Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
FASEB J ; 33(10): 11458-11468, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31343935

RESUMO

Among the earliest protein products of most cellular genes are those synthesized during the pioneer round of translation (PRT), a key step in nonsense-mediated mRNA decay (NMD) that allows scanning of new transcripts for the presence of a premature termination codon (PTC). It has been demonstrated that at least some PRT degradation products can be targeted to major histocompatibility (MHC)-I presentation. To gain new insight into this putative PRT-to-MHC-I route, we have assembled 2 pairs of reporter genes so that the 2 genes in each pair encode an identical fusion protein between a model antigenic peptide and enhanced green fluorescent protein (EGFP), one of which harbors a PTC. We expressed these genes in different mouse and human cell lines and confirmed enhanced NMD activity for the PTC(+) gene in each pair by monitoring the effect of cycloheximide on the level of the respective mRNA. We then exploited several strategies for establishing the ratio between level of peptide presentation and total amount of protein product. We consistently observed significantly higher ratios for the PTC(+) mRNAs compared with the PTC(-) ones, pointing to correlation between the turnover of otherwise identical proteins and the fate of their template mRNA. Using confocal microscopy, we showed a clear link between NMD, the presence of misfolded EGFP polypeptides, and enhanced MHC-I peptide presentation. Altogether, these findings imply that identical full-length gene products differing only in 3' noncoding sequences can be differentially degraded and targeted to the MHC-I presentation pathway, suggesting a more general role for the PRT in establishing the MHC-I peptidome.-Weinstein-Marom, H., Hendel, L., Laron, E. A., Sharabi-Nov, A., Margalit, A., Gross, G. MHC-I presentation of peptides derived from intact protein products of the pioneer round of translation.


Assuntos
Antígenos de Histocompatibilidade Classe I/genética , Peptídeos/genética , Biossíntese de Proteínas/genética , Animais , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Códon sem Sentido/genética , Genes Reporter/genética , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Camundongos , Degradação do RNAm Mediada por Códon sem Sentido/genética , RNA Mensageiro/genética
2.
J Immunol ; 201(10): 2959-2968, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30305327

RESUMO

New strategies for augmenting the actual performance of therapeutic T cells in vivo are needed for improving clinical outcome of adoptive cell therapy. Cumulative findings suggest that CD40 plays an intrinsic role in T cell costimulation. Recently, we demonstrated the ability of truncated, auto-oligomerizing CD40 derivatives to induce strong activation of APCs in a ligand-independent manner. We reasoned that constitutively active CD40 (caCD40) can similarly exert enhancing effects on human antitumor T cells. To test this assumption, we transfected human T cells with in vitro-transcribed caCD40 mRNA. In polyclonal T cells, caCD40 triggered IFN-γ secretion and upregulated CD25 and 4-1BB. In antimelanoma tumor-infiltrating lymphocytes (TILs), caCD40 induced massive production of IFN-γ, exerting a pronounced synergistic effect when coexpressed with constitutively active TLR4 devoid of its extracellular ligand binding. In unselected "young" TILs, caCD40 reproducibly increased surface expression of CD25, OX40, 4-1BB, CD127, and CD28. Three days post-mRNA electroporation of CD8 TILs, caCD40 elevated IFN-γ and TNF-α production and cytolytic activity in the presence of autologous but not HLA-I-mismatched melanoma. Enhanced killing of autologous melanoma by young TILs was observed 4 d posttransfection. These findings suggest that caCD40 can function as a potent T cell adjuvant and provide essential guidelines for similar manipulation of other key members of the TNFR family.


Assuntos
Antígenos CD40/imunologia , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos T/imunologia , Adjuvantes Imunológicos , Humanos , Imunoterapia Adotiva/métodos , Melanoma/imunologia , RNA Mensageiro , Neoplasias Cutâneas/imunologia , Células Tumorais Cultivadas
3.
Annu Rev Pharmacol Toxicol ; 56: 59-83, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26738472

RESUMO

A chimeric antigen receptor (CAR) is a recombinant fusion protein combining an antibody-derived targeting fragment with signaling domains capable of activating T cells. Recent early-phase clinical trials have demonstrated the remarkable ability of CAR-modified T cells to eliminate B cell malignancies. This review describes the choice of target antigens and CAR manipulations to maximize antitumor specificity. Benefits and current limitations of CAR-modified T cells are discussed, with a special focus on the distribution of tumor antigens on normal tissues and the risk of on-target, off-tumor toxicities in the clinical setting. We present current methodologies for pre-evaluating these risks and review the strategies for counteracting potential off-tumor effects. Successful implementation of these approaches will improve the safety and efficacy of CAR T cell therapy and extend the range of cancer patients who may be treated.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Receptores de Antígenos de Linfócitos T/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Antígenos de Neoplasias/imunologia , Antineoplásicos/imunologia , Humanos , Neoplasias/imunologia , Neoplasias/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
4.
Mol Ther ; 25(2): 456-464, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28109957

RESUMO

Chimeric major histocompatibility complex (MHC) molecules supplemented with T cell receptor (TCR) signaling motifs function as activation receptors and can redirect gene-modified T cells against pathogenic CD8 T cells. We have shown that ß2 microglobulin (ß2m) operates as a universal signaling component of MHC-I molecules when fused with the CD3-ζ chain. Linking the H-2Kd-binding insulin B chain peptide insulin B chain, amino acids 15-23 (InsB15-23) to the N terminus of ß2m/CD3-ζ, redirected polyclonal CD8 T cells against pathogenic CD8 T cells in a peptide-specific manner in the non-obese diabetic (NOD) mouse. Here, we describe mRNA electroporation for delivering peptide/ß2m/CD3-ζ genes to a reporter T cell line and purified primary mouse CD8 T cells. The peptide/ß2m/CD3-ζ products paired with endogenous MHC-I chains and transmitted strong activation signals upon MHC-I cross-linking. The reporter T cell line transfected with InsB15-23/ß2m/CD3-ζ mRNA was activated by an InsB15-23-H-2Kd-specific CD8 T cell hybrid only when the transfected T cells expressed H-2Kd. Primary NOD CD8 T cells expressing either InsB15-23/ß2m/CD3-ζ or islet-specific glucose-6-phosphatase catalytic subunit-related protein, amino acids 206-214 (IGRP206-214)/ß2m/CD3-ζ killed their respective autoreactive CD8 T cell targets in vitro. Furthermore, transfer of primary CD8 T cells transfected with InsB15-23/ß2m/CD3-ζ mRNA significantly reduced insulitis and protected NOD mice from diabetes. Our results demonstrate that mRNA encoding chimeric MHC-I receptors can redirect effector CD8 against diabetogenic CD8 T cells, offering a new approach for the treatment of type 1 diabetes.


Assuntos
Transferência Adotiva , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Imunomodulação , RNA Mensageiro/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Citotoxicidade Imunológica , Diabetes Mellitus Tipo 1/prevenção & controle , Diabetes Mellitus Tipo 1/terapia , Modelos Animais de Doenças , Feminino , Expressão Gênica , Ordem dos Genes , Vetores Genéticos/genética , Insulina/imunologia , Camundongos , Camundongos Endogâmicos NOD , Complexo Receptor-CD3 de Antígeno de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Transfecção , Microglobulina beta-2/genética
5.
Mol Ther ; 23(8): 1391-1400, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25997427

RESUMO

Recently, we have developed a novel genetic platform for improving dendritic cell (DC) induction of peptide-specific CD8 T cells, based on membrane-anchored ß2-microglobulin (ß2m) linked to a selected antigenic peptide at its N-terminus and to the cytosolic domain of toll-like receptor (TLR)4 C-terminally. In vitro transcribed mRNA transfection of antigen presenting cells resulted in polypeptides that efficiently coupled peptide presentation to cellular activation. In the present study, we evaluated the immunogenicity of such constructs in mRNA-transfected immature murine bone marrow-derived DCs. We show that the encoded peptide ß2m-TLR4 products were expressed at the cell surface up to 72 hours and stimulated the maturation of DCs. In vivo, these DCs prompted efficient peptide-specific T-cell activation and target cell killing which were superior to those induced by peptide-loaded, LPS-stimulated DCs. This superiority was also evident in the ability to protect mice from tumor progression following the administration of B16F10.9 melanoma cells and to inhibit the development of pre-established B16F10.9 tumors. Our results provide evidence that the products of two recombinant genes encoding for peptide-hß2m-TLR4 and peptide-hß2m-K(b) expressed from exogenous mRNA can cooperate to couple Major Histocompatibility Complex (MHC-I) peptide presentation to TLR-mediated signaling, offering a safe, economical and highly versatile genetic platform for a novel category of CTL-inducing vaccines.


Assuntos
Células Dendríticas/citologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Peptídeos/química , RNA Mensageiro/metabolismo , Receptor 4 Toll-Like/metabolismo , Microglobulina beta-2/metabolismo , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/citologia , Antígenos de Neoplasias/química , Antineoplásicos/química , Células da Medula Óssea/citologia , Vacinas Anticâncer/imunologia , Membrana Celular/metabolismo , Clonagem Molecular , Meios de Cultura , Citosol/metabolismo , Feminino , Humanos , Lipopolissacarídeos/química , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína , Transdução de Sinais
6.
Immunology ; 143(4): 609-17, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24943731

RESUMO

The key role played by islet-reactive CD8 and CD4 T cells in type 1 diabetes calls for new immunotherapies that target pathogenic T cells in a selective manner. We previously demonstrated that genetically linking the signalling portion of CD3-ζ onto the C-terminus of ß2 -microglobulin and an autoantigenic peptide to its N-terminus converts MHC-I complexes into functional T-cell receptor-specific receptors. CD8 T cells expressing such receptors specifically killed diabetogenic CD8 T cells, blocked T-cell-induced diabetes in immunodeficient NOD.SCID mice and suppressed disease in wild-type NOD mice. Here we describe the immunotargeting of CD4 T cells by chimeric MHC-II receptors. To this end we chose the diabetogenic NOD CD4 T-cell clone BDC2.5, which recognizes the I-A(g7) -bound 1040-31 mimotope. We assembled several constructs encoding I-A(g7) α- and ß-chains, the latter carrying mim or hen egg lysozyme peptide as control, each supplemented with CD3-ζ intracellular portion, either with or without its transmembrane domain. Following mRNA co-transfection of reporter B3Z T cells and mouse CD8 and CD4 T cells, these constructs triggered robust activation upon I-A(g7) cross-linking. A BDC2.5 T-cell hybridoma activated B3Z transfectants expressing the mimotope, but not the control peptide, in both configurations. Potent two-way activation was also evident with transgenic BDC2.5 CD4 T cells, but peptide-specific activation required the CD3-ζ transmembrane domain. Chimeric MHC-II/CD3-ζ complexes therefore allow the selective immunotargeting of islet-reactive CD4 T cells, which take part in the pathogenesis of type 1 diabetes.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Marcação de Genes , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Peptídeos/genética , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo
7.
Methods Mol Biol ; 2748: 167-186, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38070115

RESUMO

Genetic modification of tumor-infiltrating lymphocytes (TILs) or circulating T cells has become an important avenue in cancer therapy. Here we describe a comprehensive method for establishing and expanding TIL cultures and genetically modifying them with a gene of interest (GOI) via retroviral transduction or mRNA transfection. The method includes all the important steps starting with TIL extraction from tumors through to the maintenance of the genetically modified TILs. The protocol includes instructions for retroviral transduction and mRNA transfection of circulating T cells or T-cell lines. The GOIs most commonly introduced into the target cells are chimeric antigen receptors (CARs); genetic adjuvants, such as membrane-bound interleukins; and antitumor T-cell receptors (TCRs).


Assuntos
Linfócitos do Interstício Tumoral , Linfócitos T , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos T/metabolismo , Transfecção , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linhagem Celular , Linfócitos T CD8-Positivos , Imunoterapia Adotiva/métodos
8.
Front Immunol ; 14: 1227133, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37731505

RESUMO

Introduction: In this study, we report a novel therapeutic approach using B lymphocytes to attract islet-specific T cells in the non-obese diabetic (NOD) mouse model and prevent the development of autoimmune diabetes. Rather than using the antibody receptor of B cells, this approach utilizes their properties as antigen-presenting cells to T cells. Methods: Purified splenic B cells were treated with lipopolysaccharide, which increases regulatory B (Breg) cell function, then electroporated with mRNA encoding either chimeric MHC-I or MHC-II molecules covalently linked to antigenic peptides. Immunoregulatory functions of these engineered B cells (e-B cells) were tested by in vitro assays and in vivo co-transfer experiments with beta-cell-antigen-specific CD8+ or CD4+ T cells in NOD.Scid mice, respectively. Results: The e-B cells expressing chimeric MHC-I-peptide inhibited antigen-specific CD8+ T-cell cytotoxicity in vitro. The e-B cells expressing chimeric MHC-II-peptide induced antigen-specific CD4+ T cells to express the regulatory markers, PD-1, ICOS, CTLA-4, Lag3, and Nrp1. Furthermore, e-B cells encoding the chimeric MHC-I and MHC-II peptide constructs protected NOD.Scid mice from autoimmune diabetes induced by transfer of antigen-specific CD8+ and CD4+ T cells. Discussion: MHC-peptide chimeric e-B cells interacted with pathogenic T cells, and protected the host from autoimmune diabetes, in a mouse model. Thus, we have successfully expressed MHC-peptide constructs in B cells that selectively targeted antigen-specific cells, raising the possibility that this strategy could be used to endow different protective cell types to specifically regulate/remove pathogenic cells.


Assuntos
Linfócitos B Reguladores , Diabetes Mellitus Tipo 1 , Ilhotas Pancreáticas , Imunodeficiência Combinada Severa , Camundongos , Animais , Diabetes Mellitus Tipo 1/prevenção & controle , Camundongos Endogâmicos NOD , Camundongos SCID , Antígenos de Histocompatibilidade Classe II
9.
Int Immunol ; 23(7): 453-61, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21652516

RESUMO

Priming of naive CD8 T cells by dendritic cells (DCs) entails both effective antigen presentation on MHC class I products and co-stimulatory signaling. Their optimal coupling is a major goal in the development of CTL-inducing vaccines. We recently reported that a membranal derivative of the invariant MHC-I light chain, ß(2)-microglobulin (ß(2)m), markedly stabilizes MHC-I molecules and can serve as a universal platform for exceptional presentation of genetically linked peptides. To test whether it is possible to equip the resulting MHC-I complexes with an inherent ability to activate antigen-presenting cells, we engrafted the intracellular Toll/IL-1 receptor domain of mouse Toll-like receptor (TLR) 4 or TLR2 onto the peptide-ß(2)m scaffold. We evaluated the level of peptide presentation and status of cell activation conferred by such constructs in stably transfected mouse RAW264.7 macrophages and mRNA-transfected mouse DC2.4 DCs. We show that the encoded peptide-ß(2)m-TLR polypeptides are expressed at the cell surface, pair with endogenous heavy chains, stabilize MHC-I products, prompt efficient peptide-specific T-cell recognition and confer a constitutively activated phenotype on the transfected cells, as judged by the up-regulation of pro-inflammatory genes and surface co-stimulatory molecules. Our results provide evidence that the product of a single recombinant gene can couple MHC peptide presentation to TLR-mediated signaling and offer a safe, economical and highly versatile modality for a novel category of genetic CTL-inducing vaccines.


Assuntos
Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Antígenos H-2/imunologia , Peptídeos/imunologia , Animais , Vetores Genéticos/genética , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peptídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Transfecção , Microglobulina beta-2/genética , Microglobulina beta-2/imunologia
10.
Front Immunol ; 12: 780399, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34804073

RESUMO

Targeting solid tumors with absolute precision is a long-standing challenge in cancer immunotherapy. The identification of antigens, which are expressed by a large fraction of tumors of a given type and, preferably, across various types, but not by normal cells, holds the key to developing safe, off-the-shelf immunotherapies. Although the quest for widely shared, strictly tumor-specific antigens has been the focus of tremendous effort, only few such candidates have been implicated. Almost all antigens that are currently explored as targets for chimeric antigen receptor (CAR) or T cell receptor (TCR)-T cell therapy are also expressed by healthy cells and the risk of on-target off-tumor toxicity has remained a major concern. Recent studies suggest that this risk could be obviated by targeting instead combinations of two or more antigens, which are co-expressed by tumor but not normal cells and, as such, are tumor-specific. Moreover, the expression of a shared tumor antigen along with the lack of a second antigen that is expressed by normal tissues can also be exploited for precise recognition. Additional cues, antigenic or non-antigenic ones, which characterize the tumor microenvironment, could be harnessed to further increase precision. This review focuses on attempts to define the targetable signatures of tumors and assesses different strategies employing advanced synthetic biology for translating such information into safer modes of immunotherapy, implementing the principles of Boolean logic gates.


Assuntos
Imunoterapia Adotiva/métodos , Lógica , Terapia de Alvo Molecular/métodos , Neoplasias/terapia , Medicina de Precisão/métodos , Animais , Humanos
11.
J Immunother ; 44(6): 209-213, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34010245

RESUMO

The costimulatory domains incorporated into second-generation and third-generation chimeric antigen receptors (CARs) strongly influence CAR-T-cell function. Here, we explored second-generation and third-generation CARs harboring the signaling domain of the CD40 receptor as a new costimulatory element in comparison with similar CARs carrying the 4-1BB domain. In CARs of both generations, CD40 was more potent than 4-1BB in triggering the NF-κB signaling pathway. In human T cells from 2 donors, CD40 was comparable to 4-1BB in upregulating costimulatory and activation markers, inducing proinflammatory cytokine secretion and mediating target cell killing. Interestingly, differences in the response pattern of T cells from the 2 donors with respect to CD40 and 4-1BB were evident. We conclude that in human T cells, the CD40 signaling domain is a potent costimulatory element in both second-generation and third-generation CARs.


Assuntos
Antígenos CD40/imunologia , Domínios e Motivos de Interação entre Proteínas/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Antígenos CD40/química , Antígenos CD40/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular , Engenharia Genética , Humanos , Imunoterapia Adotiva/métodos , Ativação Linfocitária/imunologia , Plasmídeos/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/metabolismo
12.
J Autoimmun ; 35(4): 390-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20850948

RESUMO

Insulin is one of the earliest targeted autoantigens in the immune destruction of insulin-producing beta cells by autoreactive CD4 and CD8 T cells in type 1 diabetes. In this study, we used Non-obese diabetic (NOD) transgenic T cells engineered to express MHC class I-insulin peptide complexes linked to a T cell activation component (InsCD3-ζ), to target insulin-reactive CD8 T cells. We showed that activated, but not naïve, InsCD3-ζ CD8 T cells killed diabetogenic insulin-reactive CD8 target cells in vitro, inducing antigen-specific cell death mediated via both the release of perforin and the Fas-Fas ligand pathway. In vivo, InsCD3-ζ CD8 T cells migrated to the pancreatic lymph nodes of NOD mice after adoptive transfer. Concomitant with this, infiltration of CD8 T cells was also reduced in the pancreatic islets. Finally, in vivo, we showed that diabetes induced by adoptive transfer of insulin-reactive T cells was reduced following injection of activated InsCD3-ζ CD8 T cells. Furthermore, young NOD mice injected with InsCD3-ζ CD8 T cells developed a lower incidence and delayed onset of diabetes. Thus, using this novel system we have demonstrated that InsCD3-ζ CD8 T cells can directly kill insulin-reactive CD8 T cells in vitro and by targeting insulin-specific CD8 T cells early in the course of disease alter the progression of spontaneous diabetes in vivo in NOD mice.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Insulina/metabolismo , Fragmentos de Peptídeos/metabolismo , Transferência Adotiva , Animais , Autoantígenos/imunologia , Complexo CD3/genética , Complexo CD3/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Citotoxicidade Imunológica/genética , Diabetes Mellitus Tipo 1/terapia , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Insulina/genética , Insulina/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Fragmentos de Peptídeos/genética , Proteínas Recombinantes de Fusão/genética
13.
Front Immunol ; 11: 584148, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33488585

RESUMO

Adoptive T cell therapy (ACT) holds great promise for cancer treatment. One approach, which has regained wide interest in recent years, employs antitumor T cells isolated from tumor lesions ("tumor-infiltrating lymphocytes" or TIL). It is now appreciated that a considerable proportion of anti-melanoma TIL recognize new HLA-binding peptides resulting from somatic mutations, which occurred during tumor progression. The clinical efficacy of TIL can potentially be improved via their genetic modification, designed to enhance their survival, homing capacity, resistance to suppression, tumor killing ability and additional properties of clinical relevance. Successful implementation of such gene-based strategies critically depends on efficient and reproducible protocols for gene delivery into clinical TIL preparations. Here we describe an optimized protocol for the retroviral transduction of TIL. As the experimental system we employed anti-melanoma TIL cultures prepared from four patients, recombinant retrovirus encoding an anti-CD19 chimeric antigen receptor (CAR) as a model gene of interest and CD19+ and CD19- human cell lines serving as target cells. Transduction on day 7 of the rapid expansion protocol (REP) resulted in 69 ± 8% CAR positive TIL. Transduced, but not untransduced TIL, from the four patients responded robustly to CD19+, but not CD19- cell lines, as judged by substantial secretion of IFN-γ following co-culture. In light of the rekindled interest in antitumor TIL, this protocol can be incorporated into a broad range of gene-based approaches for improving the in-vivo survival and functionality of TIL in the clinical setting.


Assuntos
Linfócitos do Interstício Tumoral/imunologia , Retroviridae/imunologia , Antígenos CD19/imunologia , Linhagem Celular Tumoral , Humanos , Imunoterapia Adotiva/métodos , Interferon gama/imunologia , Células K562 , Ativação Linfocitária/imunologia , Melanoma/imunologia , Receptores de Antígenos Quiméricos/imunologia
14.
Biochem Biophys Res Commun ; 386(2): 402-6, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19538939

RESUMO

HIV-1 gp120 is an alleged B cell superantigen, binding certain VH3+ human antibodies. We reasoned that a CD4-VH3 fusion protein could possess higher affinity for gp120 and improved HIV-1 inhibitory capacity. To test this we produced several human IgG1 immunoligands harboring VH3. Unlike VH3-IgG1 or VH3-CD4-IgG1, CD4-VH3-IgG1 bound gp120 considerably stronger than CD4-IgG1. CD4-VH3-IgG1 exhibited approximately 1.5-2.5-fold increase in neutralization of two T-cell laboratory-adapted strains when compared to CD4-IgG1. CD4-VH3-IgG1 improved neutralization of 7/10 clade B primary isolates or pseudoviruses, exceeding 20-fold for JR-FL and 13-fold for Ba-L. It enhanced neutralization of 4/8 clade C viruses, and had negligible effect on 1/4 clade A pseudoviruses. We attribute this improvement to possible pairing of VH3 with CD4 D1 and stabilization of an Ig Fv-like structure, rather than to superantigen interactions. These novel findings support the current notion that CD4 fusion proteins can act as better HIV-1 entry inhibitors with potential clinical implications.


Assuntos
Antígenos CD4/imunologia , Proteína gp120 do Envelope de HIV/imunologia , HIV-1/imunologia , Imunoglobulina G/imunologia , Região Variável de Imunoglobulina/imunologia , Proteínas Recombinantes de Fusão/imunologia , Superantígenos/imunologia , Sequência de Aminoácidos , Antígenos CD4/genética , Humanos , Imunoglobulina G/genética , Região Variável de Imunoglobulina/genética , Ligantes , Dados de Sequência Molecular , Testes de Neutralização , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética
15.
Transpl Immunol ; 55: 101202, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30904624

RESUMO

Graft-versus-host disease (GVHD) and transplant rejection as a result of host-versus-graft (HVG) response have remained two major complications of allogeneic hematopoietic stem cell transplantation (allo-HSCT). When donors are partially HLA-mismatched unrelated or haploidentical related, their severity correlates with the degree of HLA disparity. Specific elimination of alloreactive donor or recipient T cells targeting the mismatched HLA products could markedly alleviate both complications while only minimally affecting graft-versus-tumor (GVT) response or engraftment. To redirect human CD8 T cells against alloreactive CD8 T cells we electroporate these cells with in-vitro-transcribed mRNA encoding MHC-I heavy chains fused with the signaling portion of CD3ζ. Here we show that peripheral blood human CD8 T cells expressing H-2Kb/CD3ζ or H-2Kd/CD3ζ respond to anti-MHC-I stimuli in a strictly specific manner. This study paves the way for further advancing this approach as a means to dampen GVHD and HVG that are caused by HLA disparity in allo-HSCT.


Assuntos
Complexo CD3/imunologia , Linfócitos T CD8-Positivos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Antígenos H-2/imunologia , Complexo CD3/genética , Linfócitos T CD8-Positivos/patologia , Eletroporação , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/patologia , Antígenos H-2/genética , Humanos , Células Jurkat
16.
J Immunother ; 42(2): 43-50, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30489430

RESUMO

Adoptive transfer of tumor-infiltrating lymphocytes (TILs) or gene-modified T cells expressing antitumor TCRs or chimeric antigen receptors often yields a high rate of clinical response in several types of cancer. New approaches for enhancing the functional properties of antitumor T cells could improve the clinical outcome of these treatments. To this end, we created 3 classes of genes, each designed to operate autonomously upon expression in T cells. We recently reported on the enhancing effects of constitutively active toll-like receptor 4 (caTLR4), membrane (mem) interleukin-2, memIL-12, and memIL-15, and self-oligomerizing, constitutively active CD40 (caCD40). Here, we evaluated their combined effects on peripheral blood CD8 T cells and different antimelanoma TIL cultures following mRNA electroporation. Expression in CD8 T cells induced transient production of interferon-γ and prolonged and robust upregulation of CD25, CD69, 4-1BB, and OX40. The adjuvants enhanced cytolytic activity of TILs and production of interferon-γ and TNF-α in the presence of autologous, but not mismatched, melanoma for at least 3 days after electroporation. Expression of the 3 adjuvants in young TILs from different patients markedly increased the expression of CD25, OX40, 4-1BB, CD127, and CD28 and exhibited cooperative and, at times, synergistic effects. Furthermore, predefined mixtures of mRNA encoding these adjuvants markedly enhanced the specific antitumor response of selected TILs and killing of autologous melanoma cells by young TILs. Our findings suggest that combinations of these new genetic adjuvants can substantially improve the functional properties of antitumor T cells, offering a new tool of unique versatility in adoptive cell therapy.


Assuntos
Linfócitos T CD8-Positivos/transplante , Eletroporação , Técnicas de Transferência de Genes , Imunoterapia Adotiva , Linfócitos do Interstício Tumoral/transplante , RNA Mensageiro/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Células Cultivadas , Humanos , Interferon gama , Melanoma/terapia
18.
J Immunother ; 40(2): 39-50, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28005579

RESUMO

The interaction between the CD40 receptor on antigen-presenting cells (APCs) and its trimeric ligand on CD4 T cells is essential for the initiation and progression of the adaptive immune response. Here we undertook to endow CD40 with the capacity to trigger spontaneous APC activation through ligand-independent oligomerization. To this end we exploited the GCN4 yeast transcriptional activator, which contains a leucine zipper DNA-binding motif that induces homophilic interactions. We incorporated GCN4 variants forming homodimers, trimers, or tetramers at the intracellular domain of human and mouse CD40 and replaced the extracellular portion with peptide-ß2m or other peptide tags. In parallel we examined similarly truncated CD40 monomers lacking a GCN4 motif. The oligomeric products appeared to arrange in high-molecular-weight aggregates and were considerably superior to the monomer in their ability to trigger nuclear factor kB signaling, substantiating the anticipated constitutively active (ca) phenotype. Cumulative results in human and mouse APC lines transfected with caCD40 mRNA revealed spontaneous upregulation of CD80, IL-1ß, TNFα, IL-6, and IL-12, which could be further enhanced by caTLR4 mRNA. In mouse bone-marrow-derived dendritic cells caCD40 upregulated CD80, CD86, MHC-II, and IL-12 and in human monocyte-derived dendritic cells it elevated surface CD80, CD83 CD86, CCR7, and HLA-DR. Oligomeric products carrying the peptide-ß2m extracellular portion could support MHC-I presentation of the linked peptide up to 4 days post-mRNA transfection. These findings demonstrate that the expression of a single caCD40 derivative in APCs can exert multiple immunostimulatory effects, offering a new powerful tool in the design of gene-based cancer vaccines.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/metabolismo , Vacinas Anticâncer/genética , Células Dendríticas/fisiologia , Saccharomyces cerevisiae/genética , Animais , Apresentação de Antígeno , Antígenos CD/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Antígenos CD40/genética , Diferenciação Celular , Linhagem Celular , Citocinas/metabolismo , Engenharia Genética , Antígenos de Histocompatibilidade/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Zíper de Leucina/genética , Ativação Linfocitária , Camundongos , NF-kappa B/metabolismo , Multimerização Proteica/genética , Proteínas de Saccharomyces cerevisiae/genética , Transdução de Sinais , Regulação para Cima
19.
Mol Immunol ; 42(9): 1099-109, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15829299

RESUMO

The envelope glycoprotein of HIV-1 is the principal target for entry inhibitors. The use of soluble CD4 has been found to be impractical as most clinical isolates are resistant to neutralization at feasible concentrations. CG10 is one of a small group of monoclonal antibodies specific to CD4-induced epitopes, which are structurally associated with the chemokine receptor-binding site and are capable of blocking the interaction of gp120 with its obligatory co-receptor. We have reasoned that fusing the single chain Fv of CG10 with CD4 can lead to increased HIV-1 neutralization activity and that this effect could be further enhanced by engrafting this chimeric construct onto an IgG Fc. Here we report the cloning of the genes encoding the variable regions of CG10 heavy and light chains and demonstrate that when attached to human IgG1 Fc, the single chain Fv of CG10 retains the binding properties of the original mouse antibody. Fusing CG10 single chain Fv with the gp120-binding portion of CD4 on a human IgG1 Fc backbone results in stronger binding of gp120 of different tropisms and in enhanced neutralization of laboratory-adapted strains and most, but not all, clade B and clade C isolates tested. Our findings underscore the potential use of CD4-based fusion proteins in the design of HIV immuno-therapeutics.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos CD4/imunologia , HIV-1/imunologia , Imunoglobulina G/imunologia , Antígenos CD4/genética , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Proteína gp120 do Envelope de HIV/imunologia , Humanos , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Testes de Neutralização , Receptores de HIV/imunologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/isolamento & purificação , Transfecção
20.
J Immunother ; 39(2): 60-70, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26849075

RESUMO

Proinflammatory cytokines are widely explored in different adoptive cell therapy protocols for enhancing survival and function of the transferred T cells, but their systemic administration is often associated with severe toxicity which limits their clinical use. To confine cytokine availability to the therapeutic T cells, we expressed 3 key cytokines, IL-2, IL-12, and IL-15, as integral T-cell membrane proteins. To prevent permanent activation of growth signaling pathways, we delivered these genes to T cells through mRNA electroporation. The engineered cytokines could be detected on the surface of mRNA-transfected cells and binding to their cell-surface receptors mainly occurred in cis. The 3 human cytokines supported the ex vivo growth of activated human CD8 and CD4 T cells for at least 6 days posttransfection, comparably to high-dose soluble IL-2. Similarly, membrane IL-2, membrane IL-12, and, to a lesser extent, membrane IL-15, were comparable with their soluble counterparts in supporting proliferation of splenic mouse CD8 T cells. Following electroporation of human CD8 T cells and antimelanoma tumor-infiltrating lymphocytes, membrane cytokines synergized with constitutively active toll-like receptor 4 in inducing interferon-γ secretion. Efficient cooperation with TLR4 was also evident in the upregulation of the activation molecules CD25, CD69, CD137 (4-1BB), and CD134 (OX40). Taken together, membrane cytokines expressed through mRNA transfection emerge as effective tools for enhancing T-cell proliferation and function and may have potential use in adoptive T-cell therapy.


Assuntos
Membrana Celular/metabolismo , Citocinas/metabolismo , Imunoterapia Adotiva/métodos , Linfócitos T/fisiologia , Receptor 4 Toll-Like/metabolismo , Animais , Linhagem Celular , Citocinas/genética , Eletroporação , Feminino , Humanos , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Engenharia de Proteínas , RNA Mensageiro/genética , Linfócitos T/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA