Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Oncol ; 32(1): 200775, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38596311

RESUMO

Chimeric antigen receptor (CAR) T cell therapies targeting B cell-restricted antigens CD19, CD20, or CD22 can produce potent clinical responses for some B cell malignancies, but relapse remains common. Camelid single-domain antibodies (sdAbs or nanobodies) are smaller, simpler, and easier to recombine than single-chain variable fragments (scFvs) used in most CARs, but fewer sdAb-CARs have been reported. Thus, we sought to identify a therapeutically active sdAb-CAR targeting human CD22. Immunization of an adult Llama glama with CD22 protein, sdAb-cDNA library construction, and phage panning yielded >20 sdAbs with diverse epitope and binding properties. Expressing CD22-sdAb-CAR in Jurkat cells drove varying CD22-specific reactivity not correlated with antibody affinity. Changing CD28- to CD8-transmembrane design increased CAR persistence and expression in vitro. CD22-sdAb-CAR candidates showed similar CD22-dependent CAR-T expansion in vitro, although only membrane-proximal epitope targeting CD22-sdAb-CARs activated direct cytolytic killing and extended survival in a lymphoma xenograft model. Based on enhanced survival in blinded xenograft studies, a lead CD22sdCAR-T was selected, achieving comparable complete responses to a benchmark short linker m971-scFv CAR-T in high-dose experiments. Finally, immunohistochemistry and flow cytometry confirm tissue and cellular-level specificity of the lead CD22-sdAb. This presents a complete report on preclinical development of a novel CD22sdCAR therapeutic.

2.
Front Immunol ; 13: 864868, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35935988

RESUMO

Epidermal growth factor family receptor (EGFR) is commonly overexpressed in many solid tumors and an attractive target for chimeric antigen receptor (CAR)-T therapy, but as EGFR is also expressed at lower levels in healthy tissues a therapeutic strategy must balance antigenic responsiveness against the risk of on-target off-tumor toxicity. Herein, we identify several camelid single-domain antibodies (also known as nanobodies) that are effective EGFR targeting moieties for CARs (EGFR-sdCARs) with very strong reactivity to EGFR-high and EGFR-low target cells. As a strategy to attenuate their potent antigenic sensitivity, we performed progressive truncation of the human CD8 hinge commonly used as a spacer domain in many CAR constructs. Single amino acid hinge-domain truncation progressively decreased both EGFR-sdCAR-Jurkat cell binding to EGFR-expressing targets and expression of the CD69 activation marker. Attenuated signaling in hinge-truncated EGFR-sdCAR constructs increased selectivity for antigen-dense EGFR-overexpressing cells over an EGFR-low tumor cell line or healthy donor derived EGFR-positive fibroblasts. We also provide evidence that epitope location is critical for determining hinge-domain requirement for CARs, as hinge truncation similarly decreased antigenic sensitivity of a membrane-proximal epitope targeting HER2-CAR but not a membrane-distal EGFRvIII-specific CAR. Hinge-modified EGFR-sdCAR cells showed clear functional attenuation in Jurkat-CAR-T cells and primary human CAR-T cells from multiple donors in vitro and in vivo. Overall, these results indicate that hinge length tuning provides a programmable strategy for throttling antigenic sensitivity in CARs targeting membrane-proximal epitopes, and could be employed for CAR-optimization and improved tumor selectivity.


Assuntos
Receptores de Antígenos Quiméricos , Anticorpos de Domínio Único , Epitopos , Receptores ErbB , Humanos , Imunoterapia Adotiva/métodos , Receptor ErbB-2/genética , Receptores de Antígenos Quiméricos/genética , Linfócitos T
3.
Fluids Barriers CNS ; 19(1): 38, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35650594

RESUMO

Human blood brain barrier (BBB) models derived from induced pluripotent stem cells (iPSCs) have become an important tool for the discovery and preclinical evaluation of central nervous system (CNS) targeting cell and gene-based therapies. Chimeric antigen receptor (CAR)-T cell therapy is a revolutionary form of gene-modified cell-based immunotherapy with potential for targeting solid tumors, such as glioblastomas. Crossing the BBB is an important step in the systemic application of CAR-T therapy for the treatment of glioblastomas and other CNS malignancies. In addition, even CAR-T therapies targeting non-CNS antigens, such as the well-known CD19-CAR-T therapies, are known to trigger CNS side-effects including brain swelling due to BBB disruption. In this study, we used iPSC-derived brain endothelial-like cell (iBEC) transwell co-culture model to assess BBB extravasation of CAR-T based immunotherapies targeting U87MG human glioblastoma (GBM) cells overexpressing the tumor-specific mutated protein EGFRvIII (U87vIII). Two types of anti-EGFRvIII targeting CAR-T cells, with varying tonic signaling profiles (CAR-F263 and CAR-F269), and control Mock T cells were applied on the luminal side of BBB model in vitro. CAR-F263 and CAR-F269 T cells triggered a decrease in transendothelial electrical resistance (TEER) and an increase in BBB permeability. CAR-T cell extravasation and U87vIII cytotoxicity were assessed from the abluminal compartment using flow cytometry and Incucyte real-time viability imaging, respectively. A significant decrease in U87vIII cell viability was observed over 48 h, with the most robust cytotoxicity response observed for the constitutively activated CAR-F263. CAR-F269 T cells showed a similar cytotoxic profile but were approximately four fold less efficient at killing the U87vIII cells compared to CAR-F263, despite similar transmigration rates. Visualization of CAR-T cell extravasation across the BBB was further confirmed using BBTB-on-CHIP models. The described BBB assay was able to discriminate the cytotoxic efficacies of different EGFRvIII-CARs and provide a measure of potential alterations to BBB integrity. Collectively, we illustrate how BBB models in vitro can be a valuable tool in deciphering the mechanisms of CAR-T-induced BBB disruption, accompanying toxicity and effector function on post-barrier target cells.


Assuntos
Glioblastoma , Receptores de Antígenos Quiméricos , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Imunoterapia , Receptores de Antígenos Quiméricos/metabolismo
4.
Mol Ther Methods Clin Dev ; 16: 238-254, 2020 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-32083149

RESUMO

Chimeric antigen receptor (CAR) development involves extensive empirical characterization of antigen-binding domain (ABD)/CAR constructs for clinical suitability. Here, we present a cost-efficient and rapid method for evaluating CARs in human Jurkat T cells. Using a modular CAR plasmid, a highly efficient ABD cloning strategy, plasmid electroporation, short-term co-culture, and flow-cytometric detection of CD69, this assay (referred to as CAR-J) evaluates sensitivity and specificity for ABDs. Assessing 16 novel anti-CD22 single-chain variable fragments derived from mouse monoclonal antibodies, CAR-J stratified constructs by response magnitude to CD22-expressing target cells. We also characterized 5 novel anti-EGFRvIII CARs for preclinical development, identifying candidates with varying tonic and target-specific activation characteristics. When evaluated in primary human T cells, tonic/auto-activating (without target cells) EGFRvIII-CARs induced target-independent proliferation, differentiation toward an effector phenotype, elevated activity against EGFRvIII-negative cells, and progressive loss of target-specific response upon in vitro re-challenge. These EGFRvIII CAR-T cells also showed anti-tumor activity in xenografted mice. In summary, CAR-J represents a straightforward method for high-throughput assessment of CAR constructs as genuine cell-associated antigen receptors that is particularly useful for generating large specificity datasets as well as potential downstream CAR optimization.

6.
J Leukoc Biol ; 103(2): 355-368, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29345354

RESUMO

Selectin-ligand interactions are important for leukocyte homing and functionality. The roles of selectin-ligand interactions in modulating immunity to intracellular infections are not completely understood. Mice lacking the expression of fucosyltransferase-IV and -VII (Fucosyltransferase-IV and -VII double knockout, FtDKO) exhibit deficient functionality of selectin-ligand interactions. We addressed the kinetics of infection and immunity to Listeria monocytogenes (LM), an intracellular pathogen, in FtDKO mice. These mice exhibited enhanced ability to clear infection and increased survival to a lethal dose of LM infection relative to wild-type (WT) C57BL/6J controls. This was associated with increased levels of neutrophils, monocytes, and dendritic cells (DCs) in the blood and/or infected organs. Adoptive transfer of bone marrow (BM) cells from FtDKO mice to WT mice resulted in enhanced neutrophil numbers and improved clearance of LM bacteria in recipients. In vivo depletion of myeloid innate immune cells, particularly neutrophils, monocytes, macrophages, and DCs, using anti-Ly-6G (RB6-8C5) monoclonal antibody, reduced the ability of FtDKO mice to curtail LM infection. Nevertheless, depletion using anti-Ly-6G (1A8) known to exclusively deplete neutrophils did not abrogate increased resistance of FtDKO mice to LM infection, suggesting a role for other myeloid innate immune cells in this model. Examination of BM hematopoietic progenitors through flow cytometry and cell culture colony-forming unit assay showed increased frequencies of granulocyte-macrophage progenitors in FtDKO relative to WT mice, Overall, our results indicate that functional selectin ligand deficiency enhances innate immune-mediated resistance to systemic LM infection despite defective leukocyte migration and lymphocyte homing.


Assuntos
Imunidade Inata/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Selectinas/imunologia , Animais , Citocinas/sangue , Citocinas/imunologia , Fucosiltransferases/genética , Humanos , Ligantes , Listeriose/sangue , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Selectinas/genética
7.
mBio ; 6(5): e01421-15, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26396246

RESUMO

UNLABELLED: Nontyphoidal Salmonella enterica (NTS) infections are a major burden to global public health, as they lead to diseases ranging from gastroenteritis to systemic infections and there is currently no vaccine available. Here, we describe a highly effective component vaccine against S. enterica serovar Typhimurium in both gastroenteritis and systemic murine infection models. We devised an approach to generate supernatants of S. enterica serovar Typhimurium, an organism that is highly abundant in virulence factors. Immunization of mice with this supernatant resulted in dramatic protection against a challenge with serovar Typhimurium, showing increased survival in the systemic model and decreased intestinal pathology in the gastrointestinal model. Protection correlated with specific IgA and IgG levels in the serum and specific secretory IgA levels in the feces of immunized mice. Initial characterization of the protective antigens in the bacterial culture supernatants revealed a subset of antigens that exhibited remarkable stability, a highly desirable characteristic of an effective vaccine to be used under suboptimal environmental conditions in developing countries. We were able to purify a subset of the peptides present in the supernatants and show their potential for immunization of mice against serovar Typhimurium resulting in a decreased level of colonization. This component vaccine shows promise with regard to protecting against NTS, and further work should significantly help to establish vaccines against these prevalent infections. IMPORTANCE: Salmonella enterica infections other than typhoid and paratyphoid fever are a major global health burden, as they cause high morbidity and mortality worldwide. Strategies that prevent Salmonella-related diseases are greatly needed, and there is a significant push for the development of vaccines against nontyphoidal Salmonella enterica serovars. In this work, we describe an S. Typhimurium supernatant-derived vaccine that is effective in reducing bacterial colonization in mouse models of gastroenteritis as well as invasive disease. This is a component vaccine that shows high stability to heat, a feature that is important for use under suboptimal conditions, such as those found in sub-Saharan Africa.


Assuntos
Meios de Cultura/química , Infecções por Salmonella/prevenção & controle , Vacinas contra Salmonella/administração & dosagem , Vacinas contra Salmonella/imunologia , Salmonella typhimurium/imunologia , Animais , Anticorpos Antibacterianos/sangue , Bacteriemia/microbiologia , Bacteriemia/prevenção & controle , Modelos Animais de Doenças , Fezes/química , Gastroenterite/microbiologia , Gastroenterite/prevenção & controle , Imunoglobulina A/sangue , Imunoglobulina A Secretora/análise , Imunoglobulina G/sangue , Intestinos/patologia , Camundongos , Infecções por Salmonella/microbiologia , Vacinas contra Salmonella/isolamento & purificação , Salmonella typhimurium/crescimento & desenvolvimento , Análise de Sobrevida , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/isolamento & purificação
8.
Cell Rep ; 2(6): 1710-21, 2012 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-23219554

RESUMO

Pathogens that reside in the phagosomes of infected cells persist despite the presence of potent T cell responses. We addressed the mechanism of immune evasion by using a mouse model of Salmonella typhimurium (ST). Recombinants of ST were generated that translocated antigen to the cytosol or phagosomes of infected cells. We find that the kinetics of antigen presentation and CD8(+) T cell priming is accelerated by cytosolic antigen delivery, although the magnitude of CD8(+) T cell response is not influenced by antigenic location. More importantly, only those targets that readily display antigen on the cell surface, owing to antigenic translocation to the cytosol, are recognized and killed by CD8(+) T cells. Thus, vaccination approaches developed to control phagosomal pathogens should incorporate methods for modulating antigen presentation such that infected target cells can be readily recognized by CD8(+) T cells.


Assuntos
Apresentação de Antígeno , Antígenos de Bactérias/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunidade Celular , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Doença Aguda , Animais , Antígenos de Bactérias/genética , Linfócitos T CD8-Positivos/patologia , Doença Crônica , Camundongos , Camundongos Transgênicos , Infecções por Salmonella/genética , Infecções por Salmonella/patologia , Salmonella typhimurium/genética
9.
Am J Reprod Immunol ; 63(1): 54-65, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19912156

RESUMO

PROBLEM: Listeria monocytogenes (LM) preferentially colonizes the placenta and causes fetal loss and systemic disease during pregnancy. As systemic CD8+ T-cell memory is critical in controlling LM infection, we addressed the issue as to whether it is modulated during pregnancy. METHOD OF STUDY: Pregnant mice were infected with LM and their immune response was quantified relative to the non-pregnant cohort using advanced immunological techniques. RESULTS: Pregnant mice exhibited progressive and massive placental LM infection leading to fetal resorptions. In contrast, they harbored significantly lower bacteria in spleen and liver relative to non-pregnant controls, and rapidly cleared systemic infection. Both pregnant and non-pregnant mice exhibited similar activation of systemic innate immunity. Moreover, LM infection in pregnant and non-pregnant hosts evoked strong antigen-specific cytolytic CD8+ T cells that produced IFN-gamma. Consequently, LM infection initiated during pregnancy afforded long-term protective memory to secondary infection. CONCLUSION: Maternal hosts generate a normal Listeria-specific adaptive immunity in particular CD8+ T-cell memory response suggesting that systemic listeriosis during pregnancy may be an immunopathology associated with placental infection.


Assuntos
Imunidade Adaptativa , Linfócitos T CD8-Positivos/imunologia , Células Matadoras Naturais/imunologia , Listeria monocytogenes , Listeriose/imunologia , Doenças Placentárias/imunologia , Animais , Contagem de Colônia Microbiana , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Gravidez/imunologia , Padrões de Referência
10.
J Immunol ; 178(4): 2396-406, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17277146

RESUMO

Vaccines capable of eliciting long-term T cell immunity are required for combating many diseases. Live vectors can be unsafe whereas subunit vaccines often lack potency. We previously reported induction of CD8(+) T cells to Ag entrapped in archaeal glycerolipid vesicles (archaeosomes). In this study, we evaluated the priming, phenotype, and functionality of the CD8(+) T cells induced after immunization of mice with OVA-Methanobrevibacter smithii archaeosomes (MS-OVA). A single injection of MS-OVA evoked a profound primary response but the numbers of H-2K(b)OVA(257-264)-specific CD8(+) T cells declined by 14-21 days, and <1% of primarily central phenotype (CD44(high)CD62L(high)) cells persisted. A booster injection of MS-OVA at 3-11 wk promoted massive clonal expansion and a peak effector response of approximately 20% splenic/blood OVA(257-264)-specific CD8(+) T cells. Furthermore, contraction was protracted and the memory pool (IL-7Ralpha(high)) of approximately 5% included effector (CD44(high)CD62L(low)) and central (CD44(high)CD62L(high)) phenotype cells. Recall response was observed even at >300 days. CFSE-labeled naive OT-1 (OVA(257-264) TCR transgenic) cells transferred into MS-OVA-immunized recipients cycled profoundly (>90%) within the first week of immunization indicating potent Ag presentation. Moreover, approximately 25% cycling of Ag-specific cells was seen for >50 days, suggesting an Ag depot. In vivo, CD8(+) T cells evoked by MS-OVA killed >80% of specific targets, even at day 180. MS-OVA induced responses similar in magnitude to Listeria monocytogenes-OVA, a potent live vector. Furthermore, protective CD8(+) T cells were induced in TLR2-deficient mice, suggesting nonengagement of TLR2 by archaeal lipids. Thus, an archaeosome adjuvant vaccine represents an alternative to live vectors for inducing CD8(+) T cell memory.


Assuntos
Adjuvantes Imunológicos , Linfócitos T CD8-Positivos/imunologia , Receptores de Hialuronatos/imunologia , Memória Imunológica , Selectina L/imunologia , Methanobrevibacter/imunologia , Receptor 2 Toll-Like/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Apresentação de Antígeno/imunologia , Feminino , Listeria monocytogenes/imunologia , Camundongos , Camundongos Knockout , Ovalbumina/imunologia , Receptores de Interleucina-7/imunologia , Fatores de Tempo , Receptor 2 Toll-Like/deficiência
11.
J Exp Med ; 197(4): 515-26, 2003 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-12591908

RESUMO

Natural resistance to infection with mouse cytomegalovirus (MCMV) is controlled by a dominant locus, Cmv1. Cmv1 is linked to the Ly49 family of natural killer receptors on distal chromosome 6. While some studies localized Cmv1 as distal to the Ly49 gene cluster, genetic and functional analysis identified Ly49h as a pivotal factor in resistance to MCMV. The role of these two independent genomic domains in MCMV resistance was evaluated by functional complementation using transgenesis of bacterial artificial chromosomes (BAC) in genetically susceptible mice. Phenotypic and genetic characterization of the transgenic animals traced the resistance gene to a single region spanning the Ly49h gene. The appearance of the Ly49H protein in NK cells of transgenic mice coincided with the emergence of MCMV resistance, and there was a threshold Ly49H protein level associated with full recovery. Finally, transgenic expression of Ly49H in the context of either of the two independent susceptibility alleles, Cmv1(sBALB) or Cmv1(sFVB), conferred resistance to MCMV infection. These results demonstrate that Ly49h is necessary and sufficient to confer MCMV resistance, and formally demonstrate allelism between Cmv1 and Ly49h. This panel of transgenic animals provides a unique resource to study possible pleiotropic effect of Cmv1.


Assuntos
Antígenos Ly/fisiologia , Predisposição Genética para Doença , Infecções por Herpesviridae/imunologia , Células Matadoras Naturais/imunologia , Muromegalovirus , Transgenes , Animais , Antígenos Ly/genética , Cromossomos Artificiais Bacterianos , Teste de Complementação Genética , Infecções por Herpesviridae/genética , Imunidade Inata/genética , Lectinas Tipo C , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Subfamília A de Receptores Semelhantes a Lectina de Células NK , Receptores Semelhantes a Lectina de Células NK
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...