Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
J Pediatric Infect Dis Soc ; 13(1): 75-83, 2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-38019957

RESUMO

BACKGROUND: Respiratory viral infections are common among pediatric transplant patients, with human rhinovirus (HRV) being the most frequent. In pediatric patients undergoing hemopoietic cell transplant (HCT), infection with HRV has been associated with progression to lower respiratory tract infection (LRTI) and adverse outcomes. We describe the clinical presentation and outcomes of HRV infection in children undergoing HCT. METHODS: Single-center retrospective study. HCT recipients who were positive for HRV/EV (HRV+) or negative for any respiratory virus (VN) by BioFire® FilmArray® panel between October 2014 and December 2017, were included. Primary outcomes were progression to LRTI, ICU admission, all-cause mortality at 3 and 6 months, and respiratory event-related mortality at 6 months. RESULTS: 227 patients (160 allogeneic HCT) were included. Of all patients, 108/227 (47.6%) were HRV+. From all HRV+, 95/108 (88%) were symptomatic and 68/107 (63.6%) of the diagnosis were made pretransplant. The median age of HRV+ was significantly lower than VN patients (5 vs 10 years). Cough and rhinorrhea were more frequently observed in HRV+ (53.7 and 60% vs 19.8 and 22.8%, respectively). No differences were found between both groups pretransplant and overall in rates progression to LRTI, ICU admission, mechanical ventilation, all-cause within 3 and 6 months, and mortality related with respiratory failure. No significant association was found between the severity of respiratory disease and the type of conditioning, type of transplant, or absolute lymphocyte count. CONCLUSIONS: HRV infection is frequently detected in HCT recipients but is not associated with severity of respiratory disease, need for intensive care unit or mortality, including those diagnosed before transplant, suggesting that delaying HCT in this scenario may not be needed. Multicenter larger studies are required to confirm these findings.


Assuntos
Infecções por Enterovirus , Enterovirus , Infecções por Picornaviridae , Infecções Respiratórias , Criança , Humanos , Transplante de Células/efeitos adversos , Estudos Retrospectivos , Rhinovirus , Pré-Escolar , Lactente
2.
J Clin Endocrinol Metab ; 108(6): 1273-1289, 2023 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-36611246

RESUMO

Primary adrenal insufficiency (PAI) occurs in 1 in 5 to 7000 adults. Leading etiologies are autoimmune adrenalitis in adults and congenital adrenal hyperplasia (CAH) in children. Oral replacement of cortisol is lifesaving, but poor quality of life, repeated adrenal crises, and dosing uncertainty related to lack of a validated biomarker for glucocorticoid sufficiency persists. Adrenocortical cell therapy and gene therapy may obviate many of the shortcomings of adrenal hormone replacement. Physiological cortisol secretion regulated by pituitary adrenocorticotropin could be achieved through allogeneic adrenocortical cell transplantation, production of adrenal-like steroidogenic cells from either stem cells or lineage conversion of differentiated cells, or for CAH, gene therapy to replace or repair a defective gene. The adrenal cortex is a high-turnover organ and thus failure to incorporate progenitor cells within a transplant will ultimately result in graft exhaustion. Identification of adrenocortical progenitor cells is equally important in gene therapy, for which new genetic material must be specifically integrated into the genome of progenitors to ensure a durable effect. Delivery of gene-editing machinery and a donor template, allowing targeted correction of the 21-hydroxylase gene, has the potential to achieve this. This review describes advances in adrenal cell transplants and gene therapy that may allow physiological cortisol production for children and adults with PAI.


Assuntos
Hiperplasia Suprarrenal Congênita , Insuficiência Adrenal , Criança , Adulto , Humanos , Hidrocortisona , Qualidade de Vida , Insuficiência Adrenal/genética , Insuficiência Adrenal/terapia , Insuficiência Adrenal/complicações , Hiperplasia Suprarrenal Congênita/genética , Terapia Genética/efeitos adversos , Transplante de Células/efeitos adversos
3.
J Virol ; 96(3): e0139421, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34818071

RESUMO

Humanized mouse models are based on the engraftment of human cells in immunodeficient mouse strains, most notably the NSG strain. Most used models have a major limitation in common, the development of graft-versus-host disease (GVHD). GVHD not only introduces variabilities into the research data but also leads to animal welfare concerns. A new mouse strain, B6.129S-Rag2tm1Fwa CD47tm1Fpl Il2rgtm1Wjl/J, which lacks Rag1, IL2rg, and CD47 (triple knockout [TKO]), is resistant to GVHD development. We transplanted TKO mice with human peripheral blood mononuclear cells (PBMCs) to establish a new humanized PBMC (hu-PBMC) mouse model. A cohort of these mice was infected with HIV-1 and monitored for plasma HIV viremia and CD4+ T cell depletion. The onset and progression of GVHD were monitored by clinical signs. This study demonstrates that TKO mice transplanted with human PBMCs support engraftment of human immune cells in primary and secondary lymphoid tissues, rectum, and brain. Moreover, the TKO hu-PBMC model supports HIV-1 infection via the intraperitoneal, rectal, or vaginal route, as confirmed by robust plasma HIV viremia and CD4+ T cell depletion. Lastly, TKO mice showed a delayed onset of GVHD clinical signs (∼24 days) and exhibited significant decreases in plasma levels of tumor necrosis factor beta (TNF-ß). Based on these results, the TKO hu-PBMC mouse model not only supports humanization and HIV-1 infection but also has a delayed onset of GVHD development, making this model a valuable tool in HIV research. IMPORTANCE Currently, there is no cure or vaccine for HIV infection; thus, continued research is needed to end the HIV pandemic. While many animal models are used in HIV research, none is used more than the humanized mouse model. A major limitation with current humanized mouse models is the development of graft-versus-host disease (GVHD). Here, we describe a novel humanized-PBMC mouse model that has a delayed onset GVHD development and supports and models HIV infection comparably to well-established humanized mouse models.


Assuntos
Transplante de Células , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro/etiologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Leucócitos Mononucleares/transplante , Animais , Transplante de Células/efeitos adversos , Transplante de Células/métodos , Suscetibilidade a Doenças , Feminino , HIV-1/imunologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Pesquisa , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Fatores de Tempo
4.
Methods Mol Biol ; 2174: 219-244, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32813253

RESUMO

The use of immunotherapy as an alternative treatment for cancer patients has become of great interest in the scientific community as it is required to overcome many of the currently unsolved problems such as tumor escape, immunosuppression and unwanted unspecific toxicity. The use of chimeric antigen receptor T cells has been a very successful strategy in some hematologic malignancies. However, the application of CAR T cells has been limited to solid tumors, and this has aimed the development of new generation of CARs with enhanced effectivity and specificity. Here, we review the state of the art of CAR T cell therapy with special emphasis on the current challenges and opportunities.


Assuntos
Transplante de Células/efeitos adversos , Transplante de Células/métodos , Neoplasias/terapia , Receptores de Antígenos Quiméricos , Linfócitos T/fisiologia , Genes Transgênicos Suicidas , Neoplasias Hematológicas/terapia , Humanos , Receptores de Antígenos Quiméricos/química , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/citologia
5.
Biochem Biophys Res Commun ; 534: 765-772, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33213838

RESUMO

Modifying T cells to attack tumors using engineered chimeric receptors display powerful new therapeutic capabilities. Unfortunately, the effectiveness of therapeutic T cells is limited due to the inherent T cell responses: certain facets of endogenous response programs may be toxic, and the ability to overcome the immunosuppression in TME is deficient. Here we developed a Notch receptor based synNotch T cell platform that is able to response to target tumor cells and selectively lead to CXCL10 production. Further study showed that the administration of synNotch T cells significantly inhibited the tumor growth in a humanized murine model, accompanied by the increased infiltration of CD3+T cells and elevated level of CXCL10 and IFN-γ in the tumor site. A slightly increased level of CXCL10 and limited IFN-γ were found in the serum in mice received synNotch T cells, suggesting a high security of this treatment. Finally, we demonstrated that CXCL10 is sufficient and indispensable for the synNotch T cells induced anti-tumor effect. This study provided theoretical and experimental bases for the clinical implication of CXCL10 encoding synNotch T cells.


Assuntos
Transplante de Células/métodos , Quimiocina CXCL10/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos T/imunologia , Células A549 , Animais , Transplante de Células/efeitos adversos , Quimiocina CXCL10/genética , Técnicas de Cocultura , Síndrome da Liberação de Citocina/etiologia , Proteínas Ligadas por GPI/metabolismo , Humanos , Interferon gama/metabolismo , Masculino , Mesotelina , Camundongos Endogâmicos NOD , Linfócitos T/metabolismo , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Front Immunol ; 11: 565236, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193333

RESUMO

Chimeric Antigen Receptor-T cells (CAR-T) are considered novel biological agents, designed to selectively attack cancer cells expressing specific antigens, with demonstrated clinical activity in patients affected with relapsed/refractory B-cell malignancies. In consideration of their complexity, the use of CAR-T requires dedicated clinical setting and health care practitioners with expertise in the selection, treatment, and management of toxicities and side effects. Such issue appears particularly important when contextualized in the rapid progress of CAR-T cell treatment, translating into a constant need of updating and evolution. Moreover, the clinical grade manufacturing of CAR-T cells is complex and implies articulated regulatory and organizational aspects. The main goal of this review is to summarize and provide an accurate analysis of the clinical, logistic, and regulatory requirements of CAR-T cell centers. Finally, we describe a new occupational figure called "CAR-T specialist" devoted to the establishment and coordination of the required facilities and regulatory landscape in the context of cancer centers.


Assuntos
Antígenos de Neoplasias/imunologia , Transplante de Células/efeitos adversos , Imunoterapia Adotiva/efeitos adversos , Neoplasias/terapia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Assistência ao Convalescente/métodos , Antígenos CD19/imunologia , Doadores de Sangue/legislação & jurisprudência , Transplante de Células/legislação & jurisprudência , Pessoal de Saúde/educação , Humanos , Imunoterapia Adotiva/legislação & jurisprudência , Seleção de Pacientes , Transplantes , Microambiente Tumoral/imunologia
7.
Arch Dis Child Fetal Neonatal Ed ; 105(5): 563-568, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32253200

RESUMO

Recent decades have seen the rapid progress of neonatal intensive care, and the survival rates of the most preterm infants are improving. This improvement is associated with changing patterns of morbidity and new phenotypes of bronchopulmonary dysplasia and preterm brain injury are recognised. Inflammation and immaturity are known contributors to their pathogenesis. However, a new phenomenon, the exhaustion of progenitor cells is emerging as an important factor. Current therapeutic approaches do not adequately address these new mechanisms of injury. Cell therapy, that is the use of stem and stem-like cells, with its potential to both repair and prevent injury, offers a new approach to these challenging conditions. This review will examine the rationale for cell therapy in the extremely preterm infant, the preclinical and early clinical evidence to support its use in bronchopulmonary dysplasia and preterm brain injury. Finally, it will address the challenges in translating cell therapy from the laboratory to early clinical trials.


Assuntos
Lesões Encefálicas/terapia , Displasia Broncopulmonar/terapia , Transplante de Células/métodos , Lactente Extremamente Prematuro , Doenças do Prematuro/terapia , Animais , Transplante de Células/efeitos adversos , Protocolos Clínicos , Ensaios Clínicos como Assunto , Transplante de Células-Tronco de Sangue do Cordão Umbilical/efeitos adversos , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sangue Fetal , Humanos , Recém-Nascido , Recém-Nascido de muito Baixo Peso , Terapia Intensiva Neonatal/métodos , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Fatores de Tempo
8.
Methods Mol Biol ; 2140: 43-64, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32207105

RESUMO

3D bioprinting involves engineering live cells into a 3D structure, using a 3D printer to print cells, often together with a compatible 3D scaffold. 3D-printed cells and tissues may be used for a range of purposes including medical research, in vitro drug testing, and in vivo transplantation. The inclusion of living cells and biomaterials in the 3D printing process raises ethical, policy, and regulatory issues at each stage of the bioprinting process that include the source of cells and materials, stability and biocompatibility of cells and materials, disposal of 3D-printed materials, intended use, and long-term effects. This chapter focuses on the ethical issues that arise from 3D bioprinting in the lab-from consideration of the source of cells and materials, ensuring their quality and safety, through to testing of bioprinted materials in animal and human trials. It also provides guidance on where to seek information concerning appropriate regulatory frameworks and guidelines, including on classification and patenting of 3D-bioprinted materials, and identifies regulatory gaps that deserve attention.


Assuntos
Bioimpressão/ética , Impressão Tridimensional/ética , Experimentação Animal/ética , Experimentação Animal/normas , Animais , Transplante de Células/efeitos adversos , Transplante de Células/ética , Ensaios Clínicos como Assunto/ética , Estudos de Avaliação como Assunto , Experimentação Humana/ética , Humanos , Implantes Experimentais/efeitos adversos , Implantes Experimentais/ética , Propriedade Intelectual , Patentes como Assunto , Políticas , Guias de Prática Clínica como Assunto , Células-Tronco , Engenharia Tecidual/ética , Alicerces Teciduais/efeitos adversos
9.
Jpn J Clin Oncol ; 50(6): 661-670, 2020 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-32100001

RESUMO

OBJECTIVE: Primary disease relapse (PDR) of malignant hematologic conditions after standard hematopoietic stem cell transplant (HSCT) is one of the most challenging diseases; therefore ongoing researches are aiming at relapse prevention and minimizing the transplant-related side effects. Prophylactic donor lymphocytes (pDLI) had been proposed as a valuable strategy for PDR prevention, but early studies had been discouraging due to the limited benefit and possible association with acute graft-versus-host disease (aGVHD). Therefore, we conducted a meta-analysis to evaluate the association between pDLI use, PDR, aGVHD and OS. METHOD: We performed a comprehensive literature search in MEDLINE, Cochrane library and Embase database from inception to May 2019 for studies that evaluated the association between pDLI and PDR. We conducted a random effect meta-analysis of 9 studies involving a total of 748 participants (pDLI = 398, non-pDLI = 350) and reported the pooled odd ratio (OR) for association of pDLI use, PDR, aGVHD and OS. RESULT: We found a significant decreased odd of PDR in the pDLI group (pooled OR = 0.42, 95% CI 0.30-0.58, I2 = 0%), but there was no significant increased odd of aGVHD (pooled OR of 0.98, 95% CI 0.56-1.72, I2 = 0.8%). We also found that there was an increased odd of overall survival (OS) (pooled OR 3.17, 95% CI 1.85-5.45, I2 = 50.2%). CONCLUSION: There are significantly decreased odd of PDR and increased odd of OS in the pDLI group compared to the control group, but there is no statistically significant increased odd of aGVHD as suggested by previous studies. We concluded that pDLI is a potentially valuable method for post-transplant PDR prevention.


Assuntos
Transplante de Células/efeitos adversos , Neoplasias Hematológicas/prevenção & controle , Linfócitos , Adulto , Feminino , Doença Enxerto-Hospedeiro/etiologia , Neoplasias Hematológicas/mortalidade , Neoplasias Hematológicas/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva , Adulto Jovem
10.
Biochemistry (Mosc) ; 85(Suppl 1): S108-S130, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32087056

RESUMO

It had been commonly believed for a long time, that once established, degeneration of the central nervous system (CNS) is irreparable, and that adult person merely cannot restore dead or injured neurons. The existence of stem cells (SCs) in the mature brain, an organ with minimal regenerative ability, had been ignored for many years. Currently accepted that specific structures of the adult brain contain neural SCs (NSCs) that can self-renew and generate terminally differentiated brain cells, including neurons and glia. However, their contribution to the regulation of brain activity and brain regeneration in natural aging and pathology is still a subject of ongoing studies. Since the 1970s, when Fuad Lechin suggested the existence of repair mechanisms in the brain, new exhilarating data from scientists around the world have expanded our knowledge on the mechanisms implicated in the generation of various cell phenotypes supporting the brain, regulation of brain activity by these newly generated cells, and participation of SCs in brain homeostasis and regeneration. The prospects of the SC research are truthfully infinite and hitherto challenging to forecast. Once researchers resolve the issues regarding SC expansion and maintenance, the implementation of the SC-based platform could help to treat tissues and organs impaired or damaged in many devastating human diseases. Over the past 10 years, the number of studies on SCs has increased exponentially, and we have already become witnesses of crucial discoveries in SC biology. Comprehension of the mechanisms of neurogenesis regulation is essential for the development of new therapeutic approaches for currently incurable neurodegenerative diseases and neuroblastomas. In this review, we present the latest achievements in this fast-moving field and discuss essential aspects of NSC biology, including SC regulation by hormones, neurotransmitters, and transcription factors, along with the achievements of genetic and chemical reprogramming for the safe use of SCs in vitro and in vivo.


Assuntos
Envelhecimento/metabolismo , Regeneração Nervosa/fisiologia , Células-Tronco Neurais/metabolismo , Doenças Neurodegenerativas/terapia , Adulto , Animais , Transplante de Células/efeitos adversos , Transplante de Células/métodos , Giro Denteado/efeitos dos fármacos , Giro Denteado/metabolismo , Epigênese Genética , Hormônios/metabolismo , Hormônios/farmacologia , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Doenças Neurodegenerativas/metabolismo , Neurogênese , Neurotransmissores/metabolismo , Neurotransmissores/farmacologia , Fatores de Transcrição/metabolismo
11.
Am J Ophthalmol ; 208: 242-250, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31449791

RESUMO

PURPOSE: To analyze corneal neovascularization using anterior segment optical coherence tomography angiography (AS-OCTA) in patients following cultivated oral mucosal epithelial sheet transplantation (COMET). DESIGN: Observational case series. METHODS: Nine eyes in 7 patients were analyzed. Four images of corneal quadrant were obtained by AS-OCTA from each patient during follow-up post-COMET in the Department of Ophthalmology at Osaka University Hospital. The depth of corneal neovascularization was evaluated using en face and B-scan images. Each quadrant image was classified as 1 of the following 5 types: stromal, predominantly stromal, epithelial, predominantly epithelial, or avascular. The image quality of slit-lamp photography and AS-OCTA was graded from 0 to 4. Manually segmented images of the epithelial and stromal vessels were obtained. MAIN OUTCOME MEASURES: Depth and image quality of corneal neovascularization following COMET. RESULTS: Six patients were male and 1 was female. The mean patient age was 61.3 ± 19.1 years. Thirty-six quadrant images were obtained, of which 4 (11.1%) were stromal, 16 (44.4%) were predominantly stromal, 3 (8.3%) were epithelial, 11 (30.6%) were predominantly epithelial, and 2 (5.6%) were avascular. The image quality obtained by AS-OCTA was significantly better than that obtained by slit-lamp photography (2.38 ± 0.94 vs 2.03 ± 0.90; P = .021). Segmentation images clearly demonstrated both epithelial and stromal vasculatures individually. CONCLUSIONS: AS-OCTA is useful for evaluation of depth of corneal neovascularization and has the potential to distinguish between conjunctivalization and stromal neovascularization following COMET. Findings on AS-OCTA could contribute to clinical decision making, given that retreatment is required for conjunctivalization after COMET.


Assuntos
Segmento Anterior do Olho/diagnóstico por imagem , Transplante de Células/efeitos adversos , Neovascularização da Córnea/etiologia , Células Epiteliais/transplante , Mucosa Bucal/citologia , Adulto , Idoso , Técnicas de Cultura de Células , Neovascularização da Córnea/diagnóstico por imagem , Feminino , Angiofluoresceinografia , Seguimentos , Humanos , Limbo da Córnea/patologia , Masculino , Pessoa de Meia-Idade , Células-Tronco/patologia , Tomografia de Coerência Óptica
12.
Mol Ther ; 27(6): 1114-1125, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-30962163

RESUMO

By fusing the extracellular domain of the natural killer (NK) cell receptor NKG2D to DAP12, we constructed a chimeric antigen receptor (CAR) to improve NK cell tumor responses. An RNA electroporation approach that provides transient expression of the CAR was adopted as a risk mitigation strategy. Expression of the NKG2D RNA CAR significantly augmented the cytolytic activity of NK cells against several solid tumor cell lines in vitro and provided a clear therapeutic benefit to mice with established solid tumors. Three patients with metastatic colorectal cancer were then treated with local infusion of the CAR-NK cells. Reduction of ascites generation and a marked decrease in number of tumor cells in ascites samples were observed in the first two patients treated with intraperitoneal infusion of low doses of the CAR-NK cells. The third patient with metastatic tumor sites in the liver was treated with ultrasound-guided percutaneous injection, followed by intraperitoneal infusion of the CAR-NK cells. Rapid tumor regression in the liver region was observed with Doppler ultrasound imaging and complete metabolic response in the treated liver lesions was confirmed by positron emission tomography (PET)- computed tomographic (CT) scanning. Our results highlight a promising therapeutic potential of using RNA CAR-modified NK cells to treat metastatic colorectal cancer.


Assuntos
Transferência Adotiva/métodos , Transplante de Células/métodos , Neoplasias Colorretais/terapia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Receptores de Antígenos Quiméricos/imunologia , Transferência Adotiva/efeitos adversos , Animais , Engenharia Celular/métodos , Transplante de Células/efeitos adversos , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Citotoxicidade Imunológica/genética , Estudos de Viabilidade , Feminino , Vetores Genéticos , Células HCT116 , Humanos , Células Matadoras Naturais/metabolismo , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Projetos Piloto , RNA Mensageiro/genética , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Mol Ther ; 27(6): 1126-1138, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31005597

RESUMO

Clinical success of autologous CD19-directed chimeric antigen receptor T cells (CAR Ts) in acute lymphoblastic leukemia and non-Hodgkin lymphoma suggests that CAR Ts may be a promising therapy for hematological malignancies, including multiple myeloma. However, autologous CAR T therapies have limitations that may impact clinical use, including lengthy vein-to-vein time and manufacturing constraints. Allogeneic CAR T (AlloCAR T) therapies may overcome these innate limitations of autologous CAR T therapies. Unlike autologous cell therapies, AlloCAR T therapies employ healthy donor T cells that are isolated in a manufacturing facility, engineered to express CARs with specificity for a tumor-associated antigen, and modified using gene-editing technology to limit T cell receptor (TCR)-mediated immune responses. Here, transcription activator-like effector nuclease (TALEN) gene editing of B cell maturation antigen (BCMA) CAR Ts was used to confer lymphodepletion resistance and reduced graft-versus-host disease (GvHD) potential. The safety profile of allogeneic BCMA CAR Ts was further enhanced by incorporating a CD20 mimotope-based intra-CAR off switch enabling effective CAR T elimination in the presence of rituximab. Allogeneic BCMA CAR Ts induced sustained antitumor responses in mice supplemented with human cytokines, and, most importantly, maintained their phenotype and potency after scale-up manufacturing. This novel off-the-shelf allogeneic BCMA CAR T product is a promising candidate for clinical evaluation.


Assuntos
Antígeno de Maturação de Linfócitos B/imunologia , Transplante de Células/métodos , Imunoterapia Adotiva/métodos , Mieloma Múltiplo/terapia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Linfócitos T/transplante , Animais , Antineoplásicos Imunológicos/uso terapêutico , Antígeno de Maturação de Linfócitos B/genética , Doadores de Sangue , Linhagem Celular Tumoral , Transplante de Células/efeitos adversos , Citotoxicidade Imunológica/genética , Edição de Genes , Vetores Genéticos , Doença Enxerto-Hospedeiro/terapia , Humanos , Imunoterapia Adotiva/efeitos adversos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mieloma Múltiplo/patologia , Intervalo Livre de Progressão , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Rituximab/uso terapêutico , Linfócitos T/metabolismo , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/genética , Transdução Genética , Transplante Homólogo/métodos
14.
Invest Ophthalmol Vis Sci ; 60(5): 1298-1308, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30933257

RESUMO

Purpose: To report occurrence of acute severe inflammation after surgical implantation of mycoplasma-infected induced pluripotent stem cell-derived RPE (iPS-RPE) cells into the eyes of healthy primates, and determine the immunopathological mechanisms of the inflammation. Methods: Ophthalmic allogeneic transplantation of iPS-RPE cells was performed in the subretina of major histocompatibility complex (MHC)-matched (two eyes) and MHC-mismatched (one eye) healthy cynomolgus monkeys. The clinical course after transplantation was observed using color fundus photography, fluorescence angiography, and optical coherence tomography. After the animals were killed at 1 month after surgery, eyeballs were removed and pathologically examined. Microorganisms were analyzed by PCR methods and BLAST analysis using preserved graft iPS-RPE cells and the recipients' vitreous humor. Mixed lymphocyte-RPE assay was performed on the mycoplasma-infected and noninfected iPS-RPE cells in vitro. Results: In tested eyes, abnormal findings were observed in the grafted retina 2 weeks after surgery. Here, we observed retinal vasculitis and hemorrhage, retinal detachment, and infiltration of inflammatory cells into the retina of the eyes. One month after surgery, animals were killed due to the severe immune responses observed. Using PCR methods, sequence analysis detected mycoplasma-DNA (Mycoplasma arginini species) in both the grafted RPE cells and the collected vitreous fluids of the monkeys. Mixed lymphocyte-RPE assay revealed that the infected iPS-RPE cells enhanced the proliferation of inflammatory cells in vitro. Conclusions: Transplantation of graft iPS-RPE cells contaminated with mycoplasma into the subretina caused severe ocular inflammation. Mycoplasma possesses the ability to cause immune responses in the host.


Assuntos
Transplante de Células/efeitos adversos , Infecções Oculares/microbiologia , Células-Tronco Pluripotentes Induzidas/citologia , Infecções por Mycoplasma/patologia , Mycoplasma/isolamento & purificação , Epitélio Pigmentado da Retina/transplante , Animais , Transplante de Células/métodos , DNA Forma C/análise , Modelos Animais de Doenças , Infecções Oculares/etiologia , Inflamação/patologia , Macaca fascicularis , Infecções por Mycoplasma/etiologia , Complicações Pós-Operatórias/microbiologia , Descolamento Retiniano/patologia , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/patologia , Vasculite Retiniana/patologia
15.
Eur Spine J ; 28(5): 1092-1112, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30666481

RESUMO

PURPOSE: It is an open question whether cell transplantation can provide safety and effective outcome to spinal cord injury (SCI) patient which has remained controversial for almost 40 years. This study aimed to evaluate the safety and efficacy of cell transplantation in SCI patients. METHOD: Studies of the cell transplantation for SCI were retrieved from PubMed, Embase, Medline, Cochrane Library and analyzed quantitative data by Review Manager 5.3. RESULTS: Twenty-one clinical controlled studies with 973 patients were included. The pooled results suggested that cell transplantation significantly improved ASIA score, ASIA motor score, ASIA sensory score, Barthel Index score, residual urine volume, rehabilitative time of automatic micturition. Furthermore, subgroup analysis indicated that the stem cells exhibited more potent than the non-stem cells in spinal cord repair. Cell transplantation at more than 14 days after injury showed more significant improvements than that within 14 days from injury. The dosage of cell transplantation between 1-5 × 107 and 10-20 × 107 was the potent quantity for the patient with SCI. Intrathecal injection and intravenous + intrathecal injection showed more superior to the other method. The top 5 adverse events were febrile reaction (11.5%), neurologic pain (11.3%), headache (2.6%), neurologic deterioration (2.4%), and rigidity or spasticity (1.6%). CONCLUSION: Cell transplantation appears to be a safe therapeutic strategy possessing substantial beneficial effects in the patients with SCI in clinic. Moreover, treating SCI with stem cell, the dosage of cells between 1-5 × 107 and 10-20 × 107, in intermediate or chronic phase, minimally invasive techniques, may bring more advantage to SCI patient. These slides can be retrieved under Electronic Supplementary Material.


Assuntos
Transplante de Células , Traumatismos da Medula Espinal/cirurgia , Transplante de Células/efeitos adversos , Transplante de Células/métodos , Transplante de Células/estatística & dados numéricos , Humanos , Complicações Pós-Operatórias
16.
Theranostics ; 9(1): 290-310, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30662568

RESUMO

Increasing evidence suggests the consensus that direct in vivo application of induced pluripotent stem cells (iPSCs) is infeasible may not be true. Methods: Teratoma formation and fate were examined in 53 normal and disease conditions involving brain, lung, liver, kidney, islet, skin, hind limb, and arteries. Results: Using classic teratoma generation assays, which require iPSCs to be congregated and confined, all mouse, human, and individualized autologous monkey iPSCs tested formed teratoma, while iPSC-derived cells did not. Intravenously or topically-disseminated iPSCs did not form teratomas with doses up to 2.5×108 iPSCs/kg and observation times up to 18 months, regardless of host tissue type; autologous, syngeneic, or immune-deficient host animals; presence or absence of disease; disease type; iPSC induction method; commercial or self-induced iPSCs; mouse, human, or monkey iPSCs; frequency of delivery; and sex. Matrigel-confined, but not PBS-suspended, syngeneic iPSCs delivered into the peritoneal cavity or renal capsule formed teratomas. Intravenously administered iPSCs were therapeutic with a dose as low as 5×106/kg and some iPSCs differentiated into somatic cells in injured organs. Disseminated iPSCs trafficked into injured tissue and survived significantly longer in injured than uninjured organs. In disease-free animals, no intravenously administered cell differentiated into an unwanted long-lasting cell or survived as a quiescent stem cell. In coculture, the stem cell medium and dominant cell-type status were critical for iPSCs to form cell masses. Conclusion: Teratoma can be easily and completely avoided by disseminating the cells. Direct in vivo iPSC application is feasible and can be safe.


Assuntos
Transplante de Células/efeitos adversos , Transplante de Células/métodos , Células-Tronco Pluripotentes Induzidas/transplante , Teratoma/epidemiologia , Estruturas Animais/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Haplorrinos , Camundongos , Modelos Teóricos , Teratoma/patologia
17.
Cell Transplant ; 28(1): 36-46, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30362373

RESUMO

Spinal cord injury (SCI) is a devastating disease, with a high rate of disability. In this meta-analysis, we aimed to comprehensively assess the efficacy and safety of mesenchymal stem cells (MSCs) in treating clinical SCI patients. We systematically searched the PUBMED, EMBASE, Chinese Biomedical (CBM), Web of Science and Cochrane databases using the strategy of combination of free-text words and MeSH terms. The indicators of the American Spinal Injury Association (ASIA) impairment scale (AIS)-grading improvement rate and adverse effects were displayed with an overall relative risk (RR). For the continuous variables of the ASIA motor score, light-touch score, pinprick score, activities of daily living (ADL) score, and residual urine volume, we used odds ratio (OR) to analyze the data. Eleven studies comprising 499 patients meeting all inclusion and exclusion criteria were included. No serious heterogeneity or publication bias was observed across each study. The results showed that significant improvements of total AIS grade (RR: 3.70; P < 0.001), AIS grade A (RR: 3.57; P < 0.001), ASIA sensory score (OR: 8.63; P < 0.001) and reduction of residual urine volume (OR: -36.37; P = 0.03) were observed in experimental group compared with control group. However, no significant differences of motor score (OR: 1.37, P = 0.19) and ADL score (OR: 2.61, P = 0.27) were observed between experimental and control groups. In addition, there were no serious and permanent adverse effects after cell transplantation. Cell transplantation with MSCs is effective and safe in improving the sensory and bladder functions of SCI patients.


Assuntos
Transplante de Células-Tronco Mesenquimais/efeitos adversos , Traumatismos da Medula Espinal/terapia , Transplante de Células/efeitos adversos , Resultado do Tratamento
18.
J Low Genit Tract Dis ; 23(1): 65-70, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30252710

RESUMO

OBJECTIVE: The aim of the study was to summarize and review the evidence for the efficacy and safety of adipose-derived stem cells (ADSCs) and platelet-rich plasma (PRP) for the treatment of vulvar lichen sclerosus (LS). MATERIALS AND METHODS: PubMed/MEDLINE, Ovid, Web of Science, and clinicaltrials.gov were searched from inception up to May 7, 2018. RESULTS: Seven observational studies were identified, with a total of 98 patients. Both ADSCs and PRP were reported to improve symptoms, quality of life measures, as well as clinical and histological signs of vulvar LS. There is a strong risk of biased estimates of treatment effect. CONCLUSIONS: Current evidence is weak for ADSCs and/or PRP as treatment for vulvar LS. Further research is needed before recommending this therapy.


Assuntos
Transplante de Células/métodos , Plasma Rico em Plaquetas , Líquen Escleroso Vulvar/terapia , Adulto , Idoso , Transplante de Células/efeitos adversos , Feminino , Humanos , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Estudos Observacionais como Assunto , Resultado do Tratamento
19.
Clin Chem ; 65(4): 519-529, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30593467

RESUMO

BACKGROUND: We have entered a new era of cancer therapy, with a number of immune-based therapies already used clinically as a standard of care. Adoptive cellular immunotherapy using T cells genetically modified with chimeric antigen receptors (CAR-T cells) represents a novel therapeutic approach. CAR-T cells have produced clinical responses in B-cell malignancies that are otherwise refractory to conventional therapies. Two CAR-T cell therapies obtained regulatory approval in 2017, with many more of these therapies under clinical development. CONTENT: This review focuses on the current state of adoptive cellular immunotherapy, specifically CAR-T cells, in the clinic and how this therapy differs from traditional small molecule and biologic therapies. Areas in which the clinical laboratory is affected by these novel therapies are discussed. Opportunities for the clinical laboratory to help guide these therapies are also highlighted. SUMMARY: The clinical laboratory will play an integral role in the care of patients undergoing adoptive cellular therapy with engineered T cells. There are many ways that this new therapeutic approach affects the clinical laboratory, and the clinical laboratory will likely play a critical role in managing patients that are treated with CAR-T cell therapy.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias/terapia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Transplante de Células/efeitos adversos , Transplante de Células/métodos , Humanos , Imunoterapia Adotiva/efeitos adversos , Neoplasias/imunologia , Receptores de Antígenos Quiméricos/genética
20.
Expert Rev Clin Immunol ; 14(12): 1013-1019, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30345839

RESUMO

INTRODUCTION: Conventional hemophilia treatment is based on repeated infusion of the missing clotting factor. This therapy is lifelong, expensive and can result in the formation of neutralizing antibodies, thus causing failure of the treatment and requiring higher doses of the replacement drug. Areas covered: Gene and cell therapies offer the advantage of providing a definitive and long-lasting correction of the mutated gene, promoting its physiological expression and preventing neutralizing antibody development. This review focuses on the most recent approaches that have been shown to prevent and even eradicate immune response toward the replaced factor. Expert commentary: Despite the encouraging data demonstrated by ongoing clinical trials and pre-clinical studies, more extensive investigations are necessary to establish the long-term safety and efficacy of gene therapy treatments in maintaining immune tolerance.


Assuntos
Anticorpos Neutralizantes/imunologia , Transplante de Células/métodos , Coagulantes/administração & dosagem , Fator VIII/biossíntese , Terapia Genética/métodos , Hemofilia A/terapia , Tolerância Imunológica , Animais , Transplante de Células/efeitos adversos , Coagulantes/efeitos adversos , Coagulantes/imunologia , Dependovirus/genética , Dependovirus/imunologia , Fator VIII/administração & dosagem , Fator VIII/genética , Fator VIII/imunologia , Edição de Genes , Técnicas de Transferência de Genes , Terapia Genética/efeitos adversos , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Hemofilia A/sangue , Hemofilia A/genética , Hemofilia A/imunologia , Humanos , Lentivirus/genética , Lentivirus/imunologia , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA