement cellulaire , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , Antigène CD82 , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Antigène CD82/génétique , Antigène CD82/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Régions promotrices (génétique) , Pronostic , Décitabine/pharmacologie , Métastase tumorale , Régulation négative , Gènes suppresseurs de tumeur
11.
Cancer Lett ; 593: 216949, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38729558

RÉSUMÉ

Hypomethylating agents (HMAs) are widely employed in the treatment of myeloid malignancies. However, unresponsive or resistant to HMAs occurs in approximately 50 % of patients. ASXL1, one of the most commonly mutated genes across the full spectrum of myeloid malignancies, has been reported to predict a lower overall response rate to HMAs, suggesting an essential need to develop effective therapeutic strategies for the patients with HMA failure. Here, we investigated the impact of ASXL1 on cellular responsiveness to decitabine treatment. ASXL1 deficiency increased resistance to decitabine treatment in AML cell lines and mouse bone marrow cells. Transcriptome sequencing revealed significant alterations in genes regulating cell cycle, apoptosis, and histone modification in ASXL1 deficient cells that resistant to decitabine. BIRC5 was identified as a potential target for overcoming decitabine resistance in ASXL1 deficient cells. Furthermore, our experimental evidence demonstrated that the small-molecule inhibitor of BIRC5 (YM-155) synergistically sensitized ASXL1 deficient cells to decitabine treatment. This study sheds light on the molecular mechanisms underlying the ASXL1-associated HMA resistance and proposes a promising therapeutic strategy for improving treatment outcomes in affected individuals.


Sujet(s)
Décitabine , Résistance aux médicaments antinéoplasiques , Leucémie aigüe myéloïde , Protéines de répression , Survivine , Animaux , Décitabine/pharmacologie , Humains , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Survivine/génétique , Survivine/métabolisme , Protéines de répression/génétique , Protéines de répression/métabolisme , Souris , Lignée cellulaire tumorale , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Antimétabolites antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Imidazoles , Naphtoquinones
12.
EMBO J ; 43(12): 2397-2423, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38760575

RÉSUMÉ

The nucleoside analogue decitabine (or 5-aza-dC) is used to treat several haematological cancers. Upon its triphosphorylation and incorporation into DNA, 5-aza-dC induces covalent DNA methyltransferase 1 DNA-protein crosslinks (DNMT1-DPCs), leading to DNA hypomethylation. However, 5-aza-dC's clinical outcomes vary, and relapse is common. Using genome-scale CRISPR/Cas9 screens, we map factors determining 5-aza-dC sensitivity. Unexpectedly, we find that loss of the dCMP deaminase DCTD causes 5-aza-dC resistance, suggesting that 5-aza-dUMP generation is cytotoxic. Combining results from a subsequent genetic screen in DCTD-deficient cells with the identification of the DNMT1-DPC-proximal proteome, we uncover the ubiquitin and SUMO1 E3 ligase, TOPORS, as a new DPC repair factor. TOPORS is recruited to SUMOylated DNMT1-DPCs and promotes their degradation. Our study suggests that 5-aza-dC-induced DPCs cause cytotoxicity when DPC repair is compromised, while cytotoxicity in wild-type cells arises from perturbed nucleotide metabolism, potentially laying the foundations for future identification of predictive biomarkers for decitabine treatment.


Sujet(s)
DNA (Cytosine-5-)-methyltransferase 1 , Décitabine , Ubiquitin-protein ligases , Décitabine/pharmacologie , Humains , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Antimétabolites antinéoplasiques/pharmacologie , Animaux , Sumoylation/effets des médicaments et des substances chimiques
13.
Cancer Res Commun ; 4(6): 1441-1453, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38727208

RÉSUMÉ

Programmed cell death mechanisms are important for the regulation of tumor development and progression. Evasion of and resistance to apoptosis are significant factors in tumorigenesis and drug resistance. Bypassing apoptotic pathways and eliciting another form of regulated cell death, namely necroptosis, an immunogenic cell death (ICD), may override apoptotic resistance. Here, we present the mechanistic rationale for combining tolinapant, an antagonist of the inhibitor of apoptosis proteins (IAP), with decitabine, a hypomethylating agent (HMA), in T-cell lymphoma (TCL). Tolinapant treatment alone of TCL cells in vitro and in syngeneic in vivo models demonstrated that ICD markers can be upregulated, and we have shown that epigenetic priming with decitabine further enhances this effect. The clinical relevance of ICD markers was confirmed by the direct measurement of plasma proteins from patients with peripheral TCL treated with tolinapant. We showed increased levels of necroptosis in TCL lines, along with the expression of cancer-specific antigens (such as cancer testis antigens) and increases in genes involved in IFN signaling induced by HMA treatment, together deliver a strong adaptive immune response to the tumor. These results highlight the potential of a decitabine and tolinapant combination for TCL and could lead to clinical evaluation. SIGNIFICANCE: The IAP antagonist tolinapant can induce necroptosis, a key immune-activating event, in TCL. Combination with DNA hypomethylation enhances tolinapant sensitivity and primes resistant cells by re-expressing necrosome proteins. In addition, this combination leads to increases in genes involved in IFN signaling and neoantigen expression, providing further molecular rationale for this novel therapeutic option.


Sujet(s)
Méthylation de l'ADN , Décitabine , Épigenèse génétique , Lymphome T , Humains , Épigenèse génétique/effets des médicaments et des substances chimiques , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Animaux , Décitabine/pharmacologie , Décitabine/usage thérapeutique , Souris , Lymphome T/traitement médicamenteux , Lymphome T/immunologie , Lymphome T/génétique , Lymphome T/anatomopathologie , Lignée cellulaire tumorale , Nécroptose/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
14.
Clin Epigenetics ; 16(1): 63, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38725010

RÉSUMÉ

BACKGROUND: Decitabine (DAC), a DNA methyltransferase inhibitor, has shown efficacy combined with chemotherapy for relapsed or refractory (R/R) acute myeloid leukemia (AML) in adults, but less is known about its efficacy in children. Accordingly, we conducted a study which involved a priming regimen consisting of DAC with cladribine, cytarabine, and granulocyte-stimulating factor (DAC-CLAG) and compared the efficacy and safety of this regimen with CLAG alone. METHODS: A total of 39 R/R AML children who received the CLAG or DAC-CLAG regimen in Shanghai Children's Hospital were retrospectively enrolled in this non-randomized study. These regimens were studied sequentially over time. Twenty-two patients received CLAG from 2015, while 17 patients were administered epigenetic priming with DAC before CLAG from 2020. Patients were subsequently bridged to stem cell transplantation (SCT) or consolidation chemotherapy. Complete remission (CR) and adverse effects were analyzed by Fisher's exact test, and survival was analyzed by the Kaplan-Meier method. RESULTS: DAC-CLAG conferred a numerically higher CR compared to CLAG (70.59% vs 63.64%; P = 0.740). High CR rates occurred in patients with good cytogenetics (P = 0.029) and prior induction without cladribine (P = 0.099). The 1-year event-free survival (EFS) was 64.71% ± 11.59% and 63.31% ± 10.35% in the DAC-CLAG and CLAG group (P = 0.595), and 1-year overall survival (OS) was 81.45% ± 9.72% and 77.01% ± 9.04%, respectively (P = 0.265). The 1-year OS and EFS after SCT were higher in the DAC-CLAG than in the CLAG cohort (100% vs 92.31% ± 7.39%, P = 0.072; 92.31% ± 7.39% vs 85.71% ± 9.35%, P = 0.158). Univariate analysis revealed that a good prognosis included good cytogenetics (P = 0.002), non-complex karyotype (P = 0.056), CR on reinduction (P < 0.0001), and bridging to SCT (P = 0.0007). Use of a hypomethylating agent (P = 0.049) and bridging to SCT (P = 0.011) were independent prognostic factors. Grade 3/4 hematologic toxicity and infection were the main adverse events. CONCLUSIONS: DAC prior to the CLAG regimen improved remission in pediatric R/R AML, and was feasible and well tolerated. CLAG ± DAC as a salvage therapy prior to SCT induced improved survival.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Chloro-2 désoxyadénosine , Cytarabine , Décitabine , Épigenèse génétique , Leucémie aigüe myéloïde , Humains , Décitabine/usage thérapeutique , Décitabine/administration et posologie , Décitabine/pharmacologie , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Mâle , Femelle , Enfant , Enfant d'âge préscolaire , Chloro-2 désoxyadénosine/usage thérapeutique , Chloro-2 désoxyadénosine/administration et posologie , Études rétrospectives , Cytarabine/usage thérapeutique , Cytarabine/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adolescent , Épigenèse génétique/effets des médicaments et des substances chimiques , Facteur de stimulation des colonies de granulocytes/administration et posologie , Facteur de stimulation des colonies de granulocytes/usage thérapeutique , Nourrisson , Résultat thérapeutique , Induction de rémission/méthodes
15.
Hematol Oncol ; 42(3): e3274, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38711253

RÉSUMÉ

Venetoclax, a highly selective BCL-2 inhibitor, combined with hypomethylating agents (HMAs) azacitidine or decitabine, is approved for the treatment of newly diagnosed acute myeloid leukemia (ND AML) in patients who are ineligible to receive intensive chemotherapy. Previous clinical studies initiated venetoclax plus HMA in an inpatient setting owing to concerns of tumor lysis syndrome (TLS). This study (NCT03941964) evaluated the efficacy and safety of venetoclax plus HMA in a United States community-based outpatient setting in patients with ND AML (N = 60) who were treatment naïve for AML, ineligible to receive intensive chemotherapy, had no evidence of spontaneous TLS at screening, and were deemed as appropriate candidates for outpatient initiation of venetoclax plus HMA by the investigator. Patients received venetoclax in combination with azacitidine (75 mg/m2) or decitabine (20 mg/m2) for up to 6 cycles during the study. With a median time on study of 18.3 weeks, the best response rate of composite complete remission was 66.7%, and the overall post-baseline red blood cell (RBC) and platelet transfusion independence rate was 55.0%, consistent with results of studies in which treatment was initiated in an inpatient setting. Key adverse events included nausea, anemia, thrombocytopenia, neutropenia, and white blood cell count decrease of any grade (≥50% of patients). The observed safety profile was generally consistent with that of venetoclax plus HMA observed in inpatient AML studies. With close monitoring, 2 cases of TLS were identified, appropriately managed, and the patients were able to continue study treatment. CLINICAL TRIALS REGISTRATION: This study is registered at ClinicalTrials.gov. The registration identification number is NCT03941964.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Azacitidine , Composés hétérocycliques bicycliques , Décitabine , Leucémie aigüe myéloïde , Sulfonamides , Humains , Sulfonamides/administration et posologie , Sulfonamides/usage thérapeutique , Sulfonamides/effets indésirables , Azacitidine/administration et posologie , Azacitidine/usage thérapeutique , Azacitidine/effets indésirables , Leucémie aigüe myéloïde/traitement médicamenteux , Composés hétérocycliques bicycliques/usage thérapeutique , Composés hétérocycliques bicycliques/administration et posologie , Composés hétérocycliques bicycliques/effets indésirables , Décitabine/administration et posologie , Décitabine/usage thérapeutique , Décitabine/effets indésirables , Femelle , Mâle , Sujet âgé , Adulte d'âge moyen , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Sujet âgé de 80 ans ou plus , Adulte , Patients en consultation externe
18.
Drug Metab Dispos ; 52(6): 555-564, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38565301

RÉSUMÉ

Cytochrome P450 1A2 (CYP1A2) is a known tumor suppressor in hepatocellular carcinoma (HCC), but its expression is repressed in HCC and the underlying mechanism is unclear. In this study, we investigated the epigenetic mechanisms of CYP1A2 repression and potential therapeutic implications. In HCC tumor tissues, the methylation rates of CYP1A2 CpG island (CGI) and DNA methyltransferase (DNMT) 3A protein levels were significantly higher, and there was a clear negative correlation between DNMT3A and CYP1A2 protein expression. Knockdown of DNMT3A by siRNA significantly increased CYP1A2 expression in HCC cells. Additionally, treating HCC cells with decitabine (DAC) resulted in a dose-dependent upregulation of CYP1A2 expression by reducing the methylation level of CYP1A2 CGI. Furthermore, we observed a decreased enrichment of H3K27Ac in the promoter region of CYP1A2 in HCC tissues. Treatment with the trichostatin A (TSA) restored CYP1A2 expression in HCC cells by increasing H3K27Ac levels in the CYP1A2 promoter region. Importantly, combination treatment of sorafenib with DAC or TSA resulted in a leftward shift of the dose-response curve, lower IC50 values, and reduced colony numbers in HCC cells. Our findings suggest that hypermethylation of the CGI at the promoter, mediated by the high expression of DNMT3A, and hypoacetylation of H3K27 in the CYP1A2 promoter region, leads to CYP1A2 repression in HCC. Epigenetic drugs DAC and TSA increase HCC cell sensitivity to sorafenib by restoring CYP1A2 expression. Our study provides new insights into the epigenetic regulation of CYP1A2 in HCC and highlights the potential of epigenetic drugs as a therapeutic approach for HCC. SIGNIFICANCE STATEMENT: This study marks the first exploration of the epigenetic mechanisms underlying cytochrome P450 (CYP) 1A2 suppression in hepatocellular carcinoma (HCC). Our findings reveal that heightened DNA methyltransferase expression induces hypermethylation of the CpG island at the promoter, coupled with diminished H3K27Ac levels, resulting in the repression of CYP1A2 in HCC. The use of epigenetic drugs such as decitabine and trichostatin A emerges as a novel therapeutic avenue, demonstrating their potential to restore CYP1A2 expression and enhance sorafenib sensitivity in HCC cells.


Sujet(s)
Carcinome hépatocellulaire , Cytochrome P-450 CYP1A2 , Méthylation de l'ADN , Épigenèse génétique , Tumeurs du foie , Sorafénib , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Humains , Sorafénib/pharmacologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Épigenèse génétique/effets des médicaments et des substances chimiques , Épigenèse génétique/génétique , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Cytochrome P-450 CYP1A2/génétique , Cytochrome P-450 CYP1A2/métabolisme , DNA methyltransferase 3A , Antinéoplasiques/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , DNA (cytosine-5-)-methyltransferase/génétique , DNA (cytosine-5-)-methyltransferase/métabolisme , Décitabine/pharmacologie , Ilots CpG/génétique , Acides hydroxamiques/pharmacologie , Régions promotrices (génétique)/génétique , Régions promotrices (génétique)/effets des médicaments et des substances chimiques
19.
J Nanobiotechnology ; 22(1): 174, 2024 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-38609922

RÉSUMÉ

Photothermal therapy is favored by cancer researchers due to its advantages such as controllable initiation, direct killing and immune promotion. However, the low enrichment efficiency of photosensitizer in tumor site and the limited effect of single use limits the further development of photothermal therapy. Herein, a photo-responsive multifunctional nanosystem was designed for cancer therapy, in which myeloid-derived suppressor cell (MDSC) membrane vesicle encapsulated decitabine-loaded black phosphorous (BP) nanosheets (BP@ Decitabine @MDSCs, named BDM). The BDM demonstrated excellent biosafety and biochemical characteristics, providing a suitable microenvironment for cancer cell killing. First, the BDM achieves the ability to be highly enriched at tumor sites by inheriting the ability of MDSCs to actively target tumor microenvironment. And then, BP nanosheets achieves hyperthermia and induces mitochondrial damage by its photothermal and photodynamic properties, which enhancing anti-tumor immunity mediated by immunogenic cell death (ICD). Meanwhile, intra-tumoral release of decitabine induced G2/M cell cycle arrest, further promoting tumor cell apoptosis. In vivo, the BMD showed significant inhibition of tumor growth with down-regulation of PCNA expression and increased expression of high mobility group B1 (HMGB1), calreticulin (CRT) and caspase 3. Flow cytometry revealed significantly decreased infiltration of MDSCs and M2-macrophages along with an increased proportion of CD4+, CD8+ T cells as well as CD103+ DCs, suggesting a potentiated anti-tumor immune response. In summary, BDM realizes photothermal therapy/photodynamic therapy synergized chemotherapy for cancer.


Sujet(s)
Cellules myéloïdes suppressives , Tumeurs , Photothérapie dynamique , Biomimétique , Lymphocytes T CD8+ , Décitabine/pharmacologie , Thérapie photothermique , Tumeurs/traitement médicamenteux
20.
Lancet Haematol ; 11(4): e276-e286, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38452788

RÉSUMÉ

BACKGROUND: Hypomethylating agents combined with venetoclax are effective regimens in patients with acute myeloid leukaemia who are ineligible for intensive chemotherapy. Decitabine and cedazuridine (ASTX727) is an oral formulation of decitabine that achieves equivalent area-under-curve exposure to intravenous decitabine. We performed a single centre phase 2 study to evaluate the efficacy and safety of ASTX727 plus venetoclax. METHODS: This study enrolled patients with newly diagnosed (frontline treatment group) acute myeloid leukaemia who were ineligible for intensive chemotherapy (aged ≥75 years, an Eastern Cooperative Oncology Group [ECOG] performance status of 2-3, or major comorbidities) or relapsed or refractory acute myeloid leukaemia. Being aged 18 years or older and having an ECOG performance status of 2 or less were requirements for the relapsed or refractory disease treatment cohort, without any limits in the number of previous lines of therapy. Treatment consisted of ASTX727 (cedazuridine 100 mg and decitabine 35 mg) orally for 5 days and venetoclax 400 mg orally for 21-28 days in 28-day cycles. The primary outcome was overall response rate of ASTX727 plus venetoclax. Living patients who have not completed cycle one were not evaluable for response. Safety was analysed in all patients who started treatment. This study was registered on ClinicalTrials.gov (NCT04746235) and is ongoing. The data cutoff date for this analysis was Sept 22, 2023. FINDINGS: Between March 16, 2021, and Sept 18, 2023, 62 patients were enrolled (49 frontline and 13 relapsed or refractory) with a median age of 78 years (IQR 73-82). 36 (58%) were male; 53 (85%) were White, 4 (6%) Black, 2 (3%) Asian and 3 (5%) other or did not answer. 48 (77%) of 62 patients were European LeukemiaNet 2022 adverse risk, 24 (39%) had antecedent myelodysplastic syndromes, 12 (19%) had previously failed a hypomethylating agent, ten (16%) had therapy-related acute myeloid leukaemia, and 11 (18%) had TP53 mutations. The median follow-up time was 18·3 months (IQR 8·8-23·3). The overall response rate was 30 (64%) of 47 patients (95% CI 49-77) in frontline cohort and six (46%) of 13 patients (19-75) in relapsed or refractory cohort. The most common grade 3 or worse treatment-emergent adverse events were febrile neutropenia in 11 (18%) of 62 patients, pneumonia in eight (13%), respiratory failure in five (8%), bacteraemia in four (6%), and sepsis in four (6%). Three deaths occurred in patients in remission (one sepsis, one gastrointestinal haemorrhage, and one respiratory failure) and were potentially treatment related. INTERPRETATION: ASTX727 plus venetoclax is an active fully oral regimen and safe in most older or unfit patients with acute myeloid leukaemia. Our findings should be confirmed in larger multicentric studies. FUNDING: MD Anderson Cancer Center Support Grant, Myelodysplastic Syndrome/Acute Myeloid Leukaemia Moon Shot, Leukemia SPORE, Taiho Oncology, and Astex Pharmaceuticals.


Sujet(s)
Composés hétérocycliques bicycliques , Association médicamenteuse , Leucémie aigüe myéloïde , Insuffisance respiratoire , Sepsie , Sulfonamides , Uridine/analogues et dérivés , Humains , Mâle , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Décitabine/effets indésirables , Résultat thérapeutique , Leucémie aigüe myéloïde/diagnostic , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Insuffisance respiratoire/induit chimiquement , Insuffisance respiratoire/traitement médicamenteux , Sepsie/induit chimiquement , Sepsie/traitement médicamenteux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...