Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Hum Immunol ; 83(1): 86-98, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34583856

RESUMEN

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.


Asunto(s)
Traslado Adoptivo , COVID-19/terapia , Síndrome de Liberación de Citoquinas/terapia , Citocinas/inmunología , Células Asesinas Naturales/trasplante , SARS-CoV-2/inmunología , Traslado Adoptivo/efectos adversos , Animales , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/virología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/metabolismo , Síndrome de Liberación de Citoquinas/virología , Citocinas/metabolismo , Citotoxicidad Inmunológica , Interacciones Huésped-Patógeno , Humanos , Memoria Inmunológica , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/virología , SARS-CoV-2/patogenicidad , Resultado del Tratamiento
2.
Front Immunol ; 12: 727814, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925312

RESUMEN

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.


Asunto(s)
Traslado Adoptivo/efectos adversos , Células Alogénicas/inmunología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/terapia , Trasplante de Corazón/efectos adversos , Herpesvirus Humano 4/inmunología , Tumor de Músculo Liso/complicaciones , Tumor de Músculo Liso/terapia , Linfocitos T/inmunología , Traslado Adoptivo/métodos , Infecciones por Virus de Epstein-Barr/virología , Femenino , Humanos , Trastornos Linfoproliferativos/etiología , Tumor de Músculo Liso/etiología , Trasplante Homólogo , Resultado del Tratamiento , Viremia/complicaciones , Viremia/terapia , Adulto Joven
3.
Front Immunol ; 12: 734246, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34691041

RESUMEN

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.


Asunto(s)
Traslado Adoptivo , Caspasa 9/biosíntesis , Antígenos de la Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/terapia , Linfocitos T/trasplante , Traslado Adoptivo/efectos adversos , Animales , Caspasa 9/genética , Muerte Celular , Línea Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Citotoxicidad Inmunológica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Inducción Enzimática , Femenino , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/metabolismo , Hepatitis B Crónica/virología , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Simplexvirus/enzimología , Simplexvirus/genética , Linfocitos T/enzimología , Linfocitos T/inmunología , Linfocitos T/patología , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Transducción Genética
4.
BMC Cancer ; 21(1): 877, 2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-34332557

RESUMEN

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 .


Asunto(s)
Traslado Adoptivo , Carcinoma de Pulmón de Células no Pequeñas/terapia , Gefitinib/uso terapéutico , Neoplasias Pulmonares/terapia , Mutación , Células T Asesinas Naturales/inmunología , Traslado Adoptivo/efectos adversos , Traslado Adoptivo/métodos , Antígeno B7-H1/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/etiología , Terapia Combinada , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Gefitinib/administración & dosificación , Gefitinib/efectos adversos , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/etiología , Terapia Molecular Dirigida , Células T Asesinas Naturales/metabolismo , Resultado del Tratamiento
5.
Front Immunol ; 12: 625998, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717141

RESUMEN

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.


Asunto(s)
Traslado Adoptivo , Rechazo de Injerto/terapia , Supervivencia de Injerto , Trasplante de Riñón/efectos adversos , Trasplante de Células Madre Mesenquimatosas , Células Mieloides/trasplante , Traslado Adoptivo/efectos adversos , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Humanos , Inmunosupresores/uso terapéutico , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/trasplante , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Resultado del Tratamiento
6.
Front Immunol ; 12: 624685, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679769

RESUMEN

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.


Asunto(s)
Traslado Adoptivo , Desensibilización Inmunológica , Esclerosis Múltiple/terapia , Proteínas de la Mielina/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Vacunación , Vacunas/uso terapéutico , Traslado Adoptivo/efectos adversos , Traslado Adoptivo/historia , Traslado Adoptivo/tendencias , Animales , Autoinmunidad , Desensibilización Inmunológica/efectos adversos , Desensibilización Inmunológica/historia , Desensibilización Inmunológica/tendencias , Difusión de Innovaciones , Predicción , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Tolerancia Inmunológica , Esclerosis Múltiple/historia , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Proteínas de la Mielina/efectos adversos , Proteínas de la Mielina/inmunología , Proteínas de la Mielina/metabolismo , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Vacunación/efectos adversos , Vacunación/historia , Vacunación/tendencias , Vacunas/efectos adversos
7.
Int Immunopharmacol ; 91: 107322, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33418238

RESUMEN

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.


Asunto(s)
Traslado Adoptivo , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Macrófagos/trasplante , Trasplante de Órganos , Tolerancia al Trasplante , Traslado Adoptivo/efectos adversos , Animales , Terapia Combinada , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Supervivencia de Injerto/efectos de los fármacos , Humanos , Inmunosupresores/uso terapéutico , Macrófagos/inmunología , Macrófagos/metabolismo , Trasplante de Órganos/efectos adversos , Fenotipo , Tolerancia al Trasplante/efectos de los fármacos , Resultado del Tratamiento
8.
Ann Neurol ; 89(4): 769-779, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33459417

RESUMEN

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.


Asunto(s)
Traslado Adoptivo/métodos , Virus JC , Leucoencefalopatía Multifocal Progresiva/terapia , Linfocitos T , Adolescente , Traslado Adoptivo/efectos adversos , Anciano , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Humanos , Leucoencefalopatía Multifocal Progresiva/líquido cefalorraquídeo , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Carga Viral
9.
Front Immunol ; 12: 801281, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975916

RESUMEN

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.


Asunto(s)
Traslado Adoptivo/efectos adversos , Virus BK/patogenicidad , Cistitis/terapia , Síndrome de Liberación de Citoquinas/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Hemorragia/terapia , Infecciones por Polyomavirus/terapia , Linfocitos T/trasplante , Infecciones Tumorales por Virus/terapia , Adolescente , Virus BK/inmunología , Cistitis/diagnóstico , Cistitis/inmunología , Cistitis/virología , Síndrome de Liberación de Citoquinas/diagnóstico , Resultado Fatal , Hemorragia/diagnóstico , Hemorragia/inmunología , Hemorragia/virología , Humanos , Masculino , Insuficiencia Multiorgánica/etiología , Infecciones por Polyomavirus/diagnóstico , Infecciones por Polyomavirus/inmunología , Infecciones por Polyomavirus/virología , Índice de Severidad de la Enfermedad , Linfocitos T/inmunología , Linfocitos T/virología , Resultado del Tratamiento , Infecciones Tumorales por Virus/diagnóstico , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
10.
Int Immunopharmacol ; 90: 107195, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33278746

RESUMEN

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.


Asunto(s)
Aborto Espontáneo/prevención & control , Traslado Adoptivo , Tolerancia Inmunológica , Linfocitos T Reguladores/trasplante , Aborto Espontáneo/inmunología , Aborto Espontáneo/metabolismo , Traslado Adoptivo/efectos adversos , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Fenotipo , Embarazo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
11.
Front Immunol ; 11: 1723, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33013824

RESUMEN

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.


Asunto(s)
Traslado Adoptivo , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Trasplante de Hígado , Linfocitos T/trasplante , Tolerancia al Trasplante , Traslado Adoptivo/efectos adversos , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Inmunosupresores/efectos adversos , Trasplante de Hígado/efectos adversos , Medición de Riesgo , Factores de Riesgo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Quimera por Trasplante
12.
Shock ; 54(5): 586-594, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32604223

RESUMEN

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.


Asunto(s)
Traslado Adoptivo , Subgrupos de Linfocitos B/trasplante , Betacoronavirus/patogenicidad , Infecciones por Coronavirus/terapia , Neumonía Viral/terapia , Traslado Adoptivo/efectos adversos , Animales , Subgrupos de Linfocitos B/inmunología , Betacoronavirus/inmunología , COVID-19 , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Interacciones Huésped-Patógeno , Humanos , Pandemias , Neumonía Viral/diagnóstico , Neumonía Viral/inmunología , Neumonía Viral/virología , SARS-CoV-2
13.
Front Immunol ; 11: 992, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32528477

RESUMEN

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.


Asunto(s)
Traslado Adoptivo , Antineoplásicos Inmunológicos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/trasplante , Traslado Adoptivo/efectos adversos , Animales , Antineoplásicos Inmunológicos/efectos adversos , Vacunas contra el Cáncer/efectos adversos , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Fenotipo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Investigación Biomédica Traslacional , Resultado del Tratamiento
15.
Cell Immunol ; 349: 104049, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32057353

RESUMEN

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).


Asunto(s)
Traslado Adoptivo , Artritis Experimental/terapia , Enfermedades Autoinmunes/terapia , Cistatina C/fisiología , Células Dendríticas/inmunología , Escape del Tumor/inmunología , Traslado Adoptivo/efectos adversos , Animales , Comunicación Celular , Células Cultivadas , Cistatina C/genética , Células Dendríticas/trasplante , Regulación hacia Abajo , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Granzimas/biosíntesis , Granzimas/genética , Inmunoterapia Adoptiva , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ovalbúmina/inmunología , Proteínas Citotóxicas Formadoras de Poros/biosíntesis , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Recombinantes/metabolismo , Organismos Libres de Patógenos Específicos , Especificidad del Receptor de Antígeno de Linfocitos T , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Transducción Genética
17.
Biol Blood Marrow Transplant ; 26(1): 34-43, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31605820

RESUMEN

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.


Asunto(s)
Traslado Adoptivo/efectos adversos , Leucemia Linfocítica Crónica de Células B/terapia , Linfoma no Hodgkin/terapia , Trastornos Neurocognitivos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores Quiméricos de Antígenos/administración & dosificación , Autoinforme , Adulto , Anciano , Femenino , Estudios de Seguimiento , Humanos , Leucemia Linfocítica Crónica de Células B/epidemiología , Leucemia Linfocítica Crónica de Células B/psicología , Linfoma no Hodgkin/epidemiología , Linfoma no Hodgkin/psicología , Masculino , Persona de Mediana Edad , Trastornos Neurocognitivos/epidemiología , Trastornos Neurocognitivos/etiología , Trastornos Neurocognitivos/psicología , Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/psicología
18.
Mol Ther ; 28(1): 64-74, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31636040

RESUMEN

Generation of an optimal T cell therapeutic expressing high frequencies of transgenic T cell receptor (tgTCR) is essential for improving TCR gene therapy. Upon TCR gene transfer, presence of endogenous TCRαß reduces expression of tgTCR due to TCR mixed-dimer formation and competition for binding CD3. Knockout (KO) of endogenous TCRαß was recently achieved using CRISPR/Cas9 editing of the TRAC or TRBC loci, resulting in increased expression and function of tgTCR. Here, we adopt this approach into current protocols for generating T cell populations expressing tgTCR to validate this strategy in the context of four clinically relevant TCRs. First, simultaneous editing of TRAC and TRBC loci was reproducible and resulted in high double KO efficiencies in bulk CD8 T cells. Next, tgTCR expression was significantly higher in double TRAC/BC KO conditions for all TCRs tested, including those that contained structural modifications to encourage preferential pairing. Finally, increased expression of tgTCR in edited T cell populations allowed for increased recognition of antigen expressing tumor targets and prolonged control of tumor outgrowth in a preclinical model of multiple myeloma. In conclusion, CRISPR/Cas9-mediated KO of both endogenous TCRαß chains can be incorporated in current T cell production protocols and is preferential to ensure an improved and safe clinical therapeutic.


Asunto(s)
Traslado Adoptivo/métodos , Sistemas CRISPR-Cas , Edición Génica/métodos , Terapia Genética/métodos , Mieloma Múltiple/terapia , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Traslado Adoptivo/efectos adversos , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos , Femenino , Genes Codificadores de los Receptores de Linfocitos T , Terapia Genética/efectos adversos , Voluntarios Sanos , Humanos , Células K562 , Masculino , Ratones , Ratones Endogámicos NOD , Mieloma Múltiple/patología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Transducción Genética , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Front Immunol ; 10: 2580, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31787975

RESUMEN

The safe utilization of induced pluripotent stem cell (iPSC) derivatives in clinical use is attributed to the complete elimination of the risk of forming teratomas after transplantation. The extent by which such a risk exists in immune-competent hosts is mostly unknown. Here, using humanized mice reconstituted with fetal hematopoietic stem cells and autologous thymus tissue (bone-liver-thymus humanized mice [Hu-BLT]) or following the adoptive transfer of peripheral blood mononuclear cells(PBMCs) (Hu-AT), we evaluated the capacity of immune cells to prevent or eliminate teratomas derived from human iPSCs (hiPSCs). Our results showed that the injection of hiPSCs failed to form teratomas in Hu-AT mice reconstituted with allogeneic or autologous PBMCs or purified natural killer (NK) cells alone. However, teratomas were observed in Hu-AT mice reconstituted with autologous PBMCs depleted from NK cells. In line with these results, Hu-BLT, which do not have functional NK cells, could not prevent the growth of teratomas. Finally, we found that established teratomas were not targeted by NK cells and instead were efficiently rejected by allogeneic but not autologous T cells in Hu-AT mice. Overall, our findings suggest that autologous hiPSC-derived therapies are unlikely to form teratomas in the presence of NK cells.


Asunto(s)
Células Asesinas Naturales/inmunología , Células Madre Pluripotentes/inmunología , Teratoma/prevención & control , Traslado Adoptivo/efectos adversos , Adulto , Animales , Humanos , Leucocitos Mononucleares/trasplante , Ratones , Ratones Endogámicos NOD , Ratones SCID , Linfocitos T/inmunología , Teratoma/etiología , Teratoma/inmunología , Trasplante Heterólogo
20.
Front Immunol ; 10: 2489, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31736946

RESUMEN

Epstein-Barr virus (EBV)-associated post-transplant lymphoproliferative disease (PTLD) is a life-threatening complication of T-lymphocyte deplete allogeneic hematopoietic stem cell transplantation (allo-HSCT). For patients with PTLD refractory to Rituximab, donor lymphocyte infusion (DLI) is established as a successful option for salvage therapy. However, although in vivo lymphocyte expansion has been correlated with good clinical outcome following DLI, the specificity and functional characteristics of EBV-specific T-cell responses remain poorly characterized. Here we describe two patients with Rituximab-refractory PTLD complicating T-cell deplete allo-HSCT, both of whom were successfully rescued with 1 × 106/Kg unselected stem cell donor-derived DLI. Prospective analyses revealed that complete clinical and radiological responses were associated with in vivo expansion of T and NK cells. Furthermore, EBV MHC tetramer, and interferon gamma analyses revealed a marked increase in EBV-specific T-cell frequency from 4 weeks after DLI. Reactivity was demonstrated against a range of EBV latent and lytic antigens, including those detected in tumor biopsy material. The immunodominant EBV-specific T cell response expanding in vivo following infusion matched the dominant response present in the DLI preparations prior to administration. Furthermore, differences in the repertoire of subdominant antigen-specific T-cells were also detected, suggesting that antigen-encounter in vivo can shape the immune response. These results demonstrate the value of prospectively studying in vivo T-cell responses, by facilitating the identification of important specificities required for clinical efficacy. Applying this approach on a larger scale promises to yield data which may be essential for the optimization of future adoptive immunotherapeutic strategies for PTLD.


Asunto(s)
Traslado Adoptivo , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/inmunología , Trastornos Linfoproliferativos/etiología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/inmunología , Traslado Adoptivo/efectos adversos , Traslado Adoptivo/métodos , Susceptibilidad a Enfermedades , Epítopos de Linfocito T/inmunología , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Inmunohistoquímica , Inmunofenotipificación , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Trastornos Linfoproliferativos/diagnóstico , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones , Linfocitos T/metabolismo , Donantes de Tejidos , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA