Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Blood ; 143(20): 2059-2072, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38437498

RESUMO

ABSTRACT: BRG1 (SMARCA4) and BRM (SMARCA2) are the mutually exclusive core ATPases of the chromatin remodeling BAF (BRG1/BRM-associated factor) complexes. They enable transcription factors/cofactors to access enhancers/promoter and modulate gene expressions responsible for cell growth and differentiation of acute myeloid leukemia (AML) stem/progenitor cells. In AML with MLL1 rearrangement (MLL1r) or mutant NPM1 (mtNPM1), although menin inhibitor (MI) treatment induces clinical remissions, most patients either fail to respond or relapse, some harboring menin mutations. FHD-286 is an orally bioavailable, selective inhibitor of BRG1/BRM under clinical development in AML. Present studies show that FHD-286 induces differentiation and lethality in AML cells with MLL1r or mtNPM1, concomitantly causing perturbed chromatin accessibility and repression of c-Myc, PU.1, and CDK4/6. Cotreatment with FHD-286 and decitabine, BET inhibitor (BETi) or MI, or venetoclax synergistically induced in vitro lethality in AML cells with MLL1r or mtNPM1. In models of xenografts derived from patients with AML with MLL1r or mtNPM1, FHD-286 treatment reduced AML burden, improved survival, and attenuated AML-initiating potential of stem-progenitor cells. Compared with each drug, cotreatment with FHD-286 and BETi, MI, decitabine, or venetoclax significantly reduced AML burden and improved survival, without inducing significant toxicity. These findings highlight the FHD-286-based combinations as a promising therapy for AML with MLL1r or mtNPM1.


Assuntos
DNA Helicases , Leucemia Mieloide Aguda , Proteínas Nucleares , Proteínas Proto-Oncogênicas , Fatores de Transcrição , Animais , Humanos , Camundongos , Proteínas que Contêm Bromodomínio , Linhagem Celular Tumoral , DNA Helicases/antagonistas & inibidores , DNA Helicases/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Leucemia Mieloide Aguda/genética , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Blood Cancer J ; 14(1): 25, 2024 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-38316746

RESUMO

Germline, mono-allelic mutations in RUNX1 cause familial platelet disorder (RUNX1-FPD) that evolves into myeloid malignancy (FPD-MM): MDS or AML. FPD-MM commonly harbors co-mutations in the second RUNX1 allele and/or other epigenetic regulators. Here we utilized patient-derived (PD) FPD-MM cells and established the first FPD-MM AML cell line (GMR-AML1). GMR-AML1 cells exhibited active super-enhancers of MYB, MYC, BCL2 and CDK6, augmented expressions of c-Myc, c-Myb, EVI1 and PLK1 and surface markers of AML stem cells. In longitudinally studied bone marrow cells from a patient at FPD-MM vs RUNX1-FPD state, we confirmed increased chromatin accessibility and mRNA expressions of MYB, MECOM and BCL2 in FPD-MM cells. GMR-AML1 and PD FPD-MM cells were sensitive to homoharringtonine (HHT or omacetaxine) or mebendazole-induced lethality, associated with repression of c-Myc, EVI1, PLK1, CDK6 and MCL1. Co-treatment with MB and the PLK1 inhibitor volasertib exerted synergistic in vitro lethality in GMR-AML1 cells. In luciferase-expressing GMR-AML1 xenograft model, MB, omacetaxine or volasertib monotherapy, or co-treatment with MB and volasertib, significantly reduced AML burden and improved survival in the immune-depleted mice. These findings highlight the molecular features of FPD-MM progression and demonstrate HHT, MB and/or volasertib as effective agents against cellular models of FPD-MM.


Assuntos
Transtornos Plaquetários , Leucemia Mieloide Aguda , Humanos , Animais , Camundongos , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mepesuccinato de Omacetaxina , Plaquetas/patologia , Transtornos Plaquetários/complicações , Transtornos Plaquetários/genética , Transtornos Plaquetários/patologia , Proteínas Proto-Oncogênicas c-bcl-2
3.
Leukemia ; 38(3): 545-556, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38086946

RESUMO

AML with chromosomal alterations involving 3q26 overexpresses the transcription factor (TF) EVI1, associated with therapy refractoriness and inferior overall survival in AML. Consistent with a CRISPR screen highlighting BRD4 dependency, treatment with BET inhibitor (BETi) repressed EVI1, LEF1, c-Myc, c-Myb, CDK4/6, and MCL1, and induced apoptosis of AML cells with 3q26 lesions. Tegavivint (TV, BC-2059), known to disrupt the binding of nuclear ß-catenin and TCF7L2/LEF1 with TBL1, also inhibited co-localization of EVI1 with TBL1 and dose-dependently induced apoptosis in AML cell lines and patient-derived (PD) AML cells with 3q26.2 lesions. TV treatment repressed EVI1, attenuated enhancer activity at ERG, TCF7L2, GATA2 and MECOM loci, abolished interactions between MYC enhancers, repressing AML stemness while upregulating mRNA gene-sets of interferon/inflammatory response, TGF-ß signaling and apoptosis-regulation. Co-treatment with TV and BETi or venetoclax induced synergistic in vitro lethality and reduced AML burden, improving survival of NSG mice harboring xenografts of AML with 3q26.2 lesions.


Assuntos
Antineoplásicos , Leucemia Mieloide Aguda , Humanos , Animais , Camundongos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína do Locus do Complexo MDS1 e EVI1/genética , Proteína do Locus do Complexo MDS1 e EVI1/metabolismo , Proteínas Nucleares/genética , Antineoplásicos/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Epigênese Genética , Proto-Oncogenes , Proteínas que Contêm Bromodomínio , Proteínas de Ciclo Celular/genética
4.
Blood Cancer J ; 13(1): 53, 2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37055414

RESUMO

Monotherapy with Menin inhibitor (MI), e.g., SNDX-5613, induces clinical remissions in patients with relapsed/refractory AML harboring MLL1-r or mtNPM1, but most patients either fail to respond or eventually relapse. Utilizing single-cell RNA-Seq, ChiP-Seq, ATAC-Seq, RNA-Seq, RPPA, and mass cytometry (CyTOF) analyses, present pre-clinical studies elucidate gene-expression correlates of MI efficacy in AML cells harboring MLL1-r or mtNPM1. Notably, MI-mediated genome-wide, concordant, log2 fold-perturbations in ATAC-Seq and RNA-Seq peaks were observed at the loci of MLL-FP target genes, with upregulation of mRNAs associated with AML differentiation. MI treatment also reduced the number of AML cells expressing the stem/progenitor cell signature. A protein domain-focused CRISPR-Cas9 screen in MLL1-r AML cells identified targetable co-dependencies with MI treatment, including BRD4, EP300, MOZ and KDM1A. Consistent with this, in vitro co-treatment with MI and BET, MOZ, LSD1 or CBP/p300 inhibitor induced synergistic loss of viability of AML cells with MLL1-r or mtNPM1. Co-treatment with MI and BET or CBP/p300 inhibitor also exerted significantly superior in vivo efficacy in xenograft models of AML with MLL1-r. These findings highlight novel, MI-based combinations that could prevent escape of AML stem/progenitor cells following MI monotherapy, which is responsible for therapy-refractory AML relapse.


Assuntos
Leucemia Mieloide Aguda , Proteína de Leucina Linfoide-Mieloide , Humanos , Proteínas de Ciclo Celular/genética , Epigênese Genética , Histona Desmetilases/genética , Histona-Lisina N-Metiltransferase/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Recidiva Local de Neoplasia/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/genética
5.
Lancet Reg Health Am ; 20: 100461, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36890850

RESUMO

Background: People with immune dysfunction are at higher risk of severe outcomes from COVID-19 infection, but relatively little epidemiologic information is available for mostly vaccinated population in the Omicron era. This population-based study compared relative risk of breakthrough COVID-19 hospitalisation among vaccinated people identified as clinically extremely vulnerable (CEV) vs non-CEV individuals before treatment became more widely available. Methods: COVID-19 cases and hospitalisations reported to the British Columbia Centre for Disease Control (BCCDC) between January 7, 2022 and March 14, 2022 were linked with data on their vaccination and CEV status. Case hospitalisation rates were estimated across CEV status, age groups and vaccination status. For vaccinated individuals, risk ratios for breakthrough hospitalisations were calculated for CEV and non-CEV populations matched on sex, age group, region, and vaccination characteristics. Findings: Among CEV individuals, a total of 5591 COVID-19 reported cases were included, among which 1153 were hospitalized. A third vaccine dose with mRNA vaccine offered additional protection against severe illness in both CEV and non-CEV individuals. However, 2- and 3-dose vaccinated CEV population still had a significantly higher relative risk of breakthrough COVID-19 hospitalisation compared with non-CEV individuals. Interpretation: Vaccinated CEV population remains a higher risk group in the context of circulating Omicron variant and may benefit from additional booster doses and pharmacotherapy. Funding: BC Centre for Disease Control and Provincial Health Services Authority.

6.
Leukemia ; 37(6): 1336-1348, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36977823

RESUMO

In AML with NPM1 mutation causing cytoplasmic dislocation of NPM1, treatments with Menin inhibitor (MI) and standard AML chemotherapy yield complete remissions. However, the causal and mechanistic linkage of mtNPM1 to the efficacy of these agents has not been definitively established. Utilizing CRISPR-Cas9 editing to knockout (KO) or knock-in a copy of mtNPM1 in AML cells, present studies demonstrate that KO of mtNPM1 from AML cells abrogates sensitivity to MI, selinexor (exportin-1 inhibitor), and cytarabine. Conversely, the knock-in of a copy of mtNPM1 markedly sensitized AML cells to treatment with MI or cytarabine. Following AML therapy, most elderly patients with AML with mtNPM1 and co-mutations in FLT3 suffer AML relapse with poor outcomes, creating a need for novel effective therapies. Utilizing the RNA-Seq signature of CRISPR-edited AML cells with mtNPM1 KO, we interrogated the LINCS1000-CMap data set and found several pan-HDAC inhibitors and a WEE1 tyrosine kinase inhibitor among the top expression mimickers (EMs). Additionally, treatment with adavosertib (WEE1 inhibitor) or panobinostat (pan-HDAC inhibitor) exhibited synergistic in vitro lethal activity with MI against AML cells with mtNPM1. Treatment with adavosertib or panobinostat also reduced AML burden and improved survival in AML xenograft models sensitive or resistant to MI.


Assuntos
Leucemia Mieloide Aguda , Proteínas Nucleares , Humanos , Idoso , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Panobinostat , Recidiva Local de Neoplasia , Mutação , Citarabina/farmacologia , Citarabina/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/uso terapêutico
9.
Blood Cancer J ; 12(1): 5, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017466

RESUMO

Treatment with Menin inhibitor (MI) disrupts the interaction between Menin and MLL1 or MLL1-fusion protein (FP), inhibits HOXA9/MEIS1, induces differentiation and loss of survival of AML harboring MLL1 re-arrangement (r) and FP, or expressing mutant (mt)-NPM1. Following MI treatment, although clinical responses are common, the majority of patients with AML with MLL1-r or mt-NPM1 succumb to their disease. Pre-clinical studies presented here demonstrate that genetic knockout or degradation of Menin or treatment with the MI SNDX-50469 reduces MLL1/MLL1-FP targets, associated with MI-induced differentiation and loss of viability. MI treatment also attenuates BCL2 and CDK6 levels. Co-treatment with SNDX-50469 and BCL2 inhibitor (venetoclax), or CDK6 inhibitor (abemaciclib) induces synergistic lethality in cell lines and patient-derived AML cells harboring MLL1-r or mtNPM1. Combined therapy with SNDX-5613 and venetoclax exerts superior in vivo efficacy in a cell line or PD AML cell xenografts harboring MLL1-r or mt-NPM1. Synergy with the MI-based combinations is preserved against MLL1-r AML cells expressing FLT3 mutation, also CRISPR-edited to introduce mtTP53. These findings highlight the promise of clinically testing these MI-based combinations against AML harboring MLL1-r or mtNPM1.


Assuntos
Antineoplásicos/farmacologia , Histona-Lisina N-Metiltransferase/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Proteína de Leucina Linfoide-Mieloide/genética , Nucleofosmina/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Aminopiridinas/farmacologia , Benzimidazóis/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular Tumoral , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Rearranjo Gênico/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/genética , Mutação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Sulfonamidas/farmacologia
10.
Pharmacol Rev ; 74(1): 18-47, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34987087

RESUMO

ERBB4 (HER4) is a member of the ERBB family of receptor tyrosine kinases, a family that includes the epidermal growth factor receptor (EGFR/ERBB1/HER1), ERBB2 (Neu/HER2), and ERBB3 (HER3). EGFR and ERBB2 are oncoproteins and validated targets for therapeutic intervention in a variety of solid tumors. In contrast, the role that ERBB4 plays in human malignancies is ambiguous. Thus, here we review the literature regarding ERBB4 function in human malignancies. We review the mechanisms of ERBB4 signaling with an emphasis on mechanisms of signaling specificity. In the context of this signaling specificity, we discuss the hypothesis that ERBB4 appears to function as a tumor suppressor protein and as an oncoprotein. Next, we review the literature that describes the role of ERBB4 in tumors of the bladder, liver, prostate, brain, colon, stomach, lung, bone, ovary, thyroid, hematopoietic tissues, pancreas, breast, skin, head, and neck. Whenever possible, we discuss the possibility that ERBB4 mutants function as biomarkers in these tumors. Finally, we discuss the potential roles of ERBB4 mutants in the staging of human tumors and how ERBB4 function may dictate the treatment of human tumors. SIGNIFICANCE STATEMENT: This articles reviews ERBB4 function in the context of the mechanistic model that ERBB4 homodimers function as tumor suppressors, whereas ERBB4-EGFR or ERBB4-ERBB2 heterodimers act as oncogenes. Thus, this review serves as a mechanistic framework for clinicians and scientists to consider the role of ERBB4 and ERBB4 mutants in staging and treating human tumors.


Assuntos
Neoplasias , Receptor ErbB-4 , Transdução de Sinais , Humanos , Neoplasias/genética , Receptor ErbB-4/genética
11.
Subst Abus ; 43(1): 92-98, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-32441588

RESUMO

Background:We sought to quantify the association between clinical, physiological, and contextual factors and opioid-related overdose, specifically focusing on current and past use of select prescription medications. Methods: We conducted a case-control study of individuals who experienced a non-fatal opioid-related overdose between January 2015 and November 2016 in British Columbia, Canada. We matched 8,831 cases to 44,155 controls on birth year, sex, and local health area of residence and examined 5-year prescribing history for opioids for pain, medications for opioid use disorder (MOUD), benzodiazepines/z-drugs, and other psychoactive medications. Results: The overall prevalence of prescription opioid drug use was generally low in the study population. Cases had a relatively higher use of selected prescription medications, a higher physical and mental morbidity burden, and were less connected to health services compared with controls. For opioids for pain, current therapy was associated with experiencing an overdose (OR = 8.5, 95%CI: 7.3-10); history of long-term use had a stronger association than history of short-term use (OR = 2.9, 95%CI: 2.6-3.3 vs OR = 1.7, 95%CI: 1.5-1.8, respectively). While persons on MOUD were more likely to overdose compared to persons who were not on therapy (OR = 2.0, 95%CI 1.7-2.4), recent discontinuation of MOUD greatly increased the likelihood of overdose (OR = 25.6, 95%CI 17.5-37.4). Active therapy of benzodiazepines/z-drugs and other sedating medications also significantly increased the likelihood of overdose. Conclusions: While this study supports expansion of efforts to prevent overdoses among individuals actively using opioids for pain and improve retention among those on MOUD, it is also important to address other clinical, physiological, and contextual risk and protective factors to help curb the current overdose crisis.


Assuntos
Overdose de Drogas , Drogas Ilícitas , Overdose de Opiáceos , Transtornos Relacionados ao Uso de Opioides , Medicamentos sob Prescrição , Analgésicos Opioides/uso terapêutico , Benzodiazepinas/uso terapêutico , Colúmbia Britânica/epidemiologia , Estudos de Casos e Controles , Overdose de Drogas/tratamento farmacológico , Overdose de Drogas/epidemiologia , Fentanila , Humanos , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Dor/tratamento farmacológico , Prescrições , Estudos Retrospectivos , Fatores de Risco
12.
Blood ; 139(6): 907-921, 2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-34601571

RESUMO

The majority of RUNX1 mutations in acute myeloid leukemia (AML) are missense or deletion-truncation and behave as loss-of-function mutations. Following standard therapy, AML patients expressing mtRUNX1 exhibit inferior clinical outcome than those without mutant RUNX1. Studies presented here demonstrate that as compared with AML cells lacking mtRUNX1, their isogenic counterparts harboring mtRUNX1 display impaired ribosomal biogenesis and differentiation, as well as exhibit reduced levels of wild-type RUNX1, PU.1, and c-Myc. Compared with AML cells with only wild-type RUNX1, AML cells expressing mtRUNX1 were also more sensitive to the protein translation inhibitor homoharringtonine (omacetaxine) and BCL2 inhibitor venetoclax. Homoharringtonine treatment repressed enhancers and their BRD4 occupancy and was associated with reduced levels of c-Myc, c-Myb, MCL1, and Bcl-xL. Consistent with this, cotreatment with omacetaxine and venetoclax or BET inhibitor induced synergistic in vitro lethality in AML expressing mtRUNX1. Compared with each agent alone, cotreatment with omacetaxine and venetoclax or BET inhibitor also displayed improved in vivo anti-AML efficacy, associated with improved survival of immune-depleted mice engrafted with AML cells harboring mtRUNX1. These findings highlight superior efficacy of omacetaxine-based combination therapies for AML harboring mtRUNX1.


Assuntos
Antineoplásicos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Mepesuccinato de Omacetaxina/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Inibidores da Síntese de Proteínas/farmacologia , Sulfonamidas/farmacologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Leucemia Mieloide Aguda/genética , Mutação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores
13.
Blood Cancer J ; 11(5): 98, 2021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-34016956

RESUMO

There is an unmet need to overcome nongenetic therapy-resistance to improve outcomes in AML, especially post-myeloproliferative neoplasm (MPN) secondary (s) AML. Studies presented describe effects of genetic knockout, degradation or small molecule targeted-inhibition of GFI1/LSD1 on active enhancers, altering gene-expressions and inducing differentiation and lethality in AML and (MPN) sAML cells. A protein domain-focused CRISPR screen in LSD1 (KDM1A) inhibitor (i) treated AML cells, identified BRD4, MOZ, HDAC3 and DOT1L among the codependencies. Our findings demonstrate that co-targeting LSD1 and one of these co-dependencies exerted synergistic in vitro lethality in AML and post-MPN sAML cells. Co-treatment with LSD1i and the JAKi ruxolitinib was also synergistically lethal against post-MPN sAML cells. LSD1i pre-treatment induced GFI1, PU.1 and CEBPα but depleted c-Myc, overcoming nongenetic resistance to ruxolitinib, or to BETi in post-MPN sAML cells. Co-treatment with LSD1i and BETi or ruxolitinib exerted superior in vivo efficacy against post-MPN sAML cells. These findings highlight LSD1i-based combinations that merit testing for clinical efficacy, especially to overcome nongenetic therapy-resistance in AML and post-MPN sAML.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Histona Desmetilases/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Transtornos Mieloproliferativos/tratamento farmacológico , Fatores de Transcrição/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Inativação Gênica/efeitos dos fármacos , Histona Desmetilases/genética , Humanos , Leucemia Mieloide Aguda/genética , Terapia de Alvo Molecular , Transtornos Mieloproliferativos/genética , Fatores de Transcrição/genética , Transcriptoma/efeitos dos fármacos
14.
Blood Cancer J ; 11(3): 64, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33753715

RESUMO

Ecotropic viral integration site 1 (Evi1) was discovered in 1988 as a common site of ecotropic viral integration resulting in myeloid malignancies in mice. EVI1 is an oncogenic zinc-finger transcription factor whose overexpression contributes to disease progression and an aggressive phenotype, correlating with poor clinical outcome in myeloid malignancies. Despite progress in understanding the biology of EVI1 dysregulation, significant improvements in therapeutic outcome remain elusive. Here, we highlight advances in understanding EVI1 biology and discuss how this new knowledge informs development of novel therapeutic interventions. EVI1 is overexpression is correlated with poor outcome in some epithelial cancers. However, the focus of this review is the genetic lesions, biology, and current therapeutics of myeloid malignancies overexpressing EVI1.


Assuntos
Leucemia Mieloide/genética , Proteína do Locus do Complexo MDS1 e EVI1/genética , Animais , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Instabilidade Genômica , Humanos , Leucemia Mieloide/patologia , Leucemia Mieloide/terapia , Mutação , Processamento de Proteína Pós-Traducional , Ativação Transcricional
15.
Leukemia ; 35(9): 2621-2634, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33654205

RESUMO

Richter Transformation (RT) develops in CLL as an aggressive, therapy-resistant, diffuse large B cell lymphoma (RT-DLBCL), commonly clonally-related (CLR) to the concomitant CLL. Lack of available pre-clinical human models has hampered the development of novel therapies for RT-DLBCL. Here, we report the profiles of genetic alterations, chromatin accessibility and active enhancers, gene-expressions and anti-lymphoma drug-sensitivity of three newly established, patient-derived, xenograft (PDX) models of RT-DLBCLs, including CLR and clonally-unrelated (CLUR) to concomitant CLL. The CLR and CLUR RT-DLBCL cells display active enhancers, higher single-cell RNA-Seq-determined mRNA, and protein expressions of IRF4, TCF4, and BCL2, as well as increased sensitivity to BET protein inhibitors. CRISPR knockout of IRF4 attenuated c-Myc levels and increased sensitivity to a BET protein inhibitor. Co-treatment with BET inhibitor or BET-PROTAC and ibrutinib or venetoclax exerted synergistic in vitro lethality in the RT-DLBCL cells. Finally, as compared to each agent alone, combination therapy with BET-PROTAC and venetoclax significantly reduced lymphoma burden and improved survival of immune-depleted mice engrafted with CLR-RT-DLBCL. These findings highlight a novel, potentially effective therapy for RT-DLBCL.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/metabolismo , Transformação Celular Neoplásica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Proteínas/metabolismo , Proteólise , Adenina/administração & dosagem , Adenina/análogos & derivados , Animais , Apoptose , Biomarcadores Tumorais/genética , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Humanos , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/patologia , Camundongos , Piperidinas/administração & dosagem , Proteínas/genética , Sulfonamidas/administração & dosagem , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
16.
PLoS One ; 15(12): e0243901, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33378376

RESUMO

Whereas recent clinical studies report metastatic melanoma survival rates high as 30-50%, many tumors remain nonresponsive or become resistant to current therapeutic strategies. Analyses of The Cancer Genome Atlas (TCGA) skin cutaneous melanoma (SKCM) data set suggests that a significant fraction of melanomas potentially harbor gain-of-function mutations in the gene that encodes for the ErbB4 receptor tyrosine kinase. In this work, a drug discovery strategy was developed that is based on the observation that the Q43L mutant of the naturally occurring ErbB4 agonist Neuregulin-2beta (NRG2ß) functions as a partial agonist at ErbB4. NRG2ß/Q43L stimulates tyrosine phosphorylation, fails to stimulate ErbB4-dependent cell proliferation, and inhibits agonist-induced ErbB4-dependent cell proliferation. Compounds that exhibit these characteristics likely function as ErbB4 partial agonists, and as such hold promise as therapies for ErbB4-dependent melanomas. Consequently, three highly sensitive and reproducible (Z' > 0.5) screening assays were developed and deployed for the identification of small-molecule ErbB4 partial agonists. Six compounds were identified that stimulate ErbB4 phosphorylation, fail to stimulate ErbB4-dependent cell proliferation, and appear to selectively inhibit ErbB4-dependent cell proliferation. Whereas further characterization is needed to evaluate the full therapeutic potential of these molecules, this drug discovery platform establishes reliable and scalable approaches for the discovery of ErbB4 inhibitors.


Assuntos
Proliferação de Células/genética , Melanoma/genética , Fatores de Crescimento Neural/genética , Receptor ErbB-4/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Descoberta de Drogas , Mutação com Ganho de Função/genética , Humanos , Melanoma/tratamento farmacológico , Melanoma/patologia , Fosforilação/genética , Receptor ErbB-4/agonistas , Receptor ErbB-4/antagonistas & inibidores , Transdução de Sinais/genética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
17.
Blood ; 135(15): 1255-1269, 2020 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-32068780

RESUMO

The promising activity of BET protein inhibitors (BETi's) is compromised by adaptive or innate resistance in acute myeloid leukemia (AML). Here, modeling of BETi-persister/resistance (BETi-P/R) in human postmyeloproliferative neoplasm (post-MPN) secondary AML (sAML) cells demonstrated accessible and active chromatin in specific superenhancers/enhancers, which was associated with increased levels of nuclear ß-catenin, TCF7L2, JMJD6, and c-Myc in BETi-P/R sAML cells. Following BETi treatment, c-Myc levels were rapidly restored in BETi-P/R sAML cells. CRISPR/Cas9-mediated knockout of TCF7L2 or JMJD6 reversed BETi-P/R, whereas ectopic overexpression conferred BETi-P/R in sAML cells, confirming the mechanistic role of the ß-catenin-TCF7L2-JMJD6-c-Myc axis in BETi resistance. Patient-derived, post-MPN, CD34+ sAML blasts exhibiting relative resistance to BETi, as compared with sensitive sAML blasts, displayed higher messenger RNA and protein expression of TCF7L2, JMJD6, and c-Myc and following BETi washout exhibited rapid restoration of c-Myc and JMJD6. CRISPR/Cas9 knockout of TCF7L2 and JMJD6 depleted their levels, inducing loss of viability of the sAML blasts. Disruption of colocalization of nuclear ß-catenin with TBL1 and TCF7L2 by the small-molecule inhibitor BC2059 combined with depletion of BRD4 by BET proteolysis-targeting chimera reduced c-Myc levels and exerted synergistic lethality in BETi-P/R sAML cells. This combination also reduced leukemia burden and improved survival of mice engrafted with BETi-P/R sAML cells or patient-derived AML blasts innately resistant to BETi. Therefore, multitargeted disruption of the ß-catenin-TCF7L2-JMJD6-c-Myc axis overcomes adaptive and innate BETi resistance, exhibiting preclinical efficacy against human post-MPN sAML cells.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/antagonistas & inibidores , Antineoplásicos/química , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo
18.
Int J Drug Policy ; 77: 102665, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31962283

RESUMO

BACKGROUND: North America is in the midst of an opioid overdose epidemic and it is commonly suggested that exposure to fentanyl is unknown. Using a provincial survey of harm reduction site clients, we aimed to characterize known and unknown fentanyl use and their correlates among people who use drugs in British Columbia, Canada. METHODS: We recruited 486 clients who were >18 years old and 316 agreed to provide a urine sample for substance use testing. Reported known fentanyl use was defined as a three-level categorical variable assessing recent (i.e., in the previous three days) fentanyl exposure: (i) known exposure; (ii) unknown exposure; and (iii) no exposure. We also assessed any exposure to fentanyl (Yes vs. No) confirmed by urinalysis. Survey data were summarized using descriptive statistics. Multinomial logistic regression and modified Poisson regression models were built to examine different correlates of exposure to fentanyl. RESULTS: Median age of the participants was 40 (IQR: 32-49). Out of the 303 eligible participants, 38.7% (117) reported known fentanyl use, 21.7% (66) had unknown fentanyl use, and 39.6% (120) had no recent fentanyl use. In the adjusted multinomial logistic regression model and in comparison with unknown fentanyl use, recent known fentanyl use was significantly associated with self-report of methadone use (aRRR = 3.18), heroin/morphine use (aRRR = 4.40), and crystal meth use (aRRR = 2.95). Moreover, any recent exposure to fentanyl (i.e., positive urine test for fentanyl) was significantly associated with living in urban settings (aPR = 1.49), and self-reporting recent cannabis use (aPR = 0.73), crystal meth (aPR = 1.45), and heroin/morphine use (aPR = 2.48). CONCLUSION: The landscape of illicit opioid use is changing in BC and more people are using fentanyl knowingly. The increasing prevalence of known fentanyl use is concerning and calls for further investments in public awareness and public policy efforts regarding fentanyl exposure and risks.


Assuntos
Fentanila , Redução do Dano , Drogas Ilícitas , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Adulto , Colúmbia Britânica/epidemiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/etiologia , Transtornos Relacionados ao Uso de Opioides/urina , Adulto Jovem
19.
J Allergy Clin Immunol Pract ; 8(1): 149-156.e1, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31301377

RESUMO

BACKGROUND: Because of inaccuracies in commonly used tests for food allergy, oral food challenges (OFCs) are considered the criterion standard, but OFC implementation is suboptimal. OBJECTIVE: To use a mixed-methods approach to describe OFC barriers at multiple levels and investigate solutions. METHODS: Surveys of Canadian allergists, pediatricians, and parents investigated barriers to offering or participating in OFCs, and possible solutions. Parent focus groups were held to understand these barriers and solutions. Allergist offices in British Columbia were contacted via telephone to confirm their OFC practices. RESULTS: Of 62 responding allergists, 80.6% reported performing OFCs, 72.6% reported lack of resources as an influential barrier, and 72.6% reported that creation of standard guidelines for hospital versus community OFCs would influence them to perform more OFCs. Of 101 responding pediatricians, 51.5% reported having moderate-to-extensive OFC knowledge; these pediatricians were more likely to refer to allergists who performed them (odds ratio, 2.37; 95% CI, 1.06-5.30). Of 27 pediatricians who stated they refer more to allergists who do not perform OFCs, 40.7% reported long wait times as a deterrent. The most common parent barriers from surveys (N = 110) and focus groups (N = 27) were fear and anxiety about the procedure and about experiencing reactions during OFCs, suggesting the need for better information and psychosocial resources. CONCLUSIONS: Multiple barriers prevent widespread use of OFCs. Efforts targeting OFC training for allergists, education for pediatricians, and standardized guidelines created with clinician and parent input (including consistent OFC information for families and guidance on which OFCs should be performed in-hospital versus the community) are likely to increase OFC acceptance.


Assuntos
Alérgenos , Hipersensibilidade Alimentar , Alergistas , Colúmbia Britânica , Alimentos , Humanos
20.
Dysphagia ; 35(5): 773-779, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31773332

RESUMO

Eosinophilic esophagitis (EoE) is a chronic condition that requires repeated endoscopies/biopsies to track the disease and treatment response. This invasive procedure involves risk to the patient and has significant costs. We studied whether the detection of specific proteins (cytokines and eosinophil degranulation products) from oral swabs could serve as a minimally invasive test for EoE. Swabs of the oral cavity (buccal and oropharyngeal) were obtained prior to endoscopy/biopsies in patients with EoE, possible EoE, and non-EoE patients in addition to obtaining additional esophageal biopsy tissue. ELISAs measuring the levels of cytokines IL-5, IL-8, IL-13, and eosinophil degranulation products including major basic protein (MBP), eosinophil derived neurotoxin (EDN), and eosinophil peroxidase (EPO) were performed on the samples. Comparisons were made to peak esophageal eosinophil counts. Tolerability of the swabs was evaluated. 43 patients, 4-17 years old, participated in the study. Swabs were well tolerated and all showed measurable protein. 26 patients had EoE [14 active (> 15 eosinophils/high power field), 12 non-active], 17 patients did not have EoE. Results obtained from oral swabs showed poor correlation with those from esophageal tissue. Only measurement of eosinophil degranulation products EDN and EPO from esophageal tissues showed strong correlations with eosinophil counts. In this study, the levels of cytokines and eosinophil degranulation products detected from oral swabs did not correlate with esophageal eosinophilia, and their detection would be insufficient to displace endoscopy/biopsies.


Assuntos
Esofagite Eosinofílica , Eosinófilos , Adolescente , Biomarcadores , Criança , Pré-Escolar , Neurotoxina Derivada de Eosinófilo , Esofagite Eosinofílica/diagnóstico , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...