Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 143(25): 2599-2611, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38493479

RESUMEN

ABSTRACT: Chimeric antigen receptor (CAR)-redirected immune cells hold significant therapeutic potential for oncology, autoimmune diseases, transplant medicine, and infections. All approved CAR-T therapies rely on personalized manufacturing using undirected viral gene transfer, which results in nonphysiological regulation of CAR-signaling and limits their accessibility due to logistical challenges, high costs and biosafety requirements. Random gene transfer modalities pose a risk of malignant transformation by insertional mutagenesis. Here, we propose a novel approach utilizing CRISPR-Cas gene editing to redirect T cells and natural killer (NK) cells with CARs. By transferring shorter, truncated CAR-transgenes lacking a main activation domain into the human CD3ζ (CD247) gene, functional CAR fusion-genes are generated that exploit the endogenous CD3ζ gene as the CAR's activation domain. Repurposing this T/NK-cell lineage gene facilitated physiological regulation of CAR expression and redirection of various immune cell types, including conventional T cells, TCRγ/δ T cells, regulatory T cells, and NK cells. In T cells, CD3ζ in-frame fusion eliminated TCR surface expression, reducing the risk of graft-versus-host disease in allogeneic off-the-shelf settings. CD3ζ-CD19-CAR-T cells exhibited comparable leukemia control to TCRα chain constant (TRAC)-replaced and lentivirus-transduced CAR-T cells in vivo. Tuning of CD3ζ-CAR-expression levels significantly improved the in vivo efficacy. Notably, CD3ζ gene editing enabled redirection of NK cells without impairing their canonical functions. Thus, CD3ζ gene editing is a promising platform for the development of allogeneic off-the-shelf cell therapies using redirected killer lymphocytes.


Asunto(s)
Complejo CD3 , Células Asesinas Naturales , Receptores Quiméricos de Antígenos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Humanos , Complejo CD3/genética , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Animales , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Citotoxicidad Inmunológica , Inmunoterapia Adoptiva/métodos , Edición Génica/métodos , Sistemas CRISPR-Cas , Ratones Endogámicos NOD
3.
PLoS Pathog ; 16(7): e1008560, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32667948

RESUMEN

Human cytomegalovirus (HCMV) causes serious complications to immune compromised hosts. Dendritic cells (iDCgB) expressing granulocyte-macrophage colony-stimulating factor, interferon-alpha and HCMV-gB were developed to promote de novo antiviral adaptive responses. Mice reconstituted with a human immune system (HIS) were immunized with iDCgB and challenged with HCMV, resulting into 93% protection. Immunization stimulated the expansion of functional effector memory CD8+ and CD4+ T cells recognizing gB. Machine learning analyses confirmed bone marrow T/CD4+, liver B/IgA+ and spleen B/IgG+ cells as predictive biomarkers of immunization (≈87% accuracy). CD8+ and CD4+ T cell responses against gB were validated. Splenic gB-binding IgM-/IgG+ B cells were sorted and analyzed at a single cell level. iDCgB immunizations elicited human-like IgG responses with a broad usage of various IgG heavy chain V gene segments harboring variable levels of somatic hypermutation. From this search, two gB-binding human monoclonal IgGs were generated that neutralized HCMV infection in vitro. Passive immunization with these antibodies provided proof-of-concept evidence of protection against HCMV infection. This HIS/HCMV in vivo model system supported the validation of novel active and passive immune therapies for future clinical translation.


Asunto(s)
Anticuerpos Antivirales/inmunología , Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/inmunología , Inmunización Pasiva , Inmunoglobulina G/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Antígenos Virales/inmunología , Citomegalovirus/inmunología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina G/farmacología , Ratones
4.
Am J Pathol ; 189(3): 521-539, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30593822

RESUMEN

Humanized mice developing functional human T cells endogenously and capable of recognizing cognate human leukocyte antigen-matched tumors are emerging as relevant models for studying human immuno-oncology in vivo. Herein, mice transplanted with human CD34+ stem cells and bearing endogenously developed human T cells for >15 weeks were infected with an oncogenic recombinant Epstein-Barr virus (EBV), encoding enhanced firefly luciferase and green fluorescent protein. EBV-firefly luciferase was detectable 1 week after infection by noninvasive optical imaging in the spleen, from where it spread rapidly and systemically. EBV infection resulted into a pronounced immunologic skewing regarding the expansion of CD8+ T cells in the blood outnumbering the CD4+ T and CD19+ B cells. Furthermore, within 10 weeks of infections, mice developing EBV-induced tumors had significantly higher absolute numbers of CD8+ T cells in lymphatic tissues than mice controlling tumor development. Tumor outgrowth was paralleled by an up-regulation of the programmed cell death receptor 1 on CD8+ and CD4+ T cells, indicative for T-cell dysfunction. Histopathological examinations and in situ hybridizations for EBV in tumors, spleen, liver, and kidney revealed foci of EBV-infected cells in perivascular regions in close association with programmed cell death receptor 1-positive infiltrating lymphocytes. The strong spatiotemporal correlation between tumor development and the T-cell dysfunctional status seen in this viral oncogenesis humanized model replicates observations obtained in the clinical setting.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Infecciones por Virus de Epstein-Barr/patología , Humanos , Activación de Linfocitos , Ratones , Ratones Mutantes , Neoplasias/patología , Neoplasias/virología
5.
Cancer Immunol Immunother ; 68(11): 1891-1899, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31628525

RESUMEN

Acute myeloid leukemia (AML) is the most common acute leukemia in adults and overall survival remains poor. Chemotherapy is the standard of care for intensive induction therapy. Patients who achieve a complete remission require post-remission therapies to prevent relapse. There is no standard of care for patients with minimal residual disease (MRD), and stem cell transplantation is a salvage therapy. Considering the AML genetic heterogeneity and the leukemia immune-suppressive properties, novel cellular immune therapies to effectively harness immunological responses to prevent relapse are needed. We developed a novel modality of immune therapy consisting of monocytes reprogrammed with lentiviral vectors expressing GM-CSF, IFN-α and antigens. Preclinical studies in humanized mice showed that the reprogrammed monocytes self-differentiated into highly viable induced dendritic cells (iDCs) in vivo which migrated effectively to lymph nodes, producing remarkable effects in the de novo regeneration of T and B cell responses. For the first-in-man clinical trial, the patient's monocytes will be transduced with an integrase-defective tricistronic lentiviral vector expressing GM-CSF, IFN-α and a truncated WT1 antigen. For transplanted patients, pre-clinical development of iDCs co-expressing cytomegalovirus antigens is ongoing. To simplify the product chain for a de-centralized supply model, we are currently exploring a closed automated system for a short two-day manufacturing of iDCs. A phase I clinical trial study is in preparation for immune therapy of AML patients with MRD. The proposed cell therapy can fill an important gap in the current and foreseeable future immunotherapies of AML.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Vectores Genéticos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunoterapia , Interferón-alfa/metabolismo , Leucemia Mieloide Aguda/terapia , Monocitos/inmunología , Células Dendríticas/inmunología , Humanos , Lentivirus/genética , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Medicina de Precisión , Trasplante de Células Madre
6.
Am J Pathol ; 187(6): 1380-1398, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28432872

RESUMEN

Humanized mice engrafted with human hematopoietic stem cells and developing functional human T-cell adaptive responses are in critical demand to test human-specific therapeutics. We previously showed that humanized mice immunized with long-lived induced-dendritic cells loaded with the pp65 viral antigen (iDCpp65) exhibited a faster development and maturation of T cells. Herein, we evaluated these effects in a long-term (36 weeks) nonclinical model using two stem cell donors to assess efficacy and safety. Relative to baseline, iDCpp65 immunization boosted the output of effector memory CD4+ T cells in peripheral blood and lymph nodes. No weight loss, human malignancies, or systemic graft-versus-host (GVH) disease were observed. However, for one reconstitution cohort, some mice immunized with iDCpp65 showed GVH-like signs on the skin. Histopathology analyses of the inflamed skin revealed intrafollicular and perifollicular human CD4+ cells near F4/80+ mouse macrophages around hair follicles. In spleen, CD4+ cells formed large clusters surrounded by mouse macrophages. In plasma, high levels of human T helper 2-type inflammatory cytokines were detectable, which activated in vitro the STAT5 pathway of murine macrophages. Despite this inflammatory pattern, human CD8+ T cells from mice with GVH reacted against the pp65 antigen in vitro. These results uncover a dynamic cross-species interaction between human memory T cells and mouse macrophages in the skin and lymphatic tissues of humanized mice.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Macrófagos/inmunología , Piel/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Antígenos CD34/análisis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular/inmunología , Línea Celular , Citocinas/sangre , Proteínas del Citoesqueleto , Células Dendríticas/trasplante , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/patología , Trasplante de Células Madre Hematopoyéticas , Xenoinjertos , Ratones Endogámicos NOD , Proteínas de Microfilamentos , Fosfoproteínas/inmunología , Piel/patología
7.
J Cell Physiol ; 232(7): 1669-1680, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28019659

RESUMEN

Following superficial injury, neighbouring gastric epithelial cells close the wound by rapid cell migration, a process called epithelial restitution. Na+ /H+ exchange (NHE) inhibitors interfere with restitution, but the role of the different NHE isoforms expressed in gastric pit cells has remained elusive. The role of the basolaterally expressed NHE1 (Slc9a1) and the presumably apically expressed NHE2 (Slc9a2) in epithelial restitution was investigated in the nontransformed rat gastric surface cell line RGM1. Migration velocity was assessed by loading the cells with the fluorescent dye DiR and following closure of an experimental wound over time. Since RGM1 cells expressed very low NHE2 mRNA and have low transport activity, NHE2 was introduced by lentiviral gene transfer. In medium with pH 7.4, RGM1 cells displayed slow wound healing even in the absence of growth factors and independently of NHE activity. Growth factors accelerated wound healing in a partly NHE1-dependent fashion. Preincubation with acidic pH 7.1 stimulated restitution in a NHE1-dependent fashion. When pH 7.1 was maintained during the restitution period, migratory speed was reduced to ∼10% of the speed at pH 7,4, and the residual restitution was further inhibited by NHE1 inhibition. Lentiviral NHE2 expression increased the steady-state pHi and reduced the restitution velocity after low pH preincubation, which was reversible by pharmacological NHE2 inhibition. The results demonstrate that in RGM1 cells, migratory velocity is increased by NHE1 activation, while NHE2 activity inhibit this process. A differential activation of NHE1 and NHE2 may therefore, play a role in the initiation and completion of the epithelial restitution process.


Asunto(s)
Movimiento Celular , Mucosa Gástrica/citología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Línea Celular , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Lentivirus/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/genética , Cicatrización de Heridas
8.
Virus Genes ; 53(5): 723-732, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28744797

RESUMEN

Lentiviral vectors (LVs) developed in the past two decades for research and pre-clinical purposes have entered clinical trials with remarkable safety and efficacy performances. Development and clinical testing of LVs for improvement of human immunity showed major advantages in comparison to other viral vector systems. Robust and persisted transduction efficiency of blood cells with LVs, resulted into a broad range of target cells for immune therapeutic approaches: from hematopoietic stem cells and precursor cells for correction of immune deficiencies, up to effector lymphoid and myeloid cells. T cells engineered for expression of chimeric antigen receptors (CARs) or epitope-specific transgenic T cell receptors (TCRs) are in several cancer immune therapy clinical trials worldwide. Development of engineered dendritic cells is primed for clinical trials for cancer and chronic infections. Technological adaptations for ex vivo cell manipulations are here discussed and presented based on properties and uses of the target cell. For future development of off-shelf immune therapies, direct in vivo administration of lentiviral vectors is warranted and intended. Approaches for lentiviral in vivo targeting to maximize immune therapeutic success are discussed.


Asunto(s)
Vectores Genéticos/inmunología , Sistema Inmunológico/inmunología , Sistema Inmunológico/virología , Lentivirus/inmunología , Animales , Células Dendríticas/inmunología , Células Dendríticas/virología , Terapia Genética/métodos , Humanos , Linfocitos T/inmunología , Linfocitos T/virología , Transducción Genética/métodos
9.
Retrovirology ; 13: 15, 2016 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-26969150

RESUMEN

BACKGROUND: LILRA3 is an immunostimulatory molecule which can conditionally induce the proliferation of cytotoxic cells. LILRA3 has a deletion genotype which is associated with multiple immune disorders. In this study, we wanted to analyze the regulation of LILRA3 and its significance in the context of HIV infection. RESULTS: We analyzed a panel of TLR agonists and found that ssRNA40, a TLR8 agonist, is a potent inducer of LILRA3 in healthy individuals. However, this regulation is much diminished in HIV. Comparison of TLR8 to TLR4 induction of LILRA3 indicated that LPS induces less LILRA3 than ssRNA40 among healthy controls, but not HIV patients. Levels of LILRA3 induction correlated to virus load and CD4 counts in untreated patients. Recombinant LILRA3 can induce a host of proinflammatory genes which include IL-6 and IL-1α, as well as alter the expression of MHC and costimulatory molecules in monocytes and B-cells. CONCLUSION: Our experiments point towards a beneficial role for LILRA3 in virus infections, especially in ssRNA viruses, like HIV, that engage TLR8. However, the potentially beneficial role of LILRA3 is abrogated during a HIV infection. We believe that more work has to be done to study the role of LILRA3 in infectious diseases and that there is a potential for exploring the use of LILRA3 in the treatment of virus infections.


Asunto(s)
Regulación de la Expresión Génica , Infecciones por VIH/patología , VIH/aislamiento & purificación , Monocitos/inmunología , Receptores Inmunológicos/metabolismo , Carga Viral , Adulto , Recuento de Linfocito CD4 , Femenino , Infecciones por VIH/inmunología , Humanos , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Receptor Toll-Like 8/metabolismo
10.
Cytotherapy ; 18(4): 570-80, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26971685

RESUMEN

BACKGROUND AIMS: Dendritic cell (DC)-tumor cell hybrids have been used clinically in cancer immunotherapy, but their advantage over the simple mixture of tumor cells and DCs is still a matter of controversy. In this study, we compared DC-tumor cell hybrids with the non-fused mixture of DC and tumor cells directly in their ability to induce a specific immune response. METHODS: Hybrids were obtained by electrofusion of tumor cells and monocyte-derived DCs. Cell phenotype was evaluated by flow cytometry and antigen-presenting ability by co-culture with syngeneic T cells followed by tetramer analysis and interferon (IFN)-γ ELISPOT. RESULTS: Less than half the cells in the mixture expressed DC co-stimulatory molecules. Furthermore, DCs in the mixture had significantly lower expression of MHC class I molecules than DCs in the fusion. Conversely, nearly all CD11c(+)Her2/neu(+) hybrids expressed CD80, CD86, CD83, HLA-DR and MHC class I from both tumor cells and DCs. Using tumor cells constitutively expressing a cytomegalovirus (CMV) antigen, we show that expansion of CMV-specific cytotoxic T lymphocytes (CTLs) restricted by DCs' MHC class I molecules was higher when DC-tumor hybrids were the stimulators. Furthermore, only hybrids stimulated CTLs to produce IFN-γ in response to CMV-positive target cells. CONCLUSIONS: These data show the superiority of DC-tumor cell hybrids over their simple mixture as T-cell stimulators. Hybrids expressed more co-stimulatory and MHC molecules, induced higher antigen-specific T-cell expansion and were the only cells able to induce IFN-γ-producing antigen-specific T cells. Thus, these data offer further support for cancer immunotherapeutic approaches using DC-tumor cell hybrids.


Asunto(s)
Células Dendríticas/inmunología , Células Híbridas/inmunología , Inmunidad Celular , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Presentación de Antígeno , Vacunas contra el Cáncer/inmunología , Fusión Celular , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/patología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Células Híbridas/patología , Neoplasias/patología , Linfocitos T Citotóxicos/inmunología
11.
J Immunol ; 192(10): 4636-47, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24740501

RESUMEN

De novo regeneration of immunity is a major problem after allogeneic hematopoietic stem cell transplantation (HCT). HCT modeling in severely compromised immune-deficient animals transplanted with human stem cells is currently limited because of incomplete maturation of lymphocytes and scarce adaptive responses. Dendritic cells (DC) are pivotal for the organization of lymph nodes and activation of naive T and B cells. Human DC function after HCT could be augmented with adoptively transferred donor-derived DC. In this study, we demonstrate that adoptive transfer of long-lived human DC coexpressing high levels of human IFN-α, human GM-CSF, and a clinically relevant Ag (CMV pp65 protein) promoted human lymphatic remodeling in immune-deficient NOD.Rag1(-/-).IL-2rγ(-/-) mice transplanted with human CD34(+) cells. After immunization, draining lymph nodes became replenished with terminally differentiated human follicular Th cells, plasma B cells, and memory helper and cytotoxic T cells. Human Igs against pp65 were detectable in plasma, demonstrating IgG class-switch recombination. Human T cells recovered from mice showed functional reactivity against pp65. Adoptive immunotherapy with engineered DC provides a novel strategy for de novo immune reconstitution after human HCT and a practical and effective tool for studying human lymphatic regeneration in vivo in immune deficient xenograft hosts.


Asunto(s)
Traslado Adoptivo , Células Dendríticas/trasplante , Trasplante de Células Madre Hematopoyéticas , Quimera por Trasplante/inmunología , Aloinjertos , Animales , Citomegalovirus/genética , Citomegalovirus/inmunología , Células Dendríticas/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Xenoinjertos , Humanos , Interferón-alfa/genética , Interferón-alfa/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología
12.
J Transl Med ; 13: 240, 2015 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-26198406

RESUMEN

BACKGROUND: Reactivation of latent viruses such as human cytomegalovirus (HCMV) after allogeneic hematopoietic stem cell transplantation (HSCT) results in high morbidity and mortality. Effective immunization against HCMV shortly after allo-HSCT is an unmet clinical need due to delayed adaptive T cell development. Donor-derived dendritic cells (DCs) have a critical participation in stimulation of naïve T cells and immune reconstitution, and therefore adoptive DC therapy could be used to protect patients after HSCT. However, previous methods for ex vivo generation of adoptive donor-derived DCs were complex and inconsistent, particularly regarding cell viability and potency after thawing. We have previously demonstrated in humanized mouse models of HSCT the proof-of-concept of a novel modality of lentivirus-induced DCs ("SmyleDCpp65") that accelerated antigen-specific T cell development. METHODS: Here we demonstrate the feasibility of good manufacturing practices (GMP) for production of donor-derived DCs consisting of monocytes from peripheral blood transduced with an integrase-defective lentiviral vector (IDLV, co-expressing GM-CSF, IFN-α and the cytomegalovirus antigen pp65) that were cryopreserved and thawed. RESULTS: Upscaling and standardized production of one lot of IDLV and three lots of SmyleDCpp65 under GMP-compliant conditions were feasible. Analytical parameters for quality control of SmyleDCpp65 identity after thawing and potency after culture were defined. Cell recovery, uniformity, efficacy of gene transfer, purity and viability were high and consistent. SmyleDCpp65 showed only residual and polyclonal IDLV integration, unbiased to proto-oncogenic hot-spots. Stimulation of autologous T cells by GMP-grade SmyleDCpp65 was validated. CONCLUSION: These results underscore further developments of this individualized donor-derived cell vaccine to accelerate immune reconstitution against HCMV after HSCT in clinical trials.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Células Dendríticas/citología , Lentivirus , Trasplante de Células Madre/métodos , Animales , Técnicas de Cultivo de Célula , Supervivencia Celular , Criopreservación , Citomegalovirus , Infecciones por Citomegalovirus/prevención & control , Células Dendríticas/virología , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células HEK293 , Trasplante de Células Madre Hematopoyéticas , Humanos , Interferón-alfa/metabolismo , Leucocitos Mononucleares/citología , Fosfoproteínas/metabolismo , Plásmidos/metabolismo , Transgenes , Proteínas de la Matriz Viral/metabolismo
13.
Transfusion ; 54(8): 1961-70, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24673430

RESUMEN

BACKGROUND: Endothelial cells (ECs) play a pivotal role in the allogeneic immune response upon transplantation. Semaphorin 3A (Sema3A) was implicated in the modulation of EC growth, but its effects on immunogenicity were not previously investigated. STUDY DESIGN AND METHODS: ECs were transduced with a lentiviral vector encoding for the green fluorescence protein (GFP) sequence under the control of a Class II transactivator (CIITA)-dependent promoter. Upon stimulation of nonmodified ECs with recombinant Sema3A protein, mRNA and protein levels of CIITA, HLA-DR, and Sema3A receptors were evaluated. An enzyme-linked immunosorbent assay was developed to quantify Sema3A levels in the sera of kidney-transplanted patients. RESULTS: Sema3A stimulation of lentiviral vector encoding for the GFP sequence ECs caused a significant up regulation of the transgene expression, indicating an increase in CIITA levels. Stimulation of nonmodified ECs with Sema3A resulted in an up regulation of CIITA expression, which was associated with enhanced HLA-DR levels and an increase in alloreactive CD4+ T-cell proliferation. Sema3A receptor expression was enhanced by CIITA, establishing a positive feedback loop. Higher levels of Sema3A were observed in sera of patients presenting with organ rejection. CONCLUSION: This study links Sema3A signaling in ECs with increased CIITA levels and higher HLA-DR expression, resulting in CD4+ T-cell activation, which might have important implications for tissue and organ transplantation.


Asunto(s)
Células Endoteliales/inmunología , Rechazo de Injerto/sangre , Trasplante de Riñón , Semaforina-3A/fisiología , Adulto , Linfocitos T CD4-Positivos/inmunología , Ensayo de Inmunoadsorción Enzimática , Retroalimentación Fisiológica , Femenino , Genes Reporteros , Rechazo de Injerto/inmunología , Antígenos HLA-DR/biosíntesis , Antígenos HLA-DR/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Regiones Promotoras Genéticas , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Proteínas Recombinantes de Fusión/análisis , Semaforina-3A/sangre , Semaforina-3A/genética , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Transactivadores/fisiología , Activación Transcripcional , Transgenes , Regulación hacia Arriba
14.
HLA ; 103(6): e15541, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38923358

RESUMEN

Complications due to HCMV infection or reactivation remain a challenging clinical problem in immunocompromised patients, mainly due to insufficient or absent T-cell functionality. Knowledge of viral targets is crucial to improve monitoring of high-risk patients and optimise antiviral T-cell therapy. To expand the epitope spectrum, genetically-engineered dendritic cells (DCs) and fibroblasts were designed to secrete soluble (s)HLA-A*11:01 and infected with an HCMV mutant lacking immune evasion molecules (US2-6 + 11). More than 700 HLA-A*11:01-restricted epitopes, including more than 50 epitopes derived from a broad range of HCMV open-reading-frames (ORFs) were identified by mass spectrometry and screened for HLA-A*11:01-binding using established prediction tools. The immunogenicity of the 24 highest scoring new candidates was evaluated in vitro in healthy HLA-A*11:01+/HCMV+ donors. Thus, four subdominant epitopes and one immunodominant epitope, derived from the anti-apoptotic protein UL36 and ORFL101C (A11SAL), were identified. Their HLA-A*11:01 complex stability was verified in vitro. In depth analyses revealed highly proliferative and cytotoxic memory T-cell responses against A11SAL, with T-cell responses comparable to the immunodominant HLA-A*02:01-restricted HCMVpp65NLV epitope. A11SAL-specific T cells were also detectable in vivo in immunosuppressed transplant patients and shown to be effective in an in vitro HCMV-infection model, suggesting their crucial role in inhibiting viral replication and improvement of patient's outcome. The developed in vitro pipeline is the first to utilise genetically-engineered DCs to identify naturally presented immunodominant HCMV-derived epitopes. It therefore offers advantages over in silico predictions, is transferable to other HLA alleles, and will significantly expand the repertoire of viral targets to improve therapeutic options.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Células Dendríticas , Epítopos de Linfocito T , Epítopos Inmunodominantes , Humanos , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Epítopos Inmunodominantes/inmunología , Células Dendríticas/inmunología , Epítopos de Linfocito T/inmunología , Antígeno HLA-A11/inmunología , Antígeno HLA-A11/genética , Fibroblastos/inmunología , Fibroblastos/virología , Células Presentadoras de Antígenos/inmunología
15.
Blood ; 128(25): 2874-2875, 2016 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-28007730
16.
Ann Hematol ; 92(8): 1079-90, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23616009

RESUMEN

Relapse occurs frequently after treatment of acute myeloid leukemia (AML) patients with the FMS-like tyrosine kinase 3-internal tandem duplication (ITD) mutation. The availability of immunologic biomarkers to predict patients at high risk could allow clinicians to accelerate alternative treatments such as stem cell transplantation, immunotherapy, or novel drugs. We have previously reported that first diagnostic (FD) ITD(+) AML showed immunophenotypic and functional characteristics of arrested dendritic cell (DC) precursors. In this study, we show that the high frequency of precursor DCs in 16 FD ITD(+) AML samples (Lin(-)/HLA-DR(+)/CD11c(+)/CD123(+)) was associated with a lack of terminal DCs (myeloid DCs: BDCA-1(+) or BDCA-3(+); plasmacytoid DC: BDCA-2(+)). We further evaluated prospectively the peripheral blood complete remission (CR) samples obtained from 11 ITD(+) AML patients after chemotherapy regarding the frequency of DCs and their pattern of cytokine production. Whereas the aberrant frequencies of precursor and terminal plasmacytoid DCs resolved during remission, the myeloid DC compartment did not fully recover. For an available cohort of patients (n = 4) who could be monitored over a period of >15 months after FD, we identified IL-10, TNF-α, IL-6, and IL-1ß as cytokines produced by the CR samples at high levels a few months prior to relapse. Cell-free supernatant of an FD ITD(+) AML sample stimulated monocytes obtained from two healthy donors to secrete IL-10, TNF-α, IL-6, and IL-1ß. Thus, we hypothesize that ITD(+) AML minimal residual disease can act directly as dysfunctional antigen-presenting cells or indirectly by production of factors that convert monocytes into myeloid-derived suppressor cells secreting cytokines that promote immune evasion. Monitoring these immunologic biomarkers could improve prediction of relapse.


Asunto(s)
Antígenos de Diferenciación/análisis , Citocinas/sangre , Células Dendríticas/patología , Leucemia Mieloide/inmunología , Tirosina Quinasa 3 Similar a fms/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores , Diferenciación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Células Dendríticas/química , Femenino , Genes del Tumor de Wilms , Humanos , Inmunofenotipificación , Leucemia Mieloide/sangre , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/genética , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Células Mieloides/patología , Neoplasia Residual , Pronóstico , Estudios Prospectivos , Recurrencia , Inducción de Remisión , Secuencias Repetidas en Tándem
17.
Front Immunol ; 14: 1103695, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36817460

RESUMEN

Introduction: Epstein-Barr virus (EBV) is a widely spread pathogen associated with lymphoproliferative diseases, B/ T/ NK cell lymphomas, nasopharyngeal carcinoma (NPC) and gastric carcinoma (GC). EBV lytic reactivations contribute to the genomic instability, inflammation and tumorigenesis of NPC, promoting cancer progression. Patients with NPC refractory to standard therapies show dismal survival. EBV gp350 is an envelope protein detectable in NPC specimens intracellularly and on the cell membrane of malignant cells, and is a potential viral antigen for T cell-directed immunotherapies. The potency of T cells engineered with a chimeric antigen receptor (CAR) targeting gp350 against EBV+ lymphoproliferative disease was previously shown. Methods: Here, we advanced towards preclinical and non-clinical developments of this virus-specific CAR-T cell immunotherapy against NPC. Different gp350CAR designs were inserted into a lentiviral vector (LV) backbone. Results: A construct expressing the scFv 7A1-anti-gp350 incorporating the CD8 transmembrane and CD28.CD3ζ signaling domain (ZT002) was selected. High titer ZT002 (~1x108 TU/ml) was manufactured in HEK 293T/17 suspension cells in serum free media as large-scale production under good manufacturing practices (GMP). A LV multiplicity of infection (MOI) of 1 resulted in high frequencies of functional gp350CAR+ T cells (>70%) at a low (<2) vector copy numbers in the genome. ZT002 was therefore used to establish gp350CAR-T batch run production methods. GMP upscaling and validation of T cell transduction and expansion in several runs resulted in average 3x109 gp350CAR-T cells per batch. >80% CD3+ gp350CAR-T cells bound to purified gp350 protein. In vitro cytotoxicity and cytokine secretion assays (IFN-γ and TNF-α) confirmed the specificity of gp350CAR-T cells against gp350+ NPC, GC and lymphoma cell targets. Immunocompromised B-NDG mice (NOD.CB17-PrkdcscidIl2rgtm1/Bcgen) were challenged s.c. with a EBV+ NPC C666.1 cell line expressing gp350 and then treated with escalating doses of gp350CAR-T cells or with non-transduced T cells. gp350CAR-T cells promoted antitumor responses, bio-distributed in several tissues, infiltrated in tumors and rejected gp350+ tumor cells. Discussion: These results support the use of gp350CAR-T cells generated with ZT002 as an Innovative New Drug to treat patients with solid and liquid EBV-associated malignancies.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Neoplasias Nasofaríngeas , Animales , Ratones , Herpesvirus Humano 4 , Ratones Endogámicos NOD , Carcinoma Nasofaríngeo , Linfocitos T , Receptores Quiméricos de Antígenos/inmunología
18.
Cells ; 13(1)2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38201224

RESUMEN

Antiviral neutralizing antibodies (nAbs) are commonly derived from B cells developed in immunized or infected animals and humans. Fully human antibodies are preferred for clinical use as they are potentially less immunogenic. However, the function of B cells varies depending on their homing pattern and an additional hurdle for antibody discovery in humans is the source of human tissues with an immunological microenvironment. Here, we show an efficient method to pharm human antibodies using immortalized B cells recovered from Nod.Rag.Gamma (NRG) mice reconstituting the human immune system (HIS). Humanized HIS mice were immunized either with autologous engineered dendritic cells expressing the human cytomegalovirus gB envelope protein (HCMV-gB) or with Epstein-Barr virus-like particles (EB-VLP). Human B cells recovered from spleen of HIS mice were efficiently immortalized with EBV in vitro. We show that these immortalized B cells secreted human IgGs with neutralization capacities against prototypic HCMV-gB and EBV-gp350. Taken together, we show that HIS mice can be successfully used for the generation and pharming fully human IgGs. This technology can be further explored to generate antibodies against emerging infections for diagnostic or therapeutic purposes.


Asunto(s)
Vacunas contra el Cáncer , Infecciones por Virus de Epstein-Barr , Humanos , Animales , Ratones , Bazo , Herpesvirus Humano 4 , Anticuerpos Antivirales , Inmunoglobulina G , Citomegalovirus
19.
Front Immunol ; 14: 1086433, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033919

RESUMEN

Introduction: The ubiquitous Epstein-Barr virus (EBV) is an oncogenic herpes virus associated with several human malignancies. EBV is an immune-evasive pathogen that promotes CD8+ T cell exhaustion and dysregulates CD4+ T cell functions. Burkitt lymphoma (BL) is frequently associated with EBV infections. Since BL relapses after conventional therapies are difficult to treat, we evaluated prospective off-the-shelf edited CAR-T cell therapies targeting CD19 or the EBV gp350 cell surface antigen. Methods: We used CRISPR/Cas9 gene editing methods to knock in (KI) the CD19CAR.CD28z or gp350CAR.CD28z into the T cell receptor (TCR) alpha chain (TRAC) locus. Results: Applying upscaled methods with the ExPERT ATx® MaxCyte system, KI efficacy was ~20% of the total ~2 × 108 TCR-knocked-out (KO) generated cells. KOTCRKICAR-T cells were co-cultured in vitro with the gp350+CD19+ BL cell lines Daudi (infected with type 1 EBV) or with Jiyoye (harboring a lytic type 2 EBV). Both types of CAR-T cells showed cytotoxic effects against the BL lines in vitro. CD8+ KICAR-T cells showed higher persistency than CD4+ KICAR-T cells after in vitro co-culture with BL and upregulation of the activation/exhaustion markers PD-1, LAG-3, and TIM-3. Two preclinical in vivo xenograft models were set up with Nod.Rag.Gamma mice injected intravenously (i.v.) with 2 × 105 Daudi/fLuc-GFP or with Jiyoye/fLuc-GFP cells. Compared with the non-treated controls, mice challenged with BL and treated with CD19KICAR-T cells showed delayed lymphoma dissemination with lower EBV DNA load. Notably, for the Jiyoye/fLuc-GFP model, almost exclusively CD4+ CD19KICAR-T cells were detectable at the endpoint analyses in the bone marrow, with increased frequencies of regulatory T cells (Tregs) and TIM-3+CD4+ T cells. Administration of gp350KICAR-T cells to mice after Jiyoye/GFP-fLuc challenge did not inhibit BL growth in vivo but reduced the EBV DNA load in the bone marrow and promoted gp350 antigen escape. CD8+PD-1+LAG-3+ gp350KICAR-T cells were predominant in the bone marrow. Discussion: The two types of KOTCRKICAR-T cells showed different therapeutic effects and in vivo dynamics. These findings reflect the complexities of the immune escape mechanisms of EBV, which may interfere with the CAR-T cell property and potency and should be taken into account for future clinical translation.


Asunto(s)
Linfoma de Burkitt , Infecciones por Virus de Epstein-Barr , Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Linfoma de Burkitt/terapia , Herpesvirus Humano 4 , Receptor 2 Celular del Virus de la Hepatitis A , Receptor de Muerte Celular Programada 1 , Estudios Prospectivos , Receptores de Antígenos de Linfocitos T alfa-beta
20.
bioRxiv ; 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-38116030

RESUMEN

Chimeric antigen receptor (CAR)-reprogrammed immune cells hold significant therapeutic potential for oncology, autoimmune diseases, transplant medicine, and infections. All approved CAR-T therapies rely on personalized manufacturing using undirected viral gene transfer, which results in non-physiological regulation of CAR-signaling and limits their accessibility due to logistical challenges, high costs and biosafety requirements. Here, we propose a novel approach utilizing CRISPR-Cas gene editing to redirect T cells and natural killer (NK) cells with CARs. By transferring shorter, truncated CAR-transgenes lacking a main activation domain into the human CD3 ζ (CD247) gene, functional CAR fusion-genes are generated that exploit the endogenous CD3 ζ gene as the CAR's activation domain. Repurposing this T/NK-cell lineage gene facilitated physiological regulation of CAR-expression and reprogramming of various immune cell types, including conventional T cells, TCRγ/δ T cells, regulatory T cells, and NK cells. In T cells, CD3 ζ in-frame fusion eliminated TCR surface expression, reducing the risk of graft-versus-host disease in allogeneic off-the-shelf settings. CD3 ζ-CD19-CAR-T cells exhibited comparable leukemia control to T cell receptor alpha constant ( TRAC )-replaced and lentivirus-transduced CAR-T cells in vivo . Tuning of CD3 ζ-CAR-expression levels significantly improved the in vivo efficacy. Compared to TRAC -edited CAR-T cells, integration of a Her2-CAR into CD3 ζ conveyed similar in vitro tumor lysis but reduced susceptibility to activation-induced cell death and differentiation, presumably due to lower CAR-expression levels. Notably, CD3 ζ gene editing enabled reprogramming of NK cells without impairing their canonical functions. Thus, CD3 ζ gene editing is a promising platform for the development of allogeneic off-the-shelf cell therapies using redirected killer lymphocytes. Key points: Integration of ζ-deficient CARs into CD3 ζ gene allows generation of functional TCR-ablated CAR-T cells for allogeneic off-the-shelf use CD3 ζ-editing platform allows CAR reprogramming of NK cells without affecting their canonical functions.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA