Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Hum Immunol ; 83(1): 86-98, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34583856

RESUMO

The global outbreak of coronavirus-2019 (COVID-19) still claims more lives daily around the world due to the lack of a definitive treatment and the rapid tendency of virus to mutate, which even jeopardizes vaccination efficacy. At the forefront battle against SARS-CoV-2, an effective innate response to the infection has a pivotal role in the initial control and treatment of disease. However, SARS-CoV-2 subtly interrupts the equations of immune responses, disrupting the cytolytic antiviral effects of NK cells, while seriously activating infected macrophages and other immune cells to induce an unleashed "cytokine storm", a dangerous and uncontrollable inflammatory response causing life-threatening symptoms in patients. Notably, the NK cell exhaustion with ineffective cytolytic function against the sources of exaggerated cytokine release, acts as an Achilles' heel which exacerbates the severity of COVID-19. Given this, approaches that improve NK cell cytotoxicity may benefit treatment protocols. As a suggestion, adoptive transfer of NK or CAR-NK cells with proper cytotolytic potentials and the lowest capacity of cytokine-release (for example CD56dim NK cells brightly express activating receptors), to severe COVID-19 patients may provide an effective cure especially in cases suffering from cytokine storms. More intriguingly, the ongoing evidence for persistent clonal expansion of NK memory cells characterized by an activating phenotype in response to viral infections, can benefit the future studies on vaccine development and adoptive NK cell therapy in COVID-19. Whether vaccinated volunteers or recovered patients can also be considered as suitable candidates for cell donation could be the subject of future research.


Assuntos
Transferência Adotiva , COVID-19/terapia , Síndrome da Liberação de Citocina/terapia , Citocinas/imunologia , Células Matadoras Naturais/transplante , SARS-CoV-2/imunologia , Transferência Adotiva/efeitos adversos , Animais , COVID-19/imunologia , COVID-19/metabolismo , COVID-19/virologia , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/metabolismo , Síndrome da Liberação de Citocina/virologia , Citocinas/metabolismo , Citotoxicidade Imunológica , Interações Hospedeiro-Patógeno , Humanos , Memória Imunológica , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/virologia , SARS-CoV-2/patogenicidade , Resultado do Tratamento
2.
Front Immunol ; 12: 727814, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925312

RESUMO

Posttransplant smooth muscle tumors (PTSMTs) are rare Epstein-Barr virus (EBV)-associated neoplasms, mostly occurring after solid organ transplantation. Current therapeutic strategies include surgery and reduction of immunosuppressive medication. We describe for the first time a novel treatment approach for PTSMT by adoptive cell transfer (ACT) of EBV-specific T cells to a 20-year-old patient with a medical history of cardiac transplantation, posttransplant lymphoproliferative disease, and multilocular PTSMT. During ACT, mild cytokine release syndrome occurred, while no unexpected safety signals were recorded. We observed in vivo expansion of EBV-specific T cells and reduction of EBV viremia. Best response was stable disease after 4 months with reduction of EBV viremia and normalization of lactate dehydrogenase levels. ACT with EBV-specific T cells may be a safe and efficacious therapeutic option for PTSMT that warrants further exploration.


Assuntos
Transferência Adotiva/efeitos adversos , Células Alógenas/imunologia , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/terapia , Transplante de Coração/efeitos adversos , Herpesvirus Humano 4/imunologia , Tumor de Músculo Liso/complicações , Tumor de Músculo Liso/terapia , Linfócitos T/imunologia , Transferência Adotiva/métodos , Infecções por Vírus Epstein-Barr/virologia , Feminino , Humanos , Transtornos Linfoproliferativos/etiologia , Tumor de Músculo Liso/etiologia , Transplante Homólogo , Resultado do Tratamento , Viremia/complicações , Viremia/terapia , Adulto Jovem
3.
Front Immunol ; 12: 734246, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34691041

RESUMO

T-cell therapy with T cells that are re-directed to hepatitis B virus (HBV)-infected cells by virus-specific receptors is a promising therapeutic approach for treatment of chronic hepatitis B and HBV-associated cancer. Due to the high number of target cells, however, side effects such as cytokine release syndrome or hepatotoxicity may limit safety. A safeguard mechanism, which allows depletion of transferred T cells on demand, would thus be an interesting means to increase confidence in this approach. In this study, T cells were generated by retroviral transduction to express either an HBV-specific chimeric antigen receptor (S-CAR) or T-cell receptor (TCR), and in addition either inducible caspase 9 (iC9) or herpes simplex virus thymidine kinase (HSV-TK) as a safety switch. Real-time cytotoxicity assays using HBV-replicating hepatoma cells as targets revealed that activation of both safety switches stopped cytotoxicity of S-CAR- or TCR-transduced T cells within less than one hour. In vivo, induction of iC9 led to a strong and rapid reduction of transferred S-CAR T cells adoptively transferred into AAV-HBV-infected immune incompetent mice. One to six hours after injection of the iC9 dimerizer, over 90% reduction of S-CAR T cells in the blood and the spleen and of over 99% in the liver was observed, thereby limiting hepatotoxicity and stopping cytokine secretion. Simultaneously, however, the antiviral effect of S-CAR T cells was diminished because remaining S-CAR T cells were mostly non-functional and could not be restimulated with HBsAg. A second induction of iC9 was only able to deplete T cells in the liver. In conclusion, T cells co-expressing iC9 and HBV-specific receptors efficiently recognize and kill HBV-replicating cells. Induction of T-cell death via iC9 proved to be an efficient means to deplete transferred T cells in vitro and in vivo containing unwanted hepatotoxicity.


Assuntos
Transferência Adotiva , Caspase 9/biossíntese , Antígenos da Hepatite B/imunologia , Vírus da Hepatite B/imunologia , Hepatite B Crônica/terapia , Linfócitos T/transplante , Transferência Adotiva/efeitos adversos , Animais , Caspase 9/genética , Morte Celular , Linhagem Celular , Técnicas de Cocultura , Citocinas/metabolismo , Citotoxicidade Imunológica , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Indução Enzimática , Feminino , Vírus da Hepatite B/patogenicidade , Hepatite B Crônica/imunologia , Hepatite B Crônica/metabolismo , Hepatite B Crônica/virologia , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Simplexvirus/enzimologia , Simplexvirus/genética , Linfócitos T/enzimologia , Linfócitos T/imunologia , Linfócitos T/patologia , Timidina Quinase/genética , Timidina Quinase/metabolismo , Transdução Genética
4.
BMC Cancer ; 21(1): 877, 2021 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-34332557

RESUMO

BACKGROUND: Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), such as gefitinib, have achieved good efficacy in EGFR mutation-positive non-small-cell lung cancer (NSCLC) patients, but eventual drug resistance is inevitable. Thus, new TKI-based combination therapies should be urgently explored to extend the overall survival time of these patients. CD8 + CD56+ natural killer T (NKT) cells are a natural and unique subset of lymphocytes in humans that present characteristics of T and NK cells and exert cytotoxicity on tumour cells in a granzyme B-dependent manner. The aim of this trial was to explore the efficacy and safety of CD8 + CD56+ NKT cell immunotherapy combined with gefitinib in patients with advanced EGFR-mutated NSCLC. METHODS: The study was designed as a prospective, randomized, controlled, open-label, phase I/II trial that includes 30 patients with EGFR mutation-positive stage III/IV NSCLC. All patients will be randomized in blocks at a 1:1 ratio and treated with gefitinib 250 mg/day monotherapy or combination therapy with allogeneic CD8 + CD56+ NKT cell infusions twice per month for 12 cycles or until disease progression occurs. The effectiveness of this treatment will be evaluated based on by progression-free survival (PFS), the time to progression (TTP), overall response rate (ORR), disease control rate (DCR) and overall survival (OS). The safety of the trail is being assessed based on adverse events (AEs). Recruitment and data collection, which started in December 2017, are ongoing. DISCUSSION: Although immunotherapy, including programmed death-1/programmed death-1 ligand (PD-1/PD-L1) immunotherapy, has been used for NSCLC treatment with or without EGFR-TKIs, its clear efficacy still has not been shown. Assessing the safety and therapeutic potential of allogeneic CD8 + CD56+ NKT killer cells in combination with EGFR-TKIs in NSCLC will be of great interest. TRIAL REGISTRATION: This trial (Phase I/II Trails of NKT Cell in Combination With Gefitinib For Non Small Cell Lung Cancer) was registered on 21 November 2017 with www.chictr.org.cn , ChiCTR-IIR-17013471 .


Assuntos
Transferência Adotiva , Carcinoma Pulmonar de Células não Pequenas/terapia , Gefitinibe/uso terapêutico , Neoplasias Pulmonares/terapia , Mutação , Células T Matadoras Naturais/imunologia , Transferência Adotiva/efeitos adversos , Transferência Adotiva/métodos , Antígeno B7-H1/antagonistas & inibidores , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/etiologia , Terapia Combinada , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Gefitinibe/administração & dosagem , Gefitinibe/efeitos adversos , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/etiologia , Terapia de Alvo Molecular , Células T Matadoras Naturais/metabolismo , Resultado do Tratamento
5.
Front Immunol ; 12: 625998, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717141

RESUMO

Kidney transplantation is a primary therapy for end-stage renal disease (ESRD) all the time. But it does not mean that we have fully unraveling the mystery of kidney transplantation and confer every patient favorable prognosis. Immune rejection has always been a stumbling block when we try to increase the success rate of kidney transplantation and improve long-term outcomes. Even if the immune rejection is effectively controlled in acute phase, there is a high possibility that the immune response mediated by chronically activated antibodies will trigger chronic rejection and ultimately lead to graft failure. At present, immunosuppressive agent prepared chemically is mainly used to prevent acute or chronic rejection, but it failed to increase the long-term survival rate of allografts or reduce the incidence of chronic rejection after acute rejection, and is accompanied by many adverse reactions. Therefore, many studies have begun to use immune cells to regulate the immune response in order to control allograft rejection. This article will focus on the latest study and prospects of more popular regulatory myeloid cells in the direction of renal transplantation immunotherapy and introduce their respective progress from experimental research to clinical research.


Assuntos
Transferência Adotiva , Rejeição de Enxerto/terapia , Sobrevivência de Enxerto , Transplante de Rim/efeitos adversos , Transplante de Células-Tronco Mesenquimais , Células Mieloides/transplante , Transferência Adotiva/efeitos adversos , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/transplante , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Humanos , Imunossupressores/uso terapêutico , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/transplante , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Resultado do Tratamento
6.
Front Immunol ; 12: 624685, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33679769

RESUMO

Antigen-specific therapy for multiple sclerosis may lead to a more effective therapy by induction of tolerance to a wide range of myelin-derived antigens without hampering the normal surveillance and effector function of the immune system. Numerous attempts to restore tolerance toward myelin-derived antigens have been made over the past decades, both in animal models of multiple sclerosis and in clinical trials for multiple sclerosis patients. In this review, we will give an overview of the current approaches for antigen-specific therapy that are in clinical development for multiple sclerosis as well provide an insight into the challenges for future antigen-specific treatment strategies for multiple sclerosis.


Assuntos
Transferência Adotiva , Dessensibilização Imunológica , Esclerose Múltipla/terapia , Proteínas da Mielina/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Vacinação , Vacinas/uso terapêutico , Transferência Adotiva/efeitos adversos , Transferência Adotiva/história , Transferência Adotiva/tendências , Animais , Autoimunidade , Dessensibilização Imunológica/efeitos adversos , Dessensibilização Imunológica/história , Dessensibilização Imunológica/tendências , Difusão de Inovações , Previsões , História do Século XX , História do Século XXI , Humanos , Tolerância Imunológica , Esclerose Múltipla/história , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Proteínas da Mielina/efeitos adversos , Proteínas da Mielina/imunologia , Proteínas da Mielina/metabolismo , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Vacinação/efeitos adversos , Vacinação/história , Vacinação/tendências , Vacinas/efeitos adversos
7.
Ann Neurol ; 89(4): 769-779, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33459417

RESUMO

OBJECTIVE: Progressive multifocal leukoencephalopathy (PML) is still burdened by high mortality in a subset of patients, such as those affected by hematological malignancies. The aim of this study was to analyze the safety and carry out preliminary evaluation of the efficacy of polyomavirus JC (JCPyV)-specific T cell therapy in a cohort of hematological patients with PML. METHODS: Between 2014 and 2019, 9 patients with a diagnosis of "definite PML" according to the 2013 consensus who were showing progressive clinical deterioration received JCPyV-specific T cells. Cell lines were expanded from autologous or allogenic peripheral blood mononuclear cells by stimulation with JCPyV antigen-derived peptides. RESULTS: None of the patients experienced treatment-related adverse events. In the evaluable patients, an increase in the frequency of circulating JCPyV-specific lymphocytes was observed, with a decrease or clearance of JCPyV viral load in cerebrospinal fluid. In responsive patients, transient appearance of punctate areas of contrast enhancement within, or close to, PML lesions was observed, which was interpreted as a sign of immune control and which regressed spontaneously without the need for steroid treatment. Six of 9 patients achieved PML control, with 5 alive and in good clinical condition at their last follow-up. INTERPRETATION: Among other novel treatments, T cell therapy is emerging as a viable treatment option in patients with PML, particularly for those not amenable to restoration of specific immunity. Neurologists should be encouraged to refer PML patients to specialized centers to allow access to this treatment strategy. ANN NEUROL 2021;89:769-779.


Assuntos
Transferência Adotiva/métodos , Vírus JC , Leucoencefalopatia Multifocal Progressiva/terapia , Linfócitos T , Adolescente , Transferência Adotiva/efeitos adversos , Idoso , Estudos de Coortes , Progressão da Doença , Feminino , Humanos , Leucoencefalopatia Multifocal Progressiva/líquido cefalorraquidiano , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Carga Viral
8.
Int Immunopharmacol ; 91: 107322, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33418238

RESUMO

Regulatory macrophages (Mregs) are a subtype of macrophages that are involved in regulating immune responses and inhibiting activated T lymphocyte proliferation. With advances in our basic understanding of Mregs and the revelation of their biological characteristics, Mregs have become a focus of research. In addition to promoting malignant tumor progression, Mregs also play an immunosuppressive role in inflammatory diseases and transplantation. Recent studies have shown that Mregs are closely associated with the induction of transplantation immune tolerance. Immune regulatory cell treatment as an adjunct immunosuppressive therapy offers new insights into the mechanism by which transplantation immune tolerance is established. The application of Mreg-based cellular immunotherapy has shown promise in clinical solid organ transplantation. Here, we provide a comprehensive overview of Mreg morphology, phenotype, induction and negative immunoregulatory function and discuss the role of Mregs in different transplantation models as well as their potential application value in clinical organ transplantation.


Assuntos
Transferência Adotiva , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Macrófagos/transplante , Transplante de Órgãos , Tolerância ao Transplante , Transferência Adotiva/efeitos adversos , Animais , Terapia Combinada , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Imunossupressores/uso terapêutico , Macrófagos/imunologia , Macrófagos/metabolismo , Transplante de Órgãos/efeitos adversos , Fenótipo , Tolerância ao Transplante/efeitos dos fármacos , Resultado do Tratamento
10.
Int Immunopharmacol ; 90: 107195, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33278746

RESUMO

Since half of the genes are inherited from the paternal side, the maternal immune system has to tolerate the presence of foreign paternal antigens. Regulatory T cells facilitate the development and maintenance of peripheral tissue tolerance of the fetus during pregnancy. Reduction in regulatory T cells is associated with complications of pregnancy, including spontaneous abortion. Recent studies in mouse models have shown that the adoptive transfer of Tregs can prevent spontaneous abortion in mouse models through improving maternal tolerance. Thus, adoptive cell therapy using autologous Tregs could potentially be a novel therapeutic approach for cell-based immunotherapy in women with unexplained spontaneous abortion. Besides, strategies for activating and expanding antigen-specific Tregs ex vivo and in vivo based on pharmacological agents can pave the foundation for an approach incorporating immunotherapy and pharmacotherapy. This review aims to elaborate on the current understanding of the therapeutic potential of the adoptive transfer of Tregs in the treatment of spontaneous abortion disease.


Assuntos
Aborto Espontâneo/prevenção & controle , Transferência Adotiva , Tolerância Imunológica , Linfócitos T Reguladores/transplante , Aborto Espontâneo/imunologia , Aborto Espontâneo/metabolismo , Transferência Adotiva/efeitos adversos , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Fenótipo , Gravidez , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
11.
Front Immunol ; 12: 801281, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975916

RESUMO

BK virus (BKV)-hemorrhagic cystitis (HC) is a well-known and rarely fatal complication of hematopoietic stem cell transplantation (HSCT). Treatment for BKV-HC is limited, but virus-specific T-cells (VST) represent a promising therapeutic option feasible for use posttransplant. We report on the case of a 16-year-old male with dedicator of cytokinesis 8 (DOCK8) deficiency who underwent haploidentical HSCT complicated by severe BKV-HC, catastrophic renal hemorrhage, and VST-associated cytokine release syndrome (CRS). Gross hematuria refractory to multiple interventions began with initiation of posttransplant cyclophosphamide (PT/Cy). Complete left renal arterial embolization (day +43) was ultimately indicated to control intractable renal hemorrhage. Subsequent infusion of anti-BK VSTs was complicated by CRS and progressive multiorgan failure, with postmortem analysis confirming diagnosis of hepatic sinusoidal obstruction syndrome (SOS). This case illustrates opportunities for improvement in the management of severe BKV-HC posttransplant while highlighting rare and potentially life-threatening complications of BKV-HC and VST therapy.


Assuntos
Transferência Adotiva/efeitos adversos , Vírus BK/patogenicidade , Cistite/terapia , Síndrome da Liberação de Citocina/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Hemorragia/terapia , Infecções por Polyomavirus/terapia , Linfócitos T/transplante , Infecções Tumorais por Vírus/terapia , Adolescente , Vírus BK/imunologia , Cistite/diagnóstico , Cistite/imunologia , Cistite/virologia , Síndrome da Liberação de Citocina/diagnóstico , Evolução Fatal , Hemorragia/diagnóstico , Hemorragia/imunologia , Hemorragia/virologia , Humanos , Masculino , Insuficiência de Múltiplos Órgãos/etiologia , Infecções por Polyomavirus/diagnóstico , Infecções por Polyomavirus/imunologia , Infecções por Polyomavirus/virologia , Índice de Gravidade de Doença , Linfócitos T/imunologia , Linfócitos T/virologia , Resultado do Tratamento , Infecções Tumorais por Vírus/diagnóstico , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia
12.
Transplantation ; 104(11): 2415-2423, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33125206

RESUMO

BACKGROUND: Although the outcome of kidney transplantation (KTx) has improved, various adverse effects of immunosuppressants and chronic rejection aggravate the long-term prognosis of patients. Therefore, the induction of immune tolerance may be an effective therapeutic strategy. METHODS: A clinical trial aiming at immune tolerance induction was conducted in kidney transplant recipients from HLA mismatched living donors by infusing autologous donor-specific regulatory T cells (Treg). To obtain Treg, recipient's peripheral blood mononuclear cells were cocultured with irradiated donor cells in the presence of anti-CD80/CD86 monoclonal antibody for 2 weeks. For preconditioning, splenectomy + cyclophosphamide (CP) was employed in the first series (group A; n = 9). In group B, splenectomy was substituted by preadministration of rituximab (group B; n = 3). In the latest cases, rituximab + rabbit antithymocyte globulin was administered instead of cyclophosphamide (group C; n = 4). Twelve days after KTx, the cultured cells were intravenously infused, and immunosuppressants were gradually tapered thereafter. RESULTS: Although mixed lymphocyte reaction was remarkably suppressed in a donor-specific fashion, 6 out of 9 patients from group A, 1 out of 3 from group B, and 1 out of 4 from group C developed acute rejection within 1 year after KTx. Complete cessation of immunosuppression was not achieved, and a small dose of immunosuppressants was continued. CONCLUSIONS: The adoptive transfer of autologous ex vivo-expanded Treg is 1 of the options to possibly induce alloimmune hyporesponsiveness. However, in the present study, further regimen optimization is still required and should be the focus of future investigations.


Assuntos
Transferência Adotiva , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Antígenos HLA/imunologia , Histocompatibilidade , Transplante de Rim , Linfócitos T Reguladores/transplante , Tolerância ao Transplante , Transferência Adotiva/efeitos adversos , Adulto , Células Cultivadas , Técnicas de Cocultura , Terapia Combinada , Feminino , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/imunologia , Humanos , Imunossupressores/uso terapêutico , Transplante de Rim/efeitos adversos , Doadores Vivos , Teste de Cultura Mista de Linfócitos , Masculino , Pessoa de Meia-Idade , Esplenectomia , Linfócitos T Reguladores/imunologia , Fatores de Tempo , Tóquio , Resultado do Tratamento , Adulto Jovem
13.
Front Immunol ; 11: 1723, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33013824

RESUMO

Liver transplantation (LT) has become the best chance and a routine practice for patients with end-stage liver disease and small hepatocellular carcinoma. However, life-long immunosuppressive regimens could lead to many post-LT complications, including cancer recurrence, infections, dysmetabolic syndrome, and renal injury. Impeccable management of immunosuppressive regimens is indispensable to ensure the best long-term prognosis for LT recipients. This is challenging for these patients, who probably have a post-LT graft survival of more than 10 or even 20 years. Approximately 20% of patients after LT could develop spontaneous operational tolerance. They could maintain normal graft function and histology without any immunosuppressive regimens. Operational tolerance after transplantation has been an attractive and ultimate goal in transplant immunology. The liver, as an immunoregulatory organ, generates an immune hyporesponsive microenvironment under physiological conditions. In this regard, LT recipients may be ideal candidates for studies focusing on operative tolerance. Cell-based strategies are one of the most promising methods for immune tolerance induction, including chimerism induced by hematopoietic stem cells and adoptive transfer of regulatory T cells, regulatory dendritic cells, regulatory macrophages, regulatory B cells, and mesenchymal stromal cells. The safety and the efficacy of many cell products have been evaluated by prospective clinical trials. In this review, we will summarize the latest perspectives on the clinical application of cell-based strategies in LT and will address a number of concerns and future directions regarding these cell products.


Assuntos
Transferência Adotiva , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Transplante de Fígado , Linfócitos T/transplante , Tolerância ao Transplante , Transferência Adotiva/efeitos adversos , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Imunossupressores/efeitos adversos , Transplante de Fígado/efeitos adversos , Medição de Risco , Fatores de Risco , Linfócitos T/imunologia , Linfócitos T/metabolismo , Quimeras de Transplante
14.
Shock ; 54(5): 586-594, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32604223

RESUMO

Coronavirus disease 2019 (COVID-19) is a life-threatening respiratory illness caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Its clinical presentation can vary from the asymptomatic state to acute respiratory distress syndrome (ARDS) and multi-organ dysfunction. Due to our insufficient understanding of its pathophysiology and lack of effective treatment, the morbidity and mortality of severe COVID-19 patients are high. Patients with COVID-19 develop ARDS fueled by exaggerated neutrophil influx into the lungs and cytokine storm. B-1a cells represent a unique subpopulation of B lymphocytes critical for circulating natural antibodies, innate immunity, and immunoregulation. These cells spontaneously produce natural IgM, interleukin (IL)-10, and granulocyte-monocyte colony stimulating factor (GM-CSF). Natural IgM neutralizes viruses and opsonizes bacteria, IL-10 attenuates the cytokine storm, and GM-CSF induces IgM production by B-1a cells in an autocrine manner. Indeed, B-1a cells have been shown to ameliorate influenza virus infection, sepsis, and pneumonia, all of which are similar to COVID-19. The recent discovery of B-1a cells in humans further reinforces their potentially critical role in the immune response against SARS-CoV-2 and their anticipated translational applications against viral and microbial infections. Given that B-1a cells protect against ARDS via immunoglobulin production and the anti-COVID-19 effects of convalescent plasma treatment, we recommend that studies be conducted to further examine the role of B-1a cells in the pathogenesis of COVID-19 and explore their therapeutic potential to treat COVID-19 patients.


Assuntos
Transferência Adotiva , Subpopulações de Linfócitos B/transplante , Betacoronavirus/patogenicidade , Infecções por Coronavirus/terapia , Pneumonia Viral/terapia , Transferência Adotiva/efeitos adversos , Animais , Subpopulações de Linfócitos B/imunologia , Betacoronavirus/imunologia , COVID-19 , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Interações Hospedeiro-Patógeno , Humanos , Pandemias , Pneumonia Viral/diagnóstico , Pneumonia Viral/imunologia , Pneumonia Viral/virologia , SARS-CoV-2
15.
Front Immunol ; 11: 992, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32528477

RESUMO

Despite recent advances, the eradication of cancers still represents a challenge which justifies the exploration of additional therapeutic strategies such as immunotherapies, including adoptive cell transfers. Human peripheral Vγ9Vδ2 T cells, which constitute a major transitional immunity lymphocyte subset, represent attractive candidates because of their broad and efficient anti-tumor functions, as well as their lack of alloreactivity and easy handling. Vγ9Vδ2 T cells act like immune cell stress sensors that can, in a tightly controlled manner but through yet incompletely understood mechanisms, detect subtle changes of levels of phosphorylated metabolites of isoprenoid synthesis pathways. Consequently, various anti-tumor immunotherapeutic strategies have been proposed to enhance their reactivity and cytotoxicity, as well as to reduce the deleterious events. In this review, we expose these advances based on different strategies and their validation in preclinical models. Importantly, we next discuss advantages and limits of each approach, by highlighting the importance of the use of relevant preclinical model for evaluation of safety and efficacy. Finally, we propose novel perspectives and strategies that should be explored using these models for therapeutic improvements.


Assuntos
Transferência Adotiva , Antineoplásicos Imunológicos/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/transplante , Transferência Adotiva/efeitos adversos , Animais , Antineoplásicos Imunológicos/efeitos adversos , Vacinas Anticâncer/efeitos adversos , Humanos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Fenótipo , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Pesquisa Translacional Biomédica , Resultado do Tratamento
17.
Eur J Immunol ; 50(7): 1034-1043, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32130733

RESUMO

IL-9 is involved in various T cell-dependent inflammatory models including colitis, encepahlitis, and asthma. However, the regulation and specificity of IL-9 responsiveness by T cells during immune responses remains poorly understood. Here, we addressed this question using two different models: experimental colitis induced by transfer of naive CD4+ CD45RBhigh T cells into immunodeficient mice, and OVA-specific T cell activation. In the colitis model, constitutive IL-9 expression exacerbated inflammation upon transfer of CD4+ CD45RBhigh T cells from WT but not from Il9r-/- mice, indicating that IL-9 acts directly on T cells. Suprisingly, such naïve CD4+ CD45RBhigh T cells failed to express the Il9r or respond to IL-9 in vitro, in contrast with CD4+ CD45RBlow T cells. By using OVA-specific T cells, we observed that T cells acquired the capacity to respond to IL-9 along with CD44 upregulation, after long-lasting (5 to 12 days) in vivo antigenic stimulation. Il9r expression was associated with Th2 and Th17 phenotypes. Interestingly, in contrast to the IL-2 response, antigen restimulation downregulated IL-9 responsiveness. Taken together, our results demonstrate that IL-9 does not act on naïve T cells but that IL-9 responsiveness is acquired by CD4+ T cells after in vivo activation and acquisition of memory markers such as CD44.


Assuntos
Transferência Adotiva/efeitos adversos , Colite/imunologia , Interleucina-9/imunologia , Células Th17/imunologia , Células Th2/imunologia , Animais , Colite/etiologia , Colite/genética , Colite/patologia , Modelos Animais de Doenças , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Interleucina-9/genética , Camundongos , Camundongos Knockout , Camundongos SCID , Receptores de Interleucina-9/genética , Receptores de Interleucina-9/imunologia , Células Th17/patologia , Células Th17/transplante , Células Th2/patologia , Células Th2/transplante
18.
Cell Immunol ; 349: 104049, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32057353

RESUMO

Pathogenic microorganisms utilize multiple approaches to break down host immunity in favor of their invasion, of which, cystatin C is one of the soluble factors secreted by parasites reported to affect host immunity in vivo. The cellular targets and mechanisms of action in vivo of cystatin C, however, are far from clear. As professional antigen-presenting cells, dendritic cells (DCs) are first immune cells that contact foreign pathogenic agents or their products to initiate immune responses. We previously reported that cystatin C can regulate the functions of DCs in terms of suppressed CD4+ T cell activation but enhanced Th1/Th17 differentiation via different mechanisms. Here, we further verified these regulatory effects of cystatin C on DCs in vivo. We found that the suppressive role of DC-mediated CD4+ T cell proliferation by cystatin C was partly cell-contact independent and extended to CD8+ T cells in vivo. Although cystatin C-overexpressing DCs trafficked equally as their mock-transduced counterparts, their adoptive transfer suppressed CD8+ T cell immunity and resulted in compromised tumor rejection in both vaccination and treatment regimes. Compared with their role in promoting Th17 differentiation in vivo, cystatin C-transduced DCs had far greater ability to induce T regulatory cells (Tregs), leading to collectively a higher Treg/Th17 ratio in an adoptively transferred disease model, and thus relieved Th17-dependent autoimmunity. Collectively, these data demonstrated strong in vivo evidences for immune regulatory roles of cystatin C in DCs and provided theoretical basis for the application of cystatin C-transduced cell therapy in the treatment or remission of certain autoimmune diseases. (246).


Assuntos
Transferência Adotiva , Artrite Experimental/terapia , Doenças Autoimunes/terapia , Cistatina C/fisiologia , Células Dendríticas/imunologia , Evasão Tumoral/imunologia , Transferência Adotiva/efeitos adversos , Animais , Comunicação Celular , Células Cultivadas , Cistatina C/genética , Células Dendríticas/transplante , Regulação para Baixo , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Granzimas/biossíntese , Granzimas/genética , Imunoterapia Adotiva , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Ativação Linfocitária , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Ovalbumina/imunologia , Proteínas Citotóxicas Formadoras de Poros/biossíntese , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Recombinantes/metabolismo , Organismos Livres de Patógenos Específicos , Especificidade do Receptor de Antígeno de Linfócitos T , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Transdução Genética
20.
Biol Blood Marrow Transplant ; 26(1): 34-43, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31605820

RESUMO

CD19-targeted chimeric antigen receptor (CAR) modified T cell immunotherapy is a novel treatment with promising results in patients with relapsed/refractory lymphoid malignancies. CAR T cell therapy has known early toxicities of cytokine release syndrome and neurotoxicity, but little is known about long-term neuropsychiatric adverse effects. We have used patient-reported outcomes, including Patient-Reported Outcomes Measurement Information System (PROMIS) measures, to assess neuropsychiatric and other patient-reported outcomes of 40 patients with relapse/refractory chronic lymphocytic leukemia, non-Hodgkin lymphoma, and acute lymphoblastic leukemia 1 to 5 years after treatment with CD19-targeted CAR T cells. Mean T scores of PROMIS domains of global mental health, global physical health, social function, anxiety, depression, fatigue, pain, and sleep disturbance were not clinically meaningfully different from the mean in the general US population. However, 19 patients (47.5%) reported at least 1 cognitive difficulty and/or clinically meaningful depression and/or anxiety, and 7 patients (17.5%) scored ≤40 in global mental health, indicating at least 1 standard deviation worse than the general population mean. Younger age was associated with worse long-term global mental health (P = .02), anxiety (P = .001), and depression (P= .01). Anxiety before CAR T cell therapy was associated with increased likelihood of anxiety after CAR T cell therapy (P = .001). Fifteen patients (37.5%) reported cognitive difficulties after CAR T cell therapy. Depression before CAR T cell therapy was statistically significantly associated with higher likelihood of self-reported post-CAR T cognitive difficulties (P = .02), and there was a trend for an association between acute neurotoxicity and self-reported post-CAR T cognitive difficulties (P = .08). Having more post-CAR T cognitive difficulties was associated with worse global mental health and global physical health. Our study demonstrates overall good neuropsychiatric outcomes in 40 long-term survivors after CAR T cell therapy. However, nearly 50% of patients in the cohort reported at least 1 clinically meaningful negative neuropsychiatric outcome (anxiety, depression, or cognitive difficulty), indicating that a significant number of patients would likely benefit from mental health services following CAR T cell therapy. Younger age, pre-CAR T anxiety or depression, and acute neurotoxicity may be risk factors for long-term neuropsychiatric problems in this patient population. Larger studies are needed to confirm these findings.


Assuntos
Transferência Adotiva/efeitos adversos , Leucemia Linfocítica Crônica de Células B/terapia , Linfoma não Hodgkin/terapia , Transtornos Neurocognitivos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos Quiméricos/administração & dosagem , Autorrelato , Adulto , Idoso , Feminino , Seguimentos , Humanos , Leucemia Linfocítica Crônica de Células B/epidemiologia , Leucemia Linfocítica Crônica de Células B/psicologia , Linfoma não Hodgkin/epidemiologia , Linfoma não Hodgkin/psicologia , Masculino , Pessoa de Meia-Idade , Transtornos Neurocognitivos/epidemiologia , Transtornos Neurocognitivos/etiologia , Transtornos Neurocognitivos/psicologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...