Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.884
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Blood ; 141(4): 380-390, 2023 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-36322938

RESUMO

Myeloproliferative neoplasms (MPN) are a group of clonal stem cell-derived hematopoietic malignancies driven by aberrant Janus kinase-signal transducer and activator of transcription proteins (JAK/STAT) signaling. Although these are genetically simple diseases, MPNs are phenotypically heterogeneous, reflecting underlying intratumoral heterogeneity driven by the interplay of genetic and nongenetic factors. Their evolution is determined by factors that enable certain cellular subsets to outcompete others. Therefore, techniques that resolve cellular heterogeneity at the single-cell level are ideally placed to provide new insights into MPN biology. With these insights comes the potential to uncover new approaches to predict the clinical course and treat these cancers, ultimately improving outcomes for patients. MPNs present a particularly tractable model of cancer evolution, because most patients present in an early disease phase and only a small proportion progress to aggressive disease. Therefore, it is not surprising that many groundbreaking technological advances in single-cell omics have been pioneered by their application in MPNs. In this review article, we explore how single-cell approaches have provided transformative insights into MPN disease biology, which are broadly applicable across human cancers, and discuss how these studies might be swiftly translated into clinical pathways and may eventually underpin precision medicine.


Assuntos
Neoplasias da Medula Óssea , Neoplasias Hematológicas , Transtornos Mieloproliferativos , Neoplasias , Humanos , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/terapia , Transtornos Mieloproliferativos/metabolismo , Janus Quinases/metabolismo , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/terapia , Neoplasias Hematológicas/metabolismo , Transdução de Sinais , Janus Quinase 2/genética , Mutação
2.
Br J Haematol ; 204(1): 56-67, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38083878

RESUMO

Cyclic thrombocytopenia (CTP) is characterized by periodic platelet oscillation with substantial amplitude. Most CTP cases have a thrombocytopenic background and are often misdiagnosed as immune thrombocytopenia with erratically effective treatment choices. CTP also occurs during hydroxyurea treatment in patients with myeloproliferative diseases. While the aetiology of CTP remains uncertain, here we evaluate historical, theoretical and clinical findings to provide a framework for understanding CTP pathophysiology. CTP retains the intrinsic oscillatory factors defined by the homeostatic regulation of platelet count, presenting as reciprocal platelet/thrombopoietin oscillations and stable oscillation periodicity. Moreover, CTP patients possess pathogenic factors destabilizing the platelet homeostatic system thereby creating opportunities for external perturbations to initiate and sustain the exaggerated platelet oscillations. Beyond humoral and cell-mediated autoimmunity, we propose recently uncovered germline and somatic genetic variants, such as those of MPL, STAT3 or DNMT3A, as pathogenic factors in thrombocytopenia-related CTP. Likewise, the JAK2 V617F or BCR::ABL1 translocation that drives underlying myeloproliferative diseases may also play a pathogenic role in hydroxyurea-induced CTP, where hydroxyurea treatment can serve as both a trigger and a pathogenic factor of platelet oscillation. Elucidating the pathogenic landscape of CTP provides an opportunity for targeted therapeutic approaches in the future.


Assuntos
Neoplasias da Medula Óssea , Transtornos Mieloproliferativos , Trombocitopenia , Humanos , Hidroxiureia/uso terapêutico , Trombocitopenia/etiologia , Trombocitopenia/genética , Contagem de Plaquetas , Plaquetas , Transtornos Mieloproliferativos/genética
3.
Ann Hematol ; 103(5): 1513-1523, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-37665349

RESUMO

The exact prognostic role of cardiovascular (CV) risk factors in patients with BCR::ABL1 negative chronic myeloproliferative neoplasms (MPNs) remains unknown as it is often masked by other MPN-related features that bear strong prognostic impact on thrombotic risk. Therefore, current MPN treatment is not primarily guided by presence of CV risk factors. Treatment of CV risk factors in MPN patients usually mirrors that from the general population, despite the fact that CV risk factors in MPNs have their own specificities. Moreover, the optimal target levels for different metabolic deflections in MPNs (i.e., low-density lipoprotein, serum uric acid, or glycated hemoglobin levels) have not been defined. In the current review, we separately discuss the most important aspects of every individual CV risk factor (arterial hypertension, hyperlipidemia, chronic kidney disease, smoking, diabetes mellitus, hyperuricemia, and obesity and cachexia) in MPNs, summarize recent advances in the field, and propose future directions and research areas which may be needed to appropriately manage CV risk factors in MPNs.


Assuntos
Neoplasias da Medula Óssea , Doenças Cardiovasculares , Transtornos Mieloproliferativos , Humanos , Neoplasias da Medula Óssea/genética , Neoplasias da Medula Óssea/terapia , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/etiologia , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/terapia , Fatores de Risco , Ácido Úrico/sangue , Ácido Úrico/química , Proteínas de Fusão bcr-abl
4.
J Surg Res ; 298: 240-250, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38631173

RESUMO

INTRODUCTION: Neuroblastoma (NB) is the most common extra-cranial malignancy in children. Poor survival in high-risk NB is attributed to recurrent metastatic disease. To better study metastatic disease, we used a novel mouse model to investigate differential gene expression between primary tumor cells and metastatic cells. We hypothesized that metastatic NB cells have a different gene expression profile from primary tumor cells and cultured cells. METHODS: Using three human NB cell lines (NGP, CHLA255, and SH-SY5Y), orthotopic xenografts were established in immunodeficient nod/scid gamma mice via subcapsular renal injection. Mice were sacrificed and NB cells were isolated from the primary tumor and from sites of metastasis (bone marrow, liver). RNA sequencing, gene set analysis, and pathway analysis were performed to identify differentially expressed genes and molecular pathways in the metastatic cells compared to primary tumor cells. RESULTS: There were 266 differentially expressed genes in metastatic tumor cells (bone marrow and liver combined) compared to primary tumor cells. The top upregulated gene was KCNK1 and the top downregulated genes were PDE7B and NEBL. Top upregulated pathways in the metastatic cells were involved in ion transport, cell signaling, and cell proliferation. Top downregulated pathways were involved in DNA synthesis, transcription, and cellular metabolism. CONCLUSIONS: In metastatic NB cells, our study identified the upregulation of biologic processes involved in cell cycle regulation, cell proliferation, migration, and invasion. Ongoing studies aim to validate downstream translation of these genomic alterations, as well as target these pathways to more effectively suppress and inhibit recurrent metastatic disease in NB.


Assuntos
Regulação Neoplásica da Expressão Gênica , Camundongos Endogâmicos NOD , Camundongos SCID , Neuroblastoma , Animais , Neuroblastoma/patologia , Neuroblastoma/genética , Neuroblastoma/metabolismo , Humanos , Camundongos , Linhagem Celular Tumoral , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias da Medula Óssea/secundário , Neoplasias da Medula Óssea/genética , Perfilação da Expressão Gênica , Transcriptoma
5.
Eur J Pediatr ; 183(1): 185-202, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37855927

RESUMO

Patients with high-risk neuroblastoma (HR-NB) exhibit suboptimal 5-year survival rates, leading to a widespread international preference for high-intensity chemotherapeutic regimens in these children. We analyzed the incidence and risk factors for complications during induction chemotherapy in children with HR-NB and tried to assist clinicians in predicting such complications and optimizing therapeutic strategy. The clinical data of children with HR-NB admitted to our hospital from January 2007 to December 2019 were retrospectively analyzed. The incidence, characteristics, and risk factors of complications (infection, hemorrhage, and chemotherapy-related adverse reactions (CRAR)) requiring hospitalization during induction chemotherapy in these children were explored. (1) A total of 108 patients with HR-NB were included in the final analysis. The overall infection rate was 92.6% (100/108), with the highest incidence of 71.3% observed during the first cycle. FN, bacterial infection, as well as fungal infection were common infectious complications in children with HR-NB during induction chemotherapy. (2) The overall hemorrhage rate was 24.1% (26/108), with the highest incidence of 14.8% also observed in the first cycle. Among the children with hemorrhage, there were 72% with bone marrow involved, while 65.0% of them had a high vanillylmandelic acid (VMA) value. And children with hemorrhage also exhibited neuron-specific enolase (NSE) ≥ 200 µg/L in 88.5% of cases and lactic dehydrogenase (LDH) ≥ 1000U/L in 73.1% of cases. (3) The incidence of CRAR rate was 100%, and 99.1% (107/108) patients experienced myelosuppression. The incidence of myelosuppression peaked in the third cycle, reaching up to 85.2%. Most children suffered severe myelosuppression existed with bone marrow metastases (76.3%), abnormal VMA (67.5%), and LDH ≥ 1000 U/L (60%). (4) Non-myelosuppressive adverse effects were observed in 75.9% children (82/108), with the highest incidence occurring in the third cycle at 42.6%. (5) Patients who experienced three types of complications had a lower median survival time (MST) of 54.4 months, a 3-year event-free survival (EFS) rate of (44.2 ± 10.7)%, and a 3-year overall survival (OS) rate of (75.8 ± 8.6)%, in comparison to those with only one or two complications, who had a higher MST of 59.5 months, a 3-year EFS rate of (73.5 ± 5.2)% (X2 = 10.457, P = 0.001), and a 3-year OS rate of (84.8 ± 4.1)% (X2 = 10.511, P = 0.001). CONCLUSION: The presence of bone marrow involved and increased VMA were high-risk factors for infection, while NSE ≥ 200 µg/L and LDH ≥ 1000 U/L were high-risk factors for hemorrhage. For those children who had experienced severe myelosuppression, the presence of bone marrow metastases, increased VMA, and LDH ≥ 1000 U/L were their risk factors. The presence of bone involvement was a high-risk factor for children to have non-myelosuppressive adverse effects. Complications that arise during induction chemotherapy could negatively impact the children's prognosis and overall quality of life. WHAT IS KNOWN: • The high-risk neuroblastoma (HR-NB) had a worse prognosis; there was a general international preference for high-intensity chemotherapeutic regimens in the induction phase to these children. WHAT IS NEW: • We analyzed the incidence and risk factors of complications during induction chemotherapy in children with HR-NB and tried to help clinicians predict such complications and adopt optimized therapeutic strategy.


Assuntos
Neoplasias da Medula Óssea , Neuroblastoma , Criança , Humanos , Lactente , Quimioterapia de Indução/efeitos adversos , Incidência , Estudos Retrospectivos , Qualidade de Vida , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Prognóstico , Fatores de Risco , Neoplasias da Medula Óssea/tratamento farmacológico , Hemorragia
6.
Acta Radiol ; 65(7): 841-850, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38659300

RESUMO

BACKGROUND: Fat quantification methods in magnetic resonance imaging (MRI) have been studied to differentiate bone marrow pathologies in adult patients; however, scarce literature is available in pediatric patients. PURPOSE: To evaluate the efficacy of the T1 signal intensity value (T1-SIV), out-of-phase/in-phase signal ratio (OP/IP SR), and fat fraction (FF) to differentiate between normal, benign, and malignant pathological processes. MATERIAL AND METHODS: A total of 48 pediatric patients with lumbar and pelvic MRI were classified into three groups according to bone marrow pathology (group 1, normal; group 2, benign pathology/reconversion; group 3, malignant). The efficacy of T1-SIV, OP/IP SR, and FF values in differentiating these pathologies was evaluated using Kruskal-Wallis or analysis of variance and followed by Bonferroni or Dunn-Bonferroni tests. Cutoff values for malignant infiltration were defined using ROC analysis. RESULTS: Although these values were significantly different in all three groups (P = 0.001-0.008), this difference was not sufficient to discriminate between all groups. Subgroup analyses showed significant differences in T1-SIV between groups 1-3, in OP/IP SR between groups 1-3, 2-3, and 1-2, in FF between groups 1-2 and 1-3 in various regions (P = 0.001-0.049). Cutoff values had a sensitivity and specificity of 90%-100% for OP/IP SR and FF. CONCLUSION: T1-SIV, OP/IP SR, and FF may potentially distinguish normal from pathological bone marrow. OP/IP SR and FF values detected malignant infiltration with high sensitivity and specificity in this study. However, only OP/IP SR may significantly differentiate benign and malignant bone marrow pathologies which needs to be confirmed in the future study with a larger patient population.


Assuntos
Tecido Adiposo , Doenças da Medula Óssea , Medula Óssea , Imageamento por Ressonância Magnética , Humanos , Criança , Imageamento por Ressonância Magnética/métodos , Feminino , Masculino , Diagnóstico Diferencial , Adolescente , Tecido Adiposo/diagnóstico por imagem , Tecido Adiposo/patologia , Pré-Escolar , Doenças da Medula Óssea/diagnóstico por imagem , Medula Óssea/diagnóstico por imagem , Medula Óssea/patologia , Neoplasias da Medula Óssea/diagnóstico por imagem , Sensibilidade e Especificidade , Estudos Retrospectivos , Lactente
7.
Pediatr Radiol ; 54(8): 1395-1398, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38671145

RESUMO

Bone marrow metastases-noted in 6% of patients with rhabdomyosarcoma-have been linked to very poor outcomes. Bilateral bone marrow sampling from iliac crests has been the gold standard for bone marrow examination in rhabdomyosarcoma, but sampling errors due to patchy bone marrow involvement may limit its sensitivity. Here, we report the case of a 6-year-old boy with embryonal rhabdomyosarcoma of the skull base and multiple 2-[18F]fluoro-2-deoxy-D-glucose (2-[18F]FDG)-avid bone marrow metastases visualized by positron emission tomography and computed tomography (2-[18F]FDG PET/CT). His bone marrow aspirates were tumor-free. This case illustrates the diagnostic value of 2-[18F]FDG PET/CT in the detection of bone marrow metastases in rhabdomyosarcoma patients, which may re-shape the definition of bone marrow disease and, ultimately, alter disease staging and risk stratification.


Assuntos
Neoplasias da Medula Óssea , Fluordesoxiglucose F18 , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos , Humanos , Masculino , Criança , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Neoplasias da Medula Óssea/diagnóstico por imagem , Neoplasias da Medula Óssea/patologia , Rabdomiossarcoma/diagnóstico por imagem , Rabdomiossarcoma/patologia
8.
Zhonghua Zhong Liu Za Zhi ; 46(5): 419-427, 2024 May 23.
Artigo em Chinês | MEDLINE | ID: mdl-38742355

RESUMO

Objective: To investigate the detection of bone marrow tumor cells in small cell lung cancer (SCLC) patients and their relationship with clinical features, treatment response and prognosis. Methods: A total of 113patients with newly diagnosed SCLC from January 2018 to October 2022 at Beijing Chest Hospital were prospectively enrolled. Before treatment, bone marrow was aspirated and separately submitted for tumor cells detection by liquid-based cytology and disseminated tumor cells (DTCs) detection by the substrction enrichment and immunostaining fluorescence in situ hybridization (SE-iFISH) platform. The correlation between the detection results of the two methods with patients' clinical features and treatment response was evaluated by Chi-square. Kaplan-Meier method was applied to create survival curves and the Cox regression model was used for multivariate analysis. Results: The positive rate of bone marrow liquid-based cytology in SCLC was 15.93% (18/113). The liver and bone metastases rates were significantly higher (55.56% vs 11.58% for liver metastasis, P<0.001; 77.78% vs 16.84% for bone metastasis, P<0.001) and thrombocytopenia was more common (16.67% vs 2.11%, P=0.033) in patients with tumor cells detected in liquid-based cytology than those without detected tumor cells. As for SE-iFISH, DTCs were detected in 92.92% of patients (105/113), the liver and bone metastasis rates were significantly higher (37.93% vs 11.90% for liver metastasis, P=0.002; 44.83% vs 20.23 % for bone metastasis, P=0.010), and the incidence of thrombocytopenia was significantly increased (13.79% vs 1.19%, P=0.020) in patients with DTCs≥111 per 3 ml than those with DTCs<111 per 3 ml. The positive rates of bone marrow liquid-based cytology in the disease control group and the disease progression group were 12.00% (12/100) and 46.15% (6/13), respectively, and the difference was statistically significant (P=0.002). However, the result of SE-iFISH revealed the DTCs quantities of the above two groups were 29 (8,110) and 64 (15,257) per 3 ml, and there was no statistical difference between the two groups (P=0.329). Univariate analysis depicted that the median progression-free survival (PFS) and median overall survival (OS) of liquid-based cytology positive patients were significantly shorter than those of tumor cell negative patients (6.33 months vs 9.27 months for PFS, P=0.019; 8.03 months vs 19.50 months for OS, P=0.019, P=0.033). The median PFS and median OS in patients with DTCs≥111 per 3 ml decreased significantly than those with DTCs<111 per 3 ml (6.83 months vs 9.50 months for PFS, P=0.004; 11.2 months vs 20.60 months for OS, P=0.019). Multivariate analysis showed that disease stage (HR=2.806, 95%CI:1.499-5.251, P=0.001) and DTCs quantity detected by SE-iFISH (HR=1.841, 95%CI:1.095-3.095, P=0.021) were independent factors of PFS, while disease stage was the independent factor of OS (HR=2.538, 95%CI:1.169-5.512, P=0.019). Conclusions: Both bone marrow liquid-based cytology and SE-iFISH are clinically feasible. The positive detection of liquid-based cytology or DTCs≥111 per 3 ml was correlated with distant metastasis, and DTCs≥111 per 3 ml was an independent prognostic factor of decreased PFS in SCLC.


Assuntos
Medula Óssea , Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Humanos , Carcinoma de Pequenas Células do Pulmão/patologia , Neoplasias Pulmonares/patologia , Prognóstico , Medula Óssea/patologia , Estudos Prospectivos , Feminino , Masculino , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Neoplasias Ósseas/secundário , Pessoa de Meia-Idade , Neoplasias da Medula Óssea/secundário , Taxa de Sobrevida , Células da Medula Óssea , Idoso , Trombocitopenia , Modelos de Riscos Proporcionais , Estimativa de Kaplan-Meier , Relevância Clínica
9.
Br J Haematol ; 200(5): 643-651, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36382360

RESUMO

Nestin is an intermediate filament protein, which was originally detected in neuroepithelial stem cells. Besides its use as a phenotypic marker of mesenchymal stem cells in the hematopoeitic stem cell niche, the functional interpretation of nestin+ cells remains elusive. We investigated the cellular expression of nestin in bone marrow trephine biopsies of MPN patients, following myeloablation at a stage of hypocellularity during early regeneration. Here, nestin is highly expressed in mature osteocytes, arteriolar endothelial and perivascular cells and small capillaries within the bone marrow space, but not in sinusoid lining cells. This is in stark contrast to nestin expression pattern in myeloproliferative neoplasms that show hypercellularity due to oncogenic driver mutations. Here, nestin is expressed exclusively in endothelial cells of arterioles, but not in osteocytes or small capillaries. Thus, the pattern of nestin expression following myeloablation inversely correlates with cellularity in the bone marrow. This nestin expression pattern is mimicking early postnatal transcriptional programming during bone marrow development. We show that nestin expression in osteocytes occurs across different species following transplant and also in bone marrow metastasis.


Assuntos
Neoplasias da Medula Óssea , Medula Óssea , Humanos , Nestina/genética , Nestina/metabolismo , Medula Óssea/metabolismo , Células Endoteliais/metabolismo , Osteócitos/metabolismo , Neoplasias da Medula Óssea/metabolismo
10.
J Transl Med ; 21(1): 399, 2023 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-37337244

RESUMO

BACKGROUND: Bone marrow metastasis (BMM) is underestimated in gastric cancer (GC). GC with BMM frequently complicate critical hematological abnormalities like diffused intravascular coagulation and microangiopathic hemolytic anemia, which constitute a highly aggressive GC (HAGC) subtype. HAGC present a very poor prognosis with peculiar clinical and pathological features when compared with not otherwise specified advanced GC (NAGC). But the molecular mechanisms underlying BMM from GC remain rudimentary. METHODS: The transcriptomic difference between HAGC and NAGC were analyzed. Genes that were specifically upregulated in HAGC were identified, and their effect on cell migration and invasion was studied. The function of ACTN2 gene were confirmed by GC cell lines, bone-metastatic animal model and patients' tissues. Furthermore, the molecular mechanism of ACTN2 derived-BMM was explored by multiple immunofluorescence staining, western blot, chromatin immunoprecipitation, and luciferase reporter assays. RESULTS: We elucidated the key mechanisms of BMM depending on the transcriptomic difference between HAGC and NAGC. Five genes specifically upregulated in HAGC were assessed their effect on cell migration and invasion. The ACTN2 gene encoding protein α-Actinin-2 was detected enhanced the metastatic capability and induced BMM of GC cells in mouse models. Mechanically, α-Actinin-2 was involved in filopodia formation where it promoted the Actin filament cross-linking by replacing α-Actinin-1 to form α-Actinin-2:α-Actinin-4 complexes in GC cells. Moreover, NF-κB subunit RelA and α-Actinin-2 formed heterotrimers in the nuclei of GC cells. As a direct target of RelA:α-Actinin-2 heterotrimers, the ACTN2 gene was a positive auto-regulatory loop for α-Actinin-2 expression. CONCLUSIONS: We demonstrated a link between filopodia, BMM and ACTN2 activation, where a feedforward activation loop between ACTN2 and RelA is established via actin in response to distant metastasis. Given the novel filopodia formation function and the new mechanism of BMM in GC, we propose ACTN2 as a druggable molecular vulnerability that may provide potential therapeutic benefit against BMM of GC.


Assuntos
Actinina , Neoplasias da Medula Óssea , Neoplasias Gástricas , Animais , Camundongos , Actinina/genética , Actinina/metabolismo , Linhagem Celular Tumoral , NF-kappa B/metabolismo , Pseudópodes/metabolismo , Pseudópodes/patologia , Neoplasias Gástricas/patologia
11.
Haematologica ; 108(5): 1359-1373, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36005555

RESUMO

Apart from the central role of the activated JAK/STAT signaling pathway, ASXL1 mutations are the most recurrent additional mutations in myeloproliferative neoplasms and occur much more commonly in myelofibrosis than in essential thrombocythemia and polycythemia vera. However, the mechanism of the association with ASXL1 mutations and bone marrow fibrosis remains unknown. Here, integrating our own data from patients with myeloproliferative neoplasms and a hematopoietic-specific Asxl1 deletion/Jak2V617F mouse model, we show that ASXL1 mutations are associated with advanced myeloproliferative neoplasm phenotypes and onset of myelofibrosis. ASXL1 mutations induce skewed monocyte/macrophage and neoplastic monocyte-derived fibrocyte differentiation, consequently they enhance inflammation and bone marrow fibrosis. Consistently, the loss of ASXL1 and JAK2V617F mutations in hematopoietic stem and progenitor cells leads to enhanced activation of polycomb group target genes, such as EGR1. The upregulation of EGR1, in turn, accounts for increased hematopoietic stem and progenitor cell commitment to the monocyte/macrophage lineage. Moreover, EGR1 induces the activation of TNFA and thereby further drives the differentiation of monocytes to fibrocytes. Accordingly, combined treatment with a TNFR antagonist and ruxolitinib significantly reduces fibrocyte production in vitro. Altogether, these findings demonstrate that ASXL1 mutations accelerate fibrocyte production and inflammation in myeloproliferative neoplasms via the EGR1-TNFA axis, explaining the cellular and molecular basis for bone marrow fibrosis and the proof-ofconcept for anti-fibrosis treatment.


Assuntos
Neoplasias da Medula Óssea , Transtornos Mieloproliferativos , Policitemia Vera , Mielofibrose Primária , Animais , Camundongos , Proteína 1 de Resposta de Crescimento Precoce/genética , Janus Quinase 2/genética , Mutação , Transtornos Mieloproliferativos/genética , Policitemia Vera/genética , Mielofibrose Primária/genética , Proteínas Repressoras/genética
12.
Ann Hematol ; 102(7): 1745-1759, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37233774

RESUMO

The classic BCR-ABL1-negative myeloproliferative neoplasm (MPN) is a highly heterogeneous hematologic tumor that includes three subtypes, namely polycythemia vera (PV), essential thrombocytosis (ET), and primary myelofibrosis (PMF). Despite having the same JAK2V617F mutation, the clinical manifestations of these three subtypes of MPN differ significantly, which suggests that the bone marrow (BM) immune microenvironment may also play an important role. In recent years, several studies have shown that peripheral blood monocytes play an important role in promoting MPN. However, to date, the role of BM monocytes/macrophages in MPN and their transcriptomic alterations remain incompletely understood. The purpose of this study was to clarify the role of BM monocytes/macrophages in MPN patients with the JAK2V617F mutation. MPN patients with the JAK2V617F mutation were enrolled in this study. We investigated the roles of monocytes/macrophages in the BM of MPN patients, using flow cytometry, monocyte/macrophage enrichment sorting, cytospins and Giemsa-Wright staining, and RNA-seq. Pearson correlation coefficient analysis was also used to detect the correlation between BM monocytes/macrophages and the MPN phenotype. In the present study, the proportion of CD163+ monocytes/macrophages increased significantly in all three subtypes of MPN. Interestingly, the percentages of CD163+ monocytes/macrophages are positively correlated with HGB in PV patients and PLT in ET patients. In contrast, the percentages of CD163+ monocytes/macrophages are negatively correlated with HGB and PLT in PMF patients. It was also found that CD14+CD16+ monocytes/macrophages increased and correlated with MPN clinical phenotypes. RNA-seq analyses demonstrated that the transcriptional expressions of monocytes/macrophages in MPN patients are relatively distinct. Gene expression profiles of BM monocytes/macrophages suggest a specialized function in support of megakaryopoiesis in ET patients. In contrast, BM monocytes/macrophages yielded a heterogeneous status in the support or inhibition of erythropoiesis. Significantly, BM monocytes/macrophages shaped an inflammatory microenvironment, which, in turn, promotes myelofibrosis. Thus, we characterized the roles of increased monocytes/macrophages in the occurrence and progression of MPNs. Our findings of the comprehensive transcriptomic characterization of BM monocytes/macrophages provide important resources to serve as a basis for future studies and future targets for the treatment of MPN patients.


Assuntos
Neoplasias da Medula Óssea , Transtornos Mieloproliferativos , Policitemia Vera , Trombocitemia Essencial , Humanos , Medula Óssea/patologia , Monócitos/patologia , Transtornos Mieloproliferativos/genética , Policitemia Vera/genética , Mutação , Neoplasias da Medula Óssea/patologia , Trombocitemia Essencial/genética , Janus Quinase 2/genética , Microambiente Tumoral
13.
Am J Hematol ; 98(7): 1029-1042, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37203407

RESUMO

Small molecule inhibitors targeting JAK2 provide symptomatic benefits for myeloproliferative neoplasm (MPN) patients and are among first-line therapeutic agents. However, despite all having potent capacity to suppress JAK-STAT signaling, they demonstrate distinct clinical profiles suggesting contributory effects in targeting other ancillary pathways. Here, we performed comprehensive profiling on four JAK2 inhibitors either FDA-approved (ruxolitinib, fedratinib, and pacritinib) or undergoing phase 3 studies (momelotinib) to better outline mechanistic and therapeutic efficacy. Across JAK2-mutant in vitro models, all four inhibitors demonstrated similar anti-proliferative phenotypes, whereas pacritinib yielded greatest potency on suppressing colony formation in primary samples, while momelotinib exhibited unique erythroid colony formation sparing. All inhibitors reduced leukemic engraftment, disease burden, and extended survival across patient-derived xenograft (PDX) models, with strongest effects elicited by pacritinib. Through RNA-sequencing and gene set enrichment analyses, differential suppressive degrees of JAK-STAT and inflammatory response signatures were revealed, which we validated with signaling and cytokine suspension mass cytometry across primary samples. Lastly, we assessed the capacity of JAK2 inhibitors to modulate iron regulation, uncovering potent suppression of hepcidin and SMAD signaling by pacritinib. These comparative findings provide insight into the differential and beneficial effects of ancillary targeting beyond JAK2 and may help guide the use of specific inhibitors in personalized therapy.


Assuntos
Neoplasias da Medula Óssea , Inibidores de Janus Quinases , Transtornos Mieloproliferativos , Humanos , Inibidores de Janus Quinases/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Transtornos Mieloproliferativos/genética , Janus Quinase 2/genética
14.
Am J Hematol ; 98(5): 794-800, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36869873

RESUMO

Myeloproliferative neoplasms (MPNs) are the most common etiologies of primary splanchnic vein thrombosis, present in almost forty percent of patients with Budd-Chiari syndrome or portal vein thrombosis. Diagnosis of MPNs can be difficult in these patients because key characteristics, such as elevated blood cell counts and splenomegaly, are confounded by portal hypertension or bleeding complications. In recent years, diagnostic tools have improved to provide more accurate diagnosis and classification of MPNs. Although bone marrow biopsy findings remain a major diagnostic criterion, molecular markers are playing an increasing role not only in diagnosis but also in better estimating prognosis. Therefore, though screening for JAK2V617F mutation should be the starting point of the diagnostic workup performed in all patients with splanchnic vein thrombosis, a multidisciplinary approach is needed to accurately diagnose the subtype of myeloproliferative neoplasm, recommend the useful additional tests (bone marrow biopsy, search for an additional mutation using targeted next-generation sequencing), and suggest the best treatment strategy. Indeed, providing a specific expert care pathway for patients with splanchnic vein thrombosis and underlying myeloproliferative neoplasm is crucial to determine the optimal management to reduce the risk of both hematological and hepatic complications.


Assuntos
Neoplasias da Medula Óssea , Síndrome de Budd-Chiari , Transtornos Mieloproliferativos , Trombose Venosa , Humanos , Veia Porta/patologia , Trombose Venosa/diagnóstico , Trombose Venosa/etiologia , Trombose Venosa/terapia , Síndrome de Budd-Chiari/diagnóstico , Síndrome de Budd-Chiari/etiologia , Síndrome de Budd-Chiari/terapia , Transtornos Mieloproliferativos/complicações , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/terapia , Circulação Esplâncnica , Janus Quinase 2/genética
15.
Pediatr Blood Cancer ; 70(5): e30261, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36815592

RESUMO

BACKGROUND: Risk stratification of high-risk neuroblastoma (NB) is crucial for exploring treatments. This study aimed to explore the value of minimal residual disease (MRD) based on PHOX2B levels for further risk stratification in high-risk NB. METHODS: The expression of PHOX2B was monitored at two time points (after two and six cycles of induction chemotherapy, TP1 and TP2, respectively) by real-time polymerase chain reaction (RT-PCR). The clinical characteristics between groups and survival rates were analyzed. RESULTS: The study included 151 high-risk patients. Positive expression of PHOX2B at diagnosis was seen in 129 cases. PHOX2B was mainly expressed in patients with high lactate dehydrogenase (LDH) and neuron-specific enolase (NSE) levels (p < .001), bone marrow metastasis (p < .001), more than three metastatic organs (p < .001), 11q23 loss of heterozygosity (LOH) (p = .007), and when more events occurred (p = .012). The 4-year EFS rate was significantly lower in patients with positive PHOX2B expression compared to the negative group at diagnosis (32.9% ± 6.2% vs. 74.5% ± 10.1%, p = .005). We stratified the 151 patients into three MRD risk groups: low high-risk (low-HR), with TP1 less than 10-4 and TP2 less than 10-4 ; ultra-HR, with TP1 greater than or equal to 10-2 or TP2 greater than or equal to 10-4 , and others classified as intermediate-HR. Patients in ultra-HR had the worst survival rate compared with other two groups (p = .02). In a multivariate model, MRD risk stratification based on PHOX2B levels at TP1 and TP2 was an independent prognostic factor for high-risk patients (p = .001). Patients in ultra-HR were associated with 11q23 LOH (p < .001), more than three organs of metastasis (p = .005), bone marrow metastasis (p < .001), and occurrence of more events (p = .009). CONCLUSIONS: MRD risk stratification based on PHOX2B levels at two time points (after two and six cycles of induction chemotherapy) provided a stratification system for high-risk NB, which successfully predicted treatment outcomes. Our results present an effective method for further stratification of high-risk NB.


Assuntos
Neoplasias da Medula Óssea , Neuroblastoma , Humanos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/análise , Neoplasia Residual/diagnóstico , Neuroblastoma/genética , Neuroblastoma/terapia , Neuroblastoma/diagnóstico , Prognóstico , Medição de Risco , Fatores de Transcrição/genética , Fatores de Transcrição/análise , Resultado do Tratamento
16.
Clin Radiol ; 78(7): e535-e541, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36997373

RESUMO

AIM: To evaluate the diagnostic accuracy of whole-body (WB) magnetic resonance imaging (MRI) utilising three-dimensional (3D) short tau inversion recovery (STIR) and T1-weighted in/opposed-phase MRI in the detection of neuroblastoma bone marrow metastasis compared to 2-[18F]-fluoro-2-deoxy-d-glucose (FDG) positron-emission tomography (PET)/computed tomography (CT). MATERIALS AND METHODS: A prospective study of 20 consecutive histopathologically proven neuroblastoma patients enrolled in this study from January 2021 to August 2022. WB MRI and FDG-PET/CT were performed for all cases. Bone marrow biopsy served as the standard of reference. Sensitivity, specificity, negative predictive value (NPV), positive predictive value (PPV), and accuracy were calculated. In addition, lesion-by-lesion analysis was performed and the number of bone marrow metastatic lesions in different body segments using both imaging methods was recorded and compared. RESULTS: WB MRI correctly identified true positives and true negatives in all cases with a sensitivity and specificity of 100%. In contrast, FDG-PET/CT showed two false-negative cases that resulted in a sensitivity, specificity, PPV, NPV, and accuracy of 86.7%, 100%, 100%, 71.4%, and 92%, respectively. In the lesion-by-lesion analysis, WB MRI detected more (24.3%) bone marrow metastatic lesions than FDG-PET/CT. CONCLUSION: Whole-body MRI can reliably identify neuroblastoma bone marrow infiltration, and could be an alternative to PET/CT in that regard.


Assuntos
Neoplasias da Medula Óssea , Neoplasias Ósseas , Neuroblastoma , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Medula Óssea/diagnóstico por imagem , Medula Óssea/patologia , Fluordesoxiglucose F18 , Estudos Prospectivos , Imageamento por Ressonância Magnética/métodos , Neoplasias Ósseas/secundário , Sensibilidade e Especificidade , Imagem Corporal Total/métodos , Biópsia , Neuroblastoma/diagnóstico por imagem , Neuroblastoma/patologia , Padrões de Referência , Neoplasias da Medula Óssea/diagnóstico por imagem , Tomografia por Emissão de Pósitrons
17.
Skeletal Radiol ; 52(6): 1179-1192, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36441237

RESUMO

OBJECTIVE: To compare the diagnostic accuracy of diffusion-weighted (DW)-MRI with b-values of 50 s/mm2 and 800 s/mm2 for the detection of bone marrow metastases in children and young adults with solid malignancies. METHODS: In an institutional review board-approved prospective study, we performed 51 whole-body DW-MRI scans in 19 children and young adults (14 males, 5 females; age range: 1-25 years) with metastasized cancers before (n = 19 scans) and after (n = 32 scans) chemotherapy. Two readers determined the presence of focal bone marrow lesions in 10 anatomical areas. A third reader measured ADC and SNR of focal lesions and normal marrow. Simultaneously acquired 18F-FDG-PET scans served as the standard of reference. Data of b = 50 s/mm2 and 800 s/mm2 images were compared with the Wilcoxon signed-rank test. Inter-reader agreement was evaluated with weighted kappa statistics. RESULTS: The SNR of bone marrow metastases was significantly higher compared to normal bone marrow on b = 50 s/mm2 (mean ± SD: 978.436 ± 1239.436 vs. 108.881 ± 109.813, p < 0.001) and b = 800 s/mm2 DW-MRI (499.638 ± 612.721 vs. 86.280 ± 89.120; p < 0.001). On 30 out of 32 post-treatment DW-MRI scans, reconverted marrow demonstrated low signal with low ADC values (0.385 × 10-3 ± 0.168 × 10-3mm2/s). The same number of metastases (556/588; 94.6%; p > 0.99) was detected on b = 50 s/mm2 and 800 s/mm2 images. However, both normal marrow and metastases exhibited low signals on ADC maps, limiting the ability to delineate metastases. The inter-reader agreement was substantial, with a weighted kappa of 0.783 and 0.778, respectively. CONCLUSION: Bone marrow metastases in children and young adults can be equally well detected on b = 50 s/mm2 and 800 s/mm2 images, but ADC values can be misleading.


Assuntos
Neoplasias da Medula Óssea , Neoplasias Ósseas , Masculino , Feminino , Humanos , Adulto Jovem , Criança , Lactente , Pré-Escolar , Adolescente , Adulto , Imagem de Difusão por Ressonância Magnética/métodos , Medula Óssea/diagnóstico por imagem , Medula Óssea/patologia , Estudos Prospectivos , Neoplasias Ósseas/patologia , Neoplasias da Medula Óssea/diagnóstico por imagem
18.
Zhongguo Dang Dai Er Ke Za Zhi ; 25(5): 476-482, 2023 May 15.
Artigo em Chinês | MEDLINE | ID: mdl-37272173

RESUMO

OBJECTIVES: To investigate the effectiveness of high-dose chemotherapy combined with autologous hematopoietic stem cell transplantation (ASCT) in the treatment of children with high-risk neuroblastoma (NB). METHODS: A retrospective analysis was performed on 29 children with high-risk NB who were admitted to Shanghai Children's Hospital and were treated with high-dose chemotherapy combined with ASCT from January 2013 to December 2021, and their clinical features and prognosis were analyzed. RESULTS: Among the 29 children treated by high-dose chemotherapy combined with ASCT, there were 18 boys (62%) and 11 girls (38%), with a median age of onset of 36 (27, 59) months. According to the International Neuroblastoma Staging System, 6 children (21%) had stage III NB and 23 children (79%) had stage IV NB, and the common metastatic sites at initial diagnosis were bone in 22 children (76%), bone marrow in 21 children (72%), and intracalvarium in 4 children (14%). All 29 children achieved reconstruction of hematopoietic function after ASCT. After being followed up for a median time of 25 (17, 45) months, 21 children (72%) had continuous complete remission and 8 (28%) experienced recurrence. The 3-year overall survival rate and event-free survival rate were 68.9%±16.1% and 61.4%±14.4%, respectively. Presence of bone marrow metastasis, neuron-specific enolase ≥370 ng/mL and positive bone marrow immunophenotyping might reduce the 3-year event-free survival rate (P<0.05). CONCLUSIONS: Children with high-risk NB who have bone marrow metastasis at initial diagnosis tend to have a poor prognosis. ASCT combined with high-dose chemotherapy can effectively improve the prognosis of children with NB with a favorable safety profile.


Assuntos
Neoplasias da Medula Óssea , Transplante de Células-Tronco Hematopoéticas , Neuroblastoma , Pré-Escolar , Feminino , Humanos , Masculino , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Medula Óssea/tratamento farmacológico , China , Terapia Combinada , Intervalo Livre de Doença , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Prognóstico , Estudos Retrospectivos , Transplante de Células-Tronco , Transplante Autólogo
20.
Anticancer Drugs ; 33(7): 686-690, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35324515

RESUMO

Thymic lymphoepithelioma-like carcinoma (LELC) is a rare primary malignant neoplasm originating from the thymus. Thymic LELC diagnosis is often terminal when diagnosed, some patients have lost the opportunity for surgery. Platinum- and anthracycline-based systemic chemotherapy are the first-line treatment plan; however, there is no clear consensus on therapy when first-line treatment fails because of the lack of cases of advanced thymic LELC. Here was a rare case of advanced thymic LELC with bone marrow metastasis at relapse, which is reported in a patient who responded well to toripalimab combined with anlotinib therapy. The treatment showed tolerable toxicity with good antitumor activity in the patient. As far as we know, this is the first case that the combination of toripalimab with anlotinib is effective in controlling advanced thymic LELC with bone marrow metastasis. The case reports represent an essential means by which an effective therapy for advanced thymic LELC may not be practical given the low frequency of a thymic LELC with multiple metastases.


Assuntos
Neoplasias da Medula Óssea , Neoplasias Ósseas , Carcinoma de Células Escamosas , Humanos , Imunoterapia , Platina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA