Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Oncologist ; 29(1): e90-e96, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37616529

RESUMEN

BACKGROUND: Natural killer/T-cell lymphoma (NKTCL) is a rare and heterogeneous tumor type of non-Hodgkin's lymphoma (NHL) with a poor clinical outcome. There is no standardized salvage treatment failing l-asparaginase-based regimens. Here we report our retrospective results of the combined use of selinexor and PD-1 blockade (tislelizumab) in 5 patients with NKTCL who had exhausted almost all available treatments. PATIENTS AND METHODS: A total of 5 patients with relapsed/refractory(R/R) NK/T-cell lymphomas failing prior l-asparaginase and anti-PD-1 antibody were retrospectively collected. They were treated with at least one cycle of XPO1 inhibitor plus the same anti-PD-1 antibody. Anti-PD-1 antibody (Tislelizumab) was administrated at 200 mg on day 1 every 3 weeks and selinexor doses and schedules ranged from 40 mg weekly for 2 weeks per 21-day cycle to 60 mg weekly per cycle. RESULTS: Five patients with relapsed NKTCL with extensive organ involvement including 4 central nervous system (CNS) infiltration patients were included. Four patients achieved objective responses including 3 complete responses (CR) and 1 partial response (PR). After a median follow-up time of 14.5 (range, 5-22) months, 1 patient was still in remission with CR, and the other 4 patients discontinued due to disease progression with a median progression-free survival (PFS) of 6 months and median overall survival (OS) of 12 months. Four patients with CNS involvement achieved a median OS of 8 months. Our data suggest that selinexor in combination with an anti-PD-1 antibody is a promising small molecule and immunotherapy combination regimen for patients with relapsed or refractory NKTCL.


Asunto(s)
Linfoma de Células T , Linfoma , Humanos , Asparaginasa/uso terapéutico , Estudios Retrospectivos , Receptor de Muerte Celular Programada 1/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Linfoma de Células T/tratamiento farmacológico , Células Asesinas Naturales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico
2.
Int J Cancer ; 151(5): 752-763, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35489026

RESUMEN

Optimal treatment strategies for natural killer/T-cell lymphoma (NKTCL) patients with stage IV disease have not been well defined. In this prospective phase 2 study, we evaluated the treatment using MEDA (methotrexate, etoposide, dexamethasone and pegaspargase) as induction chemotherapy and autologous hematopoietic stem cell transplantation (Auto-HSCT) for consolidation. Patients with stage IV disease without prior L-asparaginase-based chemotherapy were eligible. Four cycles of MEDA were administered as induction treatment. Patients with complete response (CR, necessary to have complete metabolic remission of PET/CT, negative plasma EBV-DNA and negative EBER staining of bone marrow biopsy tissue) were consolidated by Auto-HSCT. A total of 53 patients were enrolled. The overall response (OR) rate and CR rate after four cycles of MEDA chemotherapy were 75.5% and 56.6%, respectively. Among them, 25 patients underwent Auto-HSCT. The 4-year overall survival (OS) rate and progression-free survival (PFS) rate were 58.0% (95% CI, 43.4%-70.0%) and 43.4% (95% CI, 29.9%-56.1%), respectively. Patients who underwent Auto-HSCT had a 4-year OS rate of 92.0% (95% CI, 71.6%-97.9%) and a 4-year PFS rate of 80.0% (95% CI, 58.4%-91.1%). Grade 3/4 neutropenia and thrombocytopenia occurred in 28.3% and 17.0% of the patients, respectively. MEDA chemotherapy is an effective induction regimen with reduced grade 3/4 hematological toxicities for stage IV NKTCL. Consolidation with Auto-HSCT can be considered as a potential approach to improve the long-term survival of CR patients after induction treatment.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Tomografía Computarizada por Tomografía de Emisión de Positrones , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Supervivencia sin Enfermedad , Humanos , Estudios Prospectivos , Estudios Retrospectivos , Trasplante Autólogo , Resultado del Tratamiento
3.
Br J Haematol ; 196(4): 939-946, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34806163

RESUMEN

Early-stage natural killer/T-cell lymphoma (NK/TCL) patients usually receive a combination of chemotherapy and radiotherapy, but the optimal treatment approach has not yet been established. This study aimed to investigate the efficacy and safety profile of a novel chemotherapy regimen and sandwiched radiotherapy in early-stage NK/TCL. Patients with newly diagnosed stage IE/IIE disease were eligible. Patients were initially treated with two courses of the GELAD regimen (gemcitabine 1·0 g/m2 day 1, etoposide 60 mg/m2 days 1-3, pegaspargase 2000 units/m2 day 4, and dexamethasone 40 mg days 1-4), followed by intensity-modulated radiotherapy (IMRT; 50-56 Gy in 25-28 fractions) and two additional courses of GELAD chemotherapy. A total of 52 patients were enrolled. The overall response rate and complete response rate per Lugano 2014 criteria were 94·2% and 92·3% respectively. With a median follow-up of 32 months, the estimated four-year overall survival rate and progression-free survival rate were 94·2% [95% confidence interval (CI), 83·2% to 93·1%] and 90·4% (95% CI, 78·4% to 95·9%) respectively. The most common adverse events were related to pegaspargase. Haematological toxicities were mild, with grade 3/4 neutropenia in 15·4% of patients. Our study provides a new approach with high activity and improved safety for the treatment of early-stage NK/TCL patients. This study was registered at www.clinicaltrials.gov as NCT02733458.


Asunto(s)
Linfoma Extranodal de Células NK-T/tratamiento farmacológico , Linfoma Extranodal de Células NK-T/radioterapia , Adolescente , Adulto , Anciano , Femenino , Humanos , Linfoma Extranodal de Células NK-T/mortalidad , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Supervivencia sin Progresión , Estudios Prospectivos , Resultado del Tratamiento , Adulto Joven
4.
Cancer Immunol Immunother ; 71(9): 2197-2212, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35092480

RESUMEN

Cell-released nanovesicles can induce anti-leukemia immunity. Leukemia cell-derived exosomes (LEXs) are promising anti-tumor vaccine components for cancer immunotherapy. Nonetheless, LEX-based vaccines show modest potency in vivo, likely due to the presence of immunosuppressive PD-L1 proteins in the exosomes. We hypothesized that targeting exosomal PD-L1 could optimize LEX-based vaccines. To test this hypothesis, we compared the capacity of exosomes derived from PD-L1-silenced acute lymphocytic leukemia-derived leukemia cells (LEXPD-L1si) and non-modified exosomes to induce anti-leukemia immunity. Lentivirus-mediated PD-L1 shRNA was used to downregulate PD-L1 expression in parental leukemia cells and LEXs. LEXPD-L1si were characterized by electron microscopy, Western blotting, nanoparticle tracking analysis and flow cytometry, and their anti-leukemia immune effects were tested on immune cells and in animal models. In the present study, lentivirus-mediated PD-L1 shRNA successfully downregulated PD-L1 expression in parental leukemia cells and in LEXs. LEXPD-L1si induced better DC maturation and subsequently enhanced T cell activation, as compared with non-modified LEXs. Consistently, immunization with LEXPD-L1si induced greater T cell proliferation and Th1 cytokine release. LEXPD-L1si was a more potent inducer of antigen-specific cytotoxic lymphocyte (CTL) response. Finally, we vaccinated DBA/2 mice with exosome formulations to test their ability to induce both protective and therapeutic anti-tumor CTL responses in vivo. Vaccination with LEXPD-L1si strongly inhibited tumor growth and prolonged survival of immunized mice. Downregulation of exosomal PD-L1 expression in LEXs effectively induces more potent anti-leukemia immunity. Therefore, our strategy for optimizing LEX-based vaccine has a potential application in leukemia immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Exosomas , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Células Dendríticas , Regulación hacia Abajo , Exosomas/metabolismo , Ratones , Ratones Endogámicos DBA , ARN Interferente Pequeño/genética
5.
Biochem Biophys Res Commun ; 547: 162-168, 2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33610916

RESUMEN

Although acute myeloid leukemia (AML) is a highly heterogeneous disease with diverse genetic subsets, one hallmark of AML blasts is myeloid differentiation blockade. Extensive evidence has indicated that differentiation induction therapy represents a promising treatment strategy. Here, we identified that the pharmacological inhibition of the mitochondrial electron transport chain (ETC) complex III by antimycin A inhibits proliferation and promotes cellular differentiation of AML cells. Mechanistically, we showed that the inhibition of dihydroorotate dehydrogenase (DHODH), a rate-limiting enzyme in de novo pyrimidine biosynthesis, is involved in antimycin A-induced differentiation. The activity of antimycin A could be reversed by supplement of excessive amounts of exogenous uridine as well as orotic acid, the product of DHODH. Furthermore, we also found that complex III inhibition exerts a synergistic effect in differentiation induction combined with DHODH inhibitor brequinar as well as with the pyrimidine salvage pathway inhibitor dipyridamole. Collectively, our study uncovered the link between mitochondrial complex III and AML differentiation and may provide further insight into the potential application of mitochondrial complex III inhibitor as a mono or combination treatment in differentiation therapy of AML.


Asunto(s)
Antimicina A/análogos & derivados , Compuestos de Bifenilo/farmacología , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Antimicina A/farmacología , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dihidroorotato Deshidrogenasa , Complejo III de Transporte de Electrones/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo
6.
J Cell Mol Med ; 24(2): 1684-1699, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31782262

RESUMEN

Allogeneic haematopoietic stem cell transplantation (allo-HSCT) is the only curative method in treating haematologic malignant diseases. Graft-versus-host disease (GVHD) is a common complication post-allo-HSCT, which can be life-threatening. Mesenchymal stem cells (MSCs) as an adult stem cell with immunoregulatory function have demonstrated efficacy in steroid resistant acute GVHD (aGVHD). However, the outcome of aGVHD treated with MSCs in clinical trials varied and its underlying mechanism is still unclear. TGF-ß1 is a potent cytokine, which plays a key role in immunoregulation. In the present study, we firstly transduced the lentivirus vector containing TGF-ß1 gene with mouse bone marrow-derived MSCs. Then, we investigated the immunosuppressive effect of TGF-ß1 gene-modified MSCs on lymphocytes in vitro and its preventive and therapeutical effects on murine aGVHD model in vivo. Murine MSC was successfully isolated and identified. TGF-ß1 was efficiently transduced into mouse MSCs, and high level TGF-ß1 was detected. MSC-TGF-ß1 shared the same morphology and immunotypic features of normal MSC. In vitro, MSC-TGF-ß1 showed enhanced immunosuppressive function on lymphocyte proliferation. In vivo, MSC-TGF-ß1 showed enhanced amelioration on the severity of aGVHD both in prophylactic and therapeutic murine models. Finally, the macrophages (MØs) derived from MSC-TGF-ß1-treated mice showed a remarkably increasing of anti-inflammatory M2-like phenotype. Furthermore, the differentiation of CD4+ CD25+ Foxp3+ Treg cells was significantly increased in MSC-TGF-ß1-treated group. Taken together, we proved that MSC-TGF-ß1 showed enhanced alleviation of aGVHD severity in mice by skewing macrophages into a M2 like phenotype or increasing the proportion of Treg cells, which opens a new frontier in the treatment of aGVHD.


Asunto(s)
Diferenciación Celular , Enfermedad Injerto contra Huésped/inmunología , Macrófagos/patología , Células Madre Mesenquimatosas/metabolismo , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Enfermedad Aguda , Animales , Médula Ósea/patología , Polaridad Celular , Forma de la Célula , Modelos Animales de Enfermedad , Femenino , Enfermedad Injerto contra Huésped/patología , Proteínas Fluorescentes Verdes/metabolismo , Inmunofenotipificación , Terapia de Inmunosupresión , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Análisis de Supervivencia
7.
Acta Haematol ; 141(3): 164-175, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30808826

RESUMEN

BACKGROUND: The preferred type of postremission therapy (PRT) for intermediate-risk acute myeloid leukemia (AML) in first complete remission (CR1) is a subject of continued debate. Although allogeneic stem cell transplantation (alloSCT) is regarded as a curative strategy for AML, the efficacy of autologous stem cell transplantation (autoSCT) for patients without a matched sibling donor (MSD) has remained controversial. METHODS: To compare survival outcomes after alloSCT versus autoSCT for patients with intermediate-risk AML in CR1, we performed a meta-analysis of 11 clinical studies. The outcomes included relapse-free survival (RFS), overall survival (OS), relapse rate (RR), and treatment-related mortality (TRM). RESULTS: Compared with autoSCT, alloSCT showed better RFS, OS, and RR benefits, but higher TRM. Subgroup analysis based on donor category (MSD and matched unrelated donor [MUD]) of alloSCT showed alloSCT from MSD rather than from MUD had better OS benefits compared to autoSCT. For fms-like tyrosine kinase 3 internal tandem duplications (FLT3-ITD) wild-type patients, alloSCT and autoSCT had comparable RFS and OS outcomes. CONCLUSION: Our results suggest that, in the absence of an available MSD, autoSCT remains a viable PRT alternative for intermediate-risk AML in CR1, especially for FLT3-ITD wild-type patients.


Asunto(s)
Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/terapia , Hermanos , Trasplante de Células Madre , Donante no Emparentado , Aloinjertos , Autoinjertos , Supervivencia sin Enfermedad , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Tasa de Supervivencia , Tirosina Quinasa 3 Similar a fms/genética
8.
Biochem Biophys Res Commun ; 507(1-4): 122-127, 2018 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-30420284

RESUMEN

To explore the role of type 1 regulatory T (Tr1) cells in allogeneic bone marrow transplantation (allo-BMT), we generated Tr1 cells from naïve CD4+ T cells in donor mice that were subjected to stimulation of recipient interleukin (IL)-10 gene-modified dendritic cells (DCs). In animal models, the severity of graft-versus-host disease (GVHD) in the group co-infused with DC-induced Tr1 cells was markedly milder than that in the groups without infusion of Tr1 cells. Moreover, the co-infusion Tr1 cells significantly prolonged the leukemia-free survival in tumor-bearing mice. Besides, Tr1-induced cells mediated the shift from T helper (Th)1 to Th2 cytokines in allo-BMT recipients. In Conclusions, our findings suggest that IL-10 gene-modified DC-induced Tr1 cells could inhibit GVHD while maintaining Graft-versus-leukemia (GVL) through mediating the shift of Th1/Th2 cytokines in an allo-BMT mice model.


Asunto(s)
Células Dendríticas/inmunología , Enfermedad Injerto contra Huésped/inmunología , Efecto Injerto vs Leucemia/inmunología , Interleucina-10/genética , Linfocitos T Reguladores/inmunología , Animales , Proliferación Celular , Vectores Genéticos/metabolismo , Enfermedad Injerto contra Huésped/patología , Lentivirus/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Linfocitos T Citotóxicos/inmunología
9.
Ann Hematol ; 97(10): 1809-1816, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29754271

RESUMEN

The standard treatment in elderly patients with diffuse large B cell lymphoma (DLBCL) has not yet been finely established. We investigated the efficacy and safety of rituximab with a reduced-dose of EPOCH chemotherapy in elderly patients who had advanced DLBCL with high IPI scores. The dose of 70% EPOCH was given to patients aged 75 to 79 years, and dose of 50% to patients aged over 80 years. Thirty-one patients with a median age of 79 years (range 75-86 years) were enrolled. Patients received a median of 6 cycle's chemotherapy. The complete response rate was 71.0%. The 3-year overall survival (OS) and progression-free survival rates were 62.8 and 60.3%, respectively. The most frequent grade 3/4 adverse effects were neutropenia (3 patients, 7 events), febrile neutropenia (3 patients, 5 events), and pulmonary infection (3 patients, 3 events). Our study showed that ECOG score 3-4, bulky disease, ß2-MG > 5.0 mg/L, and loss of any IADL are prognostic factors for OS in univariate analysis. In summary, reduced-dose EPOCH-R chemotherapy for very elderly patients is very effective with acceptable toxicities. Our preliminary study may provide an alternative approach to manage very elderly fit patients with advanced and poor risk DLBCL by first-line treatment with reduced-dose EPOCH-R.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/patología , Rituximab/administración & dosificación , Factores de Edad , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Etopósido/administración & dosificación , Etopósido/efectos adversos , Femenino , Humanos , Linfoma de Células B Grandes Difuso/mortalidad , Masculino , Estadificación de Neoplasias , Prednisona/administración & dosificación , Prednisona/efectos adversos , Pronóstico , Rituximab/efectos adversos , Resultado del Tratamiento , Vincristina/administración & dosificación , Vincristina/efectos adversos
10.
Cell Physiol Biochem ; 44(1): 240-254, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29130994

RESUMEN

BACKGROUND/AIMS: Minimal residual leukemia cells (MRLs) are difficult to eradicate through traditional treatment and therefore remain to be a major threat to the long-term survival of leukemia patients. Tumor-derived exosomes (TEXs), which carry tumor associated antigens (TAA), may be a potential cell-free tumor vaccine for the specific eradication of MRLs. However, TEXs are intended to be less immunogenic due to exosomal TGF-ß1. To further optimize the efficacy of TEX-based vaccines, we investigated whether exosomes from TGF-ß1 silenced leukemia cells (LEXTGF-ß1si) had an increased potential to induce a specific antitumor effect compared with non-modified exosomes. METHODS: Exosomal TGF-ß1 was downregulated via lentiviral shRNA silencing of TGF-ß1 in leukemia cells. The characteristics of LEXTGF-ß1si were determined via electron microscopy, western blot analysis, and flow cytometry. The antitumor effect of LEXTGF-ß1si was evaluated by detecting the properties of LEXTGF-ß1si-pulsed dendritic cells (DCs), CD4+ T-cell proliferation, Th1 cytokine secretion, specific CTL activity, and NK cell function. Moreover, to verify the superiority of LEXTGF-ß1si immunization, LEXTGF-ß1si was subcutaneously injected into DBA/2 mice: either followed by tumor challenge or tumor bearing. RESULTS: The lentiviral shRNA silencing of TGF-ß1 in parental leukemia cells successfully downregulated the TGF-ß1 level in leukemia cell derived exosomes (LEX). LEXTGF-ß1si was uptaken by DCs and was more potent in promoting DC function by upregulating the surface expression of costimulatory factors and MHC class II molecules, while inducing the secretion of IL-12p70 and TNF-α. Furthermore, immunization with LEXTGF-ß1si facilitated CD4+ T-cell proliferation and Th1 cytokine secretion, and stimulated stronger specific cytotoxic lymphocyte (CTL) response and nature killer (NK) cell cytotoxicity more efficiently compared to non-modified LEX. In mice models, immunization with LEXTGF-ß1si resulted in a more potent capability to inhibit tumor growth and to prolong survival, suggesting that LEXTGF-ß1si was more effective in both protective and therapeutic antitumor tests than non-modified LEX. CONCLUSIONS: These data suggested that down-regulation of exosomal TGF-ß1 effectively induced potent anti-tumor immunity. Our strategy of optimizing exosome vaccine may have promising potential for leukemia immunotherapy.


Asunto(s)
Exosomas/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación hacia Abajo , Exosomas/metabolismo , Humanos , Inmunización , Inmunoterapia , Interleucina-12/metabolismo , Células Asesinas Naturales/inmunología , Leucemia/metabolismo , Leucemia/patología , Leucemia/terapia , Ratones , Ratones Endogámicos DBA , Interferencia de ARN , Linfocitos T Citotóxicos/inmunología , Células TH1/citología , Células TH1/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/genética , Factor de Necrosis Tumoral alfa/metabolismo
11.
Cell Physiol Biochem ; 43(1): 353-366, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28869950

RESUMEN

BACKGROUND/AIMS: Tr1 cells can induce peripheral tolerance to self- and foreign antigens, and have been developed as a therapeutic tool for the induction of tolerance to transplanted tissue. We explored the feasibility of generating Tr1 cells by using IL-10 gene-modified recipient DCs (DCLV-IL-10) to stimulate donor naive CD4+ T cells. We also investigated some biological properties of Tr1 cells. METHODS: DCLV-IL-10 were generated through DCs transduced with a lentivirus vector carrying the IL-10 gene, and Tr1 cells were produced by using DCLV-IL-10 to stimulate naive CD4+ T cells. The effects of Tr1 cells on T-cell proliferation and the occurrence of graft versus host disease (GVHD) following allogeneic stem-cell transplantation (allo-HSCT) were investigated. RESULTS: The DCLV-IL-10-induced Tr1 cells co-expressed LAG-3 and CD49b. Moreover, they also expressed CD4, CD25, and IL-10, but not Foxp3, and secreted significantly higher levels of IL-10 (1,729.36 ± 185.79 pg/mL; P < 0.001) and INF-γ (1,524.48 ± 168.65 pg/mL; P < 0.01) than the control T cells upon the stimulation by allogeneic DCs. Tr1 cells markedly suppressed T-lymphocyte proliferation and the mixed lymphocytic response (MLR) in vitro. The mice used in the allo-HSCT model had longer survival times and lower clinical and pathological GVHD scores than the control mice. CONCLUSION: IL-10 gene-modified DC-induced Tr1 cells may be used as a potent cellular therapy for the prevention of GVHD after allo-HSCT.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Interleucina-10/metabolismo , Trasplante de Células Madre , Linfocitos T Reguladores/trasplante , Animales , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Enfermedad Injerto contra Huésped/epidemiología , Interleucina-10/análisis , Interleucina-10/genética , Interleucina-12/análisis , Interleucina-12/genética , Interleucina-12/metabolismo , Interleucina-6/análisis , Interleucina-6/genética , Interleucina-6/metabolismo , Lentivirus/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Trasplante Homólogo/efectos adversos
12.
Cancer Immunol Immunother ; 66(10): 1321-1331, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28601924

RESUMEN

Tumor-derived exosomes (TEX) can induce a specific antitumor immune response and have been developed as a promising tumor vaccine. Despite promising preclinical data, TEX exhibit relatively low efficacy and limited clinical benefit in clinical trials. In the present study, we investigated whether exosomes from the TGF-ß1 silenced L1210 cells (LEXTGF-ß1si) can enhance the efficacy of DC-based vaccines. We silenced TGF-ß1 in L1210 cells with a lentiviral shRNA vector and prepared the LEXTGF-ß1si. It was shown that LEXTGF-ß1si can significantly decrease TGF-ß1 expression of dendritic cells (DC) and effectively promote their maturation and immune function. In addition, DC pulsed with LEXTGF-ß1si (DCLEX-TGF-ß1si) more effectively promoted CD4+ T cell proliferation in vitro and Th1 cytokine secretion and induced tumor-specific CTL response. This response was higher in potency compared to that noted by the other two formulations. Moreover, DCLEX-TGF-ß1si inhibited tumor growth more efficiently than other formulations did as the preventive or therapeutic tumor vaccine. Accordingly, these findings revealed that DCLEX-TGF-ß1si induced a more potent antigen-specific anti-leukemic immunity than DC pulsed with exosomes from non-manipulated L1210 cells. This indicated that the targeting of DC by LEXTGF-ß1si may be used as a promising approach for leukemia immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Inmunoterapia/métodos , Leucemia/genética , Factor de Crecimiento Transformador beta1/inmunología , Animales , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Exosomas , Femenino , Humanos , Ratones
14.
Ann Hematol ; 94(5): 761-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25519475

RESUMEN

To better understand the predictive factors and improve clinical outcome of allogeneic transplant for patients with myelodysplastic syndrome (MDS), we retrospectively analyzed the post-transplant outcome of 60 Southeast Asian patients with MDS. Multivariate analysis showed that WHO classification-based Prognostic Scoring System (WPSS) significantly affect overall survival (OS), progression-free survival (PFS), cumulative incidence of relapse (CIR), and cumulative incidence of non-relapse mortality (CINRM). Stratified by WPSS into very low/low, intermediate, high, and very high-risk categories, 3-year OS was 100, 61, 37, and 18% (p = 0.02); PFS was 100, 55, 32, and 18% (p = 0.014); CIR was 12, 24, 38, and 59% (p = 0.024); CINRM was 0, 6, 12, and 26% (p = 0.037), respectively. WHO classification, Revised International Prognostic Scoring System (IPSS-R), IPSS-R-defined cytogenetic risk groups, donor gender, and acute and chronic graft vs host disease (GVHD) also influenced different aspects of transplant outcome. We found that WPSS is a powerful predictor of post-transplant outcome. WPSS provides an important model not only for prognostication but also for exploration of further post-transplant measures such as immunological maneuvers or novel therapy to improve the poor outcome of high-risk patients.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Síndromes Mielodisplásicos/terapia , Trasplante Homólogo , Asia Sudoriental , Supervivencia sin Enfermedad , Femenino , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/terapia , Humanos , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/mortalidad , Síndromes Mielodisplásicos/patología , Pronóstico , Estudios Retrospectivos , Resultado del Tratamiento
15.
Vaccine ; 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38960787

RESUMEN

OBJECTIVES: Leukemia cell-derived exosomes (LEXs), carrying leukemia cell-specific antigens, can serve as a source of antigen for dendritic cell (DC) vaccine loading. However, LEX-targeted DC-based vaccines have demonstrated limited antitumor immune effects in clinical trials, attributed to the low immunogenicity of LEXs and the scant levels of costimulatory molecules on DCs. The costimulatory molecules CD80 and CD86, which are crucial to DC function, play a significant role in enhancing immune efficacy. In this study, we explored the anti-leukemia immune response of costimulatory molecule gene-modified LEX-targeted DCs (LEX-8086) in vitro and in animal models. METHODS: DCs were incubated with LEX-8086 to produce LEX-8086-targeted DCs (DCsLEX-8086). ELISA, cytotoxicity assays and flow cytometry utilized to assess the antitumor efficacy of DCsLEX8086 in vitro. Flow cytometry was used to evaluate the immunomodulatory function of DCsLEX8086 in animal models. RESULTS: Our findings indicated that LEX-8086 enhanced the maturation and antigen-presenting ability of DCs. Immunization with DCsLEX8086 significantly activated CD8+ T cells and boosted the CTL response in vitro. More importantly, DCsLEX-8086 effectively suppressed tumor growth and exerted anti-leukemia effects in both prophylactic and therapeutic animal models. Furthermore, DCsLEX-8086 promoted the proportion of CD4+ T cells, CD8+ T cells and M1 macrophages in the tumor environments both prophylactically and therapeutically. Treatment with DCsLEX-8086 showed no significant difference in the levels of M2 macrophages but decreased the proportion of Tregs within the tumor bed during therapeutic experiments. CONCLUSION: The results suggested that DCsLEX-8086 induces a more effective anti-leukemia immunity compared to DCsLEX-null in vivo and in vitro. DCsLEX-8086 might achieve antitumor effects by elevating the numbers of CD4+ T cells, CD8+ T cells, and M1 macrophages in tumors. Our findings indicate that DCsLEX-8086 could be leveraged to develop a new, highly effective vaccine for anti-leukemia immunity.

16.
Clin Cancer Res ; 30(7): 1248-1255, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38190117

RESUMEN

PURPOSE: Patients with peripheral T-cell lymphomas (PTCL) in the relapsed or refractory (r/r) setting have only a limited number of therapies available, and the prognosis is extremely poor. SHR2554 is an oral inhibitor against EZH2, a rational therapeutic target for lymphomas. PATIENTS AND METHODS: This was a multicenter, two-part, phase I study of SHR2554 in r/r mature lymphoid neoplasms. In part I, 350 mg twice daily was established as the recommended phase II dose (RP2D) based on the findings during dose escalation and expansion; subsequently, selected lymphoma subtypes were recruited in clinical expansion cohorts to receive SHR2554 at RP2D. Here, we provide an in-depth assessment of SHR2554 at RP2D in subpopulation with r/r PTCL. RESULTS: Twenty-eight patients were included for analysis (17 angioimmunoblastic T-cell lymphoma and 11 not otherwise specified). Eighteen (64%) patients had received ≥2 lines of previous anticancer therapies. The objective response rate was 61% [95% confidence interval (CI), 41-78]. Responses were still ongoing in 59% (10/17) of the responders; estimated median duration of response was 12.3 months (95% CI, 7.4-not reached). Median progression-free survival was 11.1 months (95% CI, 5.3-22.0), and 12-month overall survival rate was 92% (95% CI, 72-98). The most common grade 3 or 4 treatment-related adverse events were decreased platelet count [nine (32%)] as well as decreased white blood cell count, decreased neutrophil count, and anemia [four (14%) for each]. No treatment-related deaths were reported. CONCLUSIONS: This extended follow-up analysis further supports SHR2554 as a therapeutic opportunity for patients with r/r PTCL.


Asunto(s)
Linfoma de Células T Periférico , Humanos , Linfoma de Células T Periférico/tratamiento farmacológico , Linfoma de Células T Periférico/genética , Linfoma de Células T Periférico/patología , Resultado del Tratamiento , Proteína Potenciadora del Homólogo Zeste 2 , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Pronóstico , Inhibidores Enzimáticos/uso terapéutico
17.
Biochem Biophys Res Commun ; 436(1): 60-5, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23707941

RESUMEN

Tumor cell-derived exosomes (TEX) have been widely used to induce antitumor immune responses in animal models and clinical trials. However, the efficiency of the antitumor immunity that is induced by TEX is still relatively weak. In this study, we compared the antitumor immunities between EG7 tumor cell-derived exosomes (EXO(EG7)) and EXO(EG7)-targeted dendritic cells (DC(EXO)). We found that EXO(EG7) harbored OVA and peptide major histocompatibility complex I (pMHC-I), which were expressed on its parental EG7 tmor cells, and they could transfer OVA and pMHC-I to dendritic cells (DCs) in vitro. DC(EXO) could more efficiently induce antitumor immunity than EXO(EG7). In addition, we showed that the immune stimulatory effects of EXO(EG7) were dependent on the host DCs and, whereas those of DC(EXO) were not, indicating the important role of the host DCs in TEX vaccines. Taken together, TEX-targeted DCs may be more effective for EXO-based vaccines for the induction of antitumor immunity.


Asunto(s)
Linfocitos T CD8-positivos/citología , Células Dendríticas/citología , Exosomas/metabolismo , Neoplasias/inmunología , Linfocitos T Citotóxicos/citología , Animales , Biotina/química , Células de la Médula Ósea/citología , Proliferación Celular , Células Dendríticas/inmunología , Femenino , Inmunoterapia/métodos , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo
18.
Breast Cancer Res Treat ; 140(2): 273-84, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23881522

RESUMEN

One of the major obstacles in human epidermal growth factor receptor 2 (HER2)-specific trastuzumab antibody immunotherapy of HER2-positive breast cancer is the development of trastuzumab resistance, warranting the search for other therapeutic strategies. Using mouse models, we previously demonstrated that ovalbumin (OVA)-specific dendritic cell (DC)-released exosome (EXOOVA)-targeted CD4(+) T cell-based (OVA-TEXO) vaccine stimulates efficient cytotoxic T lymphocyte (CTL) responses via exosomal peptide/major histocompatibility complex (pMHC)-I, exosomal CD80 and endogenous IL-2 signaling; and long-term CTL memory by means of via endogenous CD40L signaling. In this study, using two-photon microscopy, we provide the first visual evidence on targeting OVA-TEXO to cognate CD8(+) T cells in vivo via exosomal pMHC-I complex. We prepared HER2/neu-specific Neu-TEXO and HER2-TEXO vaccines using adenoviral vector (AdVneu and AdVHER2)-transfected DC (DCneu and DCHER2)-released EXOs (EXOneu and EXOHER2), and assessed their stimulatory effects on HER2/neu-specific CTL responses and antitumor immunity. We demonstrate that Neu-TEXO vaccine is capable of stimulating efficient neu-specific CTL responses, leading to protective immunity against neu-expressing Tg1-1 breast cancer in all 6/6 transgenic (Tg) FVBneuN mice with neu-specific self-immune tolerance. We also demonstrate that HER2-TEXO vaccine is capable of inducing HER2-specific CTL responses and protective immunity against transgene HLA-A2(+)HER2(+) BL6-10A2/HER2 B16 melanoma in 2/8 double Tg HLA-A2/HER2 mice with HER2-specific self-immune tolerance. The remaining 6/8 mice had significantly prolonged survival. Furthermore, we demonstrate that HER2-TEXO vaccine stimulates responses of CD8(+) T cells capable of not only inducing killing activity to HLA-A2(+)HER2(+) BL6-10A2/HER2 melanoma and trastuzumab-resistant BT474A2 breast cancer cells in vitro but also eradicating 6-day palpable HER2(+) BT474A2 breast cancer (3-4 mm in diameter) in athymic nude mice. Therefore, the novel T cell-based HER2-TEXO vaccine may provide a new therapeutic alternative for women with HER2(+) breast cancer, especially for trastuzumab-resistant HER2(+) breast cancer patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/uso terapéutico , Resistencia a Antineoplásicos/inmunología , Antígeno HLA-A2/inmunología , Linfocitos T Citotóxicos/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Resistencia a Antineoplásicos/genética , Exosomas/inmunología , Femenino , Humanos , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Desnudos , Ratones Transgénicos , Receptor ErbB-2/inmunología , Linfocitos T Citotóxicos/inmunología , Trastuzumab
19.
Front Immunol ; 14: 1284936, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38124750

RESUMEN

Background: Mesenchymal stem cells (MSCs) can alleviate graft-versus-host disease (GVHD) in hematopoietic stem cell transplantation (HSCT). MSCs-derived exosomes (MEXs) can mirror the biological function of their parent cells. Whether MEXs can alleviate GVHD like their parent cells or not is unclear. In this study, we investigate the effects of MEXs on GVHD and graft-versus-leukemia (GVL) effect in vitro and in HSCT animal models. Method: MSCs were produced using bone marrow mononuclear cells (MNCs), and MEXs were separated from the supernatants of MSCs. Electron microscopy, western blot, and nanoparticle tracking analysis (NTA) were used to determine the characteristics of MEXs. The immunomodulatory function of MEXs and their effects on GVHD and GVL were examined in vitro and in vivo. Result: Like other cell-type derived exosomes, our data revealed that MEXs were also disc-shaped vesicles with a diameter of 100-200 nm under electron microscopy and were positive for the exosomal hallmark proteins. MEXs can notably inhibit the expression of costimulatory molecules and functional cytokine secretion of dendritic cells (DCs). Meanwhile, MEXs can exert suppressive effects on T lymphocyte proliferation and activation. Moreover, MEXs can also encourage the polarization of macrophages toward the M2 type. In animal HSCT models, MEXs can promote the differentiation of Treg cells in spleens, decrease the GVHD score, increase the survival rate of mice, and preserve the cytotoxic antileukemia effects of CD8+ T lymphocytes from recipient mice. Conclusion: These findings showed that MEXs exert their effects by inhibiting the immunomodulatory function of DCs, macrophages, and T lymphocytes. In the animal model, MEXs ameliorate the clinical symptoms of GVHD, while maintaining the antitumor effects of CD8+ T lymphocytes. Therefore, it can be inferred that MEXs can separate GVHD from GVL in HSCT. Our study suggests that MEXs have broad clinical application potential in the prevention and treatment of GVHD in HSCT in the near future.


Asunto(s)
Exosomas , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Células Madre Mesenquimatosas , Ratones , Animales , Exosomas/patología , Trasplante Homólogo , Enfermedad Injerto contra Huésped/patología , Modelos Animales
20.
J Clin Oncol ; 41(16): 3032-3041, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36996373

RESUMEN

PURPOSE: Relapsed or refractory extranodal natural killer/T-cell lymphoma (R/R ENKTL) is a rare and aggressive type of non-Hodgkin lymphoma with limited treatment options. This phase II study evaluated the efficacy and safety of sugemalimab, an anti-PD-L1 monoclonal antibody, in R/R ENKTL. METHODS: Eligible patients received sugemalimab 1,200 mg intravenously once every 3 weeks for up to 24 months or until progression, death, or study withdrawal. The primary end point was objective response rate (ORR) assessed by an independent radiologic review committee. Key secondary end points included ORR assessed by the investigators, complete response rate, duration of response, and safety. RESULTS: At the data cutoff (February 23, 2022), 80 patients were enrolled and followed for a median of 18.7 months. At baseline, 54 (67.5%) had stage IV disease and 39 (48.8%) had received ≥2 lines of prior systemic therapy. Independent radiologic review committee-assessed ORR was 44.9% (95% CI, 33.6 to 56.6); 28 (35.9%) patients achieved a complete response and seven (9.0%) achieved a partial response, with a 12-month duration of response rate of 82.5% (95% CI, 62.0 to 92.6). Investigator-assessed ORR was 45.6% (95% CI, 34.3 to 57.2), and 24 (30.4%) patients achieved a complete response. Most treatment-emergent adverse events were grade 1-2 in severity, and grade ≥ 3 events were reported in 32 (40.0%) patients. CONCLUSION: Sugemalimab showed robust and durable antitumor activity in R/R ENKTL. Treatment was well tolerated with expected safety profile for this drug class.


Asunto(s)
Linfoma Extranodal de Células NK-T , Humanos , Linfoma Extranodal de Células NK-T/tratamiento farmacológico , Resultado del Tratamiento , Anticuerpos Monoclonales , Células Asesinas Naturales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA