Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Blood ; 141(22): 2738-2755, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36857629

RESUMO

Primary resistance to tyrosine kinase inhibitors (TKIs) is a significant barrier to optimal outcomes in chronic myeloid leukemia (CML), but factors contributing to response heterogeneity remain unclear. Using single-cell RNA (scRNA) sequencing, we identified 8 statistically significant features in pretreatment bone marrow, which correlated with either sensitivity (major molecular response or MMR) or extreme resistance to imatinib (eventual blast crisis [BC] transformation). Employing machine-learning, we identified leukemic stem cell (LSC) and natural killer (NK) cell gene expression profiles predicting imatinib response with >80% accuracy, including no false positives for predicting BC. A canonical erythroid-specifying (TAL1/KLF1/GATA1) regulon was a hallmark of LSCs from patients with MMR and was associated with erythroid progenitor [ERP] expansion in vivo (P < .05), and a 2- to 10-fold (6.3-fold in group A vs 1.09-fold in group C) erythroid over myeloid bias in vitro. Notably, ERPs demonstrated exquisite TKI sensitivity compared with myeloid progenitors (P < .001). These LSC features were lost with progressive resistance, and MYC- and IRF1-driven inflammatory regulons were evident in patients who progressed to transformation. Patients with MMR also exhibited a 56-fold expansion (P < .01) of a normally rare subset of hyperfunctional adaptive-like NK cells, which diminished with progressive resistance, whereas patients destined for BC accumulated inhibitory NKG2A+ NK cells favoring NK cell tolerance. Finally, we developed antibody panels to validate our scRNA-seq findings. These panels may be useful for prospective studies of primary resistance, and in assessing the contribution of predetermined vs acquired factors in TKI response heterogeneity.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva , Inibidores de Proteínas Quinases , Humanos , Mesilato de Imatinib/farmacologia , Mesilato de Imatinib/uso terapêutico , Estudos Prospectivos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Crise Blástica , Resistencia a Medicamentos Antineoplásicos/genética
2.
Mol Carcinog ; 62(12): 1846-1859, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37589421

RESUMO

Pulmonary lymphoepithelioma-like carcinoma (pLELC) is a rare malignancy that lacks specific biomarkers. N6-methyladenosine (m6 A) is the most widespread internal modification of messenger RNA (mRNA), and its dysregulation is involved in the development of many cancers. However, the expression of m6 A genes in pLELC and their roles are unknown. We obtained an exosomal transcriptome data set of patients diagnosed with pLELC and healthy controls using RNA sequencing and identified differentially expressed genes (DEGs) in the two groups using R software. The differential expression of the 37 m6 A genes in the two sets of samples was further analyzed, and receiver operating characteristic (ROC) curves were plotted for each gene to identify their grouping ability. The STRING database was used to construct a protein-protein interaction network for m6 A genes. An mRNA-miRNA regulatory network of m6 A-related DEGs was constructed using the miRNet database, and a prediction score formula was established. A nomogram was constructed based on the candidate m6 A genes and prediction scores. The expression of key genes was determined through the immunohistochemical (IHC) staining of clinical tissue sections. Using ROC curves, nine m6 A genes were revealed to have classification efficacy in both groups of samples. We screened seven m6 A-related DEGs (MAN2C1, HNRNPCL1, FUS, EIF6, DIP2A, COA3, and BUD13) that were beneficial for grouping and constructed nomogram models. Through IHC, we identified FUS and EIF6 as being possibly involved in the occurrence and development of pLELC. The m6 A gene expression patterns in pLELC-derived exosomes were significantly different from those in healthy controls. We screened several key genes to facilitate the development of diagnostic markers for pulmonary lymphoepithelioma.


Assuntos
Carcinoma de Células Escamosas , Humanos , Metilação , Perfilação da Expressão Gênica , Transcriptoma , Adenosina/genética , RNA Mensageiro/genética
3.
Discov Oncol ; 14(1): 129, 2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37452162

RESUMO

Enrichment of Veillonella parvula in the lung microbiota is strongly associated with non-small cell lung cancer (NSCLC) and induces the progression of lung adenocarcinoma in vivo, but its actual role and mechanism remain unexplored. This study analyzed the correlation between NSCLC and V. parvula abundance based on 16 s rRNA sequencing results. The effects of V. parvula on the progression of lung adenocarcinoma were observed in vivo and in vitro using a C57 bl/6j mouse tumor-bearing model, a bacterial cell co-culture model, combined with transcriptome sequencing, and a TCGA database to explore and validate the growth promotion of lung adenocarcinoma by V. parvula and its molecular mechanism. 16 s rRNA sequencing revealed that V. parvula was significantly enriched in lung adenocarcinoma. In vivo, V. parvula promoted the growth of lung adenocarcinoma in mice by suppressing the infiltration of tumor-associated T lymphocytes and peripheral T lymphocytes. It showed a higher affinity for lung adenocarcinoma in vitro and promoted lung adenocarcinoma cell proliferation through adhesion or intracellular invasion. Further analysis of differential gene expression and KEGG enrichment by transcriptome sequencing revealed that V. parvula induced CCN4 expression and activated NOD-like receptor and NF-κB signaling pathway in lung adenocarcinoma cells. Further analysis clarified that V. parvula promoted activation of the NF-κB pathway via Nod2/CCN4 signaling, which promoted lung adenocarcinoma cell proliferation. Thus, V. parvula mediates activation of the Nod2/CCN4/NF-κB signaling pathway to promote non-small cell lung adenocarcinoma progression, thereby providing a potential target for diagnosing and treating lung adenocarcinoma.

4.
J Cancer Res Clin Oncol ; 149(3): 1185-1193, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35377040

RESUMO

PURPOSE: Primary pulmonary lympho-epithelioma-like carcinoma (PPLELC) is a rare subtype of primary non-small cell lung cancer (NSCLC). Currently, there is still lack of research data on anti-angiogenic therapy of advanced PPLELC. The purpose of this study was to investigate the efficacy and safety of anti-angiogenic therapy combined with chemotherapy compared with traditional chemotherapy for these patients. METHODS: Advanced PPLELC patients admitted to six grade A hospitals from January 2013 to January 2021 were selected. The patients received anti-angiogenic therapy combined with chemotherapy (AT group) or chemotherapy (CT group) alone. RESULTS: A total of 65 patients were included in this study, including 31 patients in the AT group treated with anti-angiogenic therapy combined with chemotherapy and 34 patients in the CT group treated with chemotherapy alone. As of October 1, 2021, the median progression-free survival (PFS) in the AT group was 11.2 months [95% confidence interval (CI), 5.9-16.5]. The median PFS in the CT group was 7.0 months [95%CI, 5.1-8.9] [Hazard Ratio (HR), 0.49; 95%CI, 0.29-0.83; P = 0.008]. The 1-year PFS rates were 41.9% and 17.6%, respectively. The overall response rates (ORR) of two groups were 45.2% (95% CI, 0.27-0.64), 38.2% (95% CI, 0.21-0.56), (P = 0.571). The disease control rates (DCR) of two groups were 93.5% (95% CI, 0.84-1.03), 88.2% (95% CI, 0.77-1.00), (P = 0.756). CONCLUSION: Among patients with advanced PPLELC, the PFS of patients with anti-angiogenic therapy combined with chemotherapy is better than that of patients with chemotherapy alone. Anti-angiogenic therapy combined with chemotherapy is an optional treatment scheme.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Carcinoma , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Intervalo Livre de Progressão , Imunoterapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
5.
Bioengineered ; 13(3): 6665-6677, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35254206

RESUMO

The role of lung microbiota in non-small cell lung cancer remains unclear. We investigated the characteristics and functional roles of lung microbiota in non-small cell lung cancer. Bronchoalveolar lavage fluid samples were obtained from patients with non-small cell lung cancer (n = 46) and with benign lung disease (n = 29). The differences in composition and gene expression in the microbiota between the samples were analyzed using 16s rRNA sequencing. The oncogenic genus (Veillonella) was then evaluated in the progression of lung cancer in C57 BL/6 mice. Compared to benign lung disease, the lung microbiota in non-small cell lung cancer was significantly altered, both in terms of α- and ß-diversity. In terms of bacterial composition, the non-small cell lung cancer group was enriched with two Phyla (Firmicutes, Bacteroidetes) and three genera (Streptococcus, Prevotella, Veillonella). Prevotella and Veillonella were most strongly associated with non-small cell lung cancer, and Veillonella significantly promoted the progression of lung cancer in vivo. Moreover, metabolic prediction revealed that ribosomes, biosynthesis of secondary metabolites, and pyrimidine metabolism were among the enriched pathways that may be involved in the progression of non-small cell lung cancer. Overall, results suggest that the progression of non-small cell lung cancer is followed by significant changes in the composition and function of the lung microbiota. These differing genera may be potential diagnostic markers and therapeutic targets.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Microbiota , Animais , Humanos , Pulmão , Neoplasias Pulmonares/genética , Camundongos , Microbiota/genética , RNA Ribossômico 16S/genética
6.
Leukemia ; 34(7): 1787-1798, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32051529

RESUMO

Patients with chronic myeloid leukemia (CML) who are treated with tyrosine kinase inhibitors (TKIs) experience significant heterogeneity regarding depth and speed of responses. Factors intrinsic and extrinsic to CML cells contribute to response heterogeneity and TKI resistance. Among extrinsic factors, cytokine-mediated TKI resistance has been demonstrated in CML progenitors, but the underlying mechanisms remain obscure. Using RNA-sequencing, we identified differentially expressed splicing factors in primary CD34+ chronic phase (CP) CML progenitors and controls. We found SRSF1 expression to be increased as a result of both BCR-ABL1- and cytokine-mediated signaling. SRSF1 overexpression conferred cytokine independence to untransformed hematopoietic cells and impaired imatinib sensitivity in CML cells, while SRSF1 depletion in CD34+ CP CML cells prevented the ability of extrinsic cytokines to decrease imatinib sensitivity. Mechanistically, PRKCH and PLCH1 were upregulated by elevated SRSF1 levels, and contributed to impaired imatinib sensitivity. Importantly, very high SRSF1 levels in the bone marrow of CML patients at presentation correlated with poorer clinical TKI responses. In summary, we find SRSF1 levels to be maintained in CD34+ CP CML progenitors by cytokines despite effective BCR-ABL1 inhibition, and that elevated levels promote impaired imatinib responses. Together, our data support an SRSF1/PRKCH/PLCH1 axis in contributing to cytokine-induced impaired imatinib sensitivity in CML.


Assuntos
Medula Óssea/patologia , Citocinas/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Mesilato de Imatinib/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Células-Tronco Neoplásicas/patologia , Fatores de Processamento de Serina-Arginina/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Inibidores de Proteínas Quinases/farmacologia , Fatores de Processamento de Serina-Arginina/genética , Células Tumorais Cultivadas
7.
Chin Med J (Engl) ; 130(2): 211-218, 2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-28091414

RESUMO

OBJECTIVE: Leukemia is the most common pediatric malignancy and a major cause of morbidity and mortality in children. Among all subtypes, a lack of consensus exists regarding the diagnosis and treatment of acute myeloid leukemia (AML). Patient survival rates have remained modest for the past three decades in AML. Recently, targeted therapy has emerged as a promising treatment. DATA SOURCES: We searched the PubMed database for recently published research papers on diagnostic development, target therapy, and other novel therapies of AML. Clinical trial information was obtained from ClinicalTrials.gov. For the major purpose of this review that is to outline the latest therapeutic development of AML, we only listed the ongoing clinical trials for reference. However, the published results of complete clinical trials were also mentioned. STUDY SELECTION: This article reviewed the latest developments related to the diagnosis and treatment of AML. In the first portion, we provided some novel insights on the molecular basis of AML, as well as provided an update on the classification of AML. In the second portion, we summarized the results of research on potential molecular therapeutic agents including monoclonal antibodies, tyrosine kinase/Fms-like tyrosine kinase 3 (FLT3) inhibitors, epigenetic/demethylating agents, and cellular therapeutic agents. We will also highlight ongoing research and clinical trials in pediatric AML. RESULTS: We described clonal evolution and how it changes our view on leukemogenesis, treatment responses, and disease relapse. Pediatric-specific genomic mapping was discussed with a novel diagnostic method highlighted. In the later portion of this review, we summarized the researches on potential molecular therapeutic agents including monoclonal antibodies, tyrosine kinase/FLT3 inhibitors, epigenetic/demethylating agents, and cellular therapeutic agents. CONCLUSION: Gene sequencing techniques should set the basis for next-generation diagnostic methods of AML, and target therapy should be the focus of future clinical research in the exploration of therapeutic possibilities.


Assuntos
Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Humanos , Inibidores de Proteínas Quinases/uso terapêutico , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA