Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Cells ; 12(18)2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37759484

RESUMO

T cells expressing chimeric antigen receptors (CARs) are at the forefront of clinical treatment of cancers. Still, the nanoscale organization of CARs at the interface of CAR-Ts with target cells, which is essential for TCR-mediated T cell activation, remains poorly understood. Here, we studied the nanoscale organization of CARs targeting CD138 proteoglycans in such fixed and live interfaces, generated optimally for single-molecule localization microscopy. CARs showed significant self-association in nanoclusters that was enhanced in interfaces with on-target cells (SKOV-3, CAG, FaDu) relative to negative cells (OVCAR-3). CARs also segregated more efficiently from the abundant membrane phosphatase CD45 in CAR-T cells forming such interfaces. CAR clustering and segregation from CD45 correlated with the effector functions of Ca++ influx and target cell killing. Our results shed new light on the nanoscale organization of CARs on the surfaces of CAR-Ts engaging on- and off-target cells, and its potential significance for CAR-Ts' efficacy and safety.


Assuntos
Neoplasias Ovarianas , Receptores de Antígenos Quiméricos , Humanos , Feminino , Receptores de Antígenos Quiméricos/metabolismo , Apoptose , Linhagem Celular Tumoral , Sinapses/metabolismo
2.
Biomedicines ; 10(9)2022 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-36140319

RESUMO

High-grade serous ovarian carcinoma (HGSOC) is the most common type of epithelial ovarian cancer. The majority of cases are diagnosed at advanced stages, when intraperitoneal (IP) spread has already occurred. Despite significant surgical and chemotherapeutic advances in HGSOC treatment over the past decades, survival rates with HGSOC have only modestly improved. Chimeric antigen receptor (CAR)-T cells enable T cells to directly bind to tumor-associated antigens in a major histocompatibility complex-independent manner, thereby inducing tumor rejection. While CAR-T cell therapy shows great promise in hematological malignancies, its use in solid tumors is limited. Therefore, innovative approaches are needed to increase the specificity of CAR-modified T cells against solid tumors. The aim of this study was to assess the efficacy and safety of intraperitoneal (IP) versus intravenous (IV) CAR-T cell therapy in the treatment of HGSOC. We constructed a CAR that targets the ErbB2/HER2 protein (ErbB2CAR), which is overexpressed in HGSOC, and evaluated the functionality of ErbB2CAR on ovarian cancer cell lines (OVCAR8, SKOV3, and NAR). Our findings show that an IP injection of ErbB2CAR-T cells to tumor-bearing mice led to disease remission and increased survival compared to the IV route. Moreover, we found that IP-injected ErbB2CART cells circulate to a lesser extent, making them safer for non-tumor tissues than IV-injected cells. Further supporting our findings, we show that the effect of ErbB2CAR-T cells on primary HGSOC tumors is correlated with ErbB2 expression. Together, these data demonstrate the advantages of an IP administration of CAR-T cells over IV administration, offering not only a safer strategy but also the potential for counteracting the effect of ErbB2CAR in HGSOC. Significance: IP-injected ErbB2CAR-T cells led to disease remission and increased survival compared to the IV route. These findings demonstrate the advantages of IP administration, offering a safe treatment strategy with the potential for counteracting the effect of ErbB2CAR in HGSOC.

3.
Hum Gene Ther ; 32(19-20): 1224-1241, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34494460

RESUMO

Chimeric antigen receptor (CAR)-T cells are genetically engineered T cells, directed against a tumor-associated antigen. Extracellular vesicles (EVs) derived from CAR-T cells (CAR-T EVs) may preserve CAR-T activity and overcome one of the major obstacles responsible for CAR-T cell failure in patients with solid tumors. This study aimed to compare CAR-T EVs with their parental cells and explore their cell penetration and cytotoxic activity. Anti-HER-2 CARs were stimulated with specific target cells. EVs were isolated from the cell media and characterized for their content and functions. We found that CAR-T EVs contained a mixture of small and large EVs. Stimulated anti-HER-2+ CAR-T EVs expressed lower cytokine levels compared with their parental CAR-T cells (such as interferon gamma). Higher levels of granzyme B were found in CAR-T EVs (≥20 × ) compared with EVs from unstimulated cells (p < 0.001). Anti-HER-2+ CAR-T EVs bound and penetrated specifically into HER-2 expressing target cells. Similar cytotoxic effects measured by caspase-3/7 activity were found in CAR-T cells and their derived EVs. However, while the CAR-T cells induced massive apoptosis during the first 24 h, CAR-T EVs required 60 - 90 h. In summary, CAR-T EVs provide a novel potent immunotherapy approach that may be effective against solid tumors.


Assuntos
Vesículas Extracelulares , Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Imunoterapia Adotiva , Neoplasias/terapia , Receptores de Antígenos Quiméricos/genética , Linfócitos T
4.
Nat Commun ; 12(1): 3615, 2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-34127674

RESUMO

Glioblastoma is considered one of the most aggressive malignancies in adult and pediatric patients. Despite decades of research no curative treatment is available and it thus remains associated with a very dismal prognosis. Although recent pre-clinical and clinical studies have demonstrated the feasibility of chimeric antigen receptors (CAR) T cell immunotherapeutic approach in glioblastoma, tumor heterogeneity and antigen loss remain among one of the most important challenges to be addressed. In this study, we identify p32/gC1qR/HABP/C1qBP to be specifically expressed on the surface of glioma cells, making it a suitable tumor associated antigen for redirected CAR T cell therapy. We generate p32 CAR T cells and find them to recognize and specifically eliminate p32 expressing glioma cells and tumor derived endothelial cells in vitro and to control tumor growth in orthotopic syngeneic and xenograft mouse models. Thus, p32 CAR T cells may serve as a therapeutic option for glioblastoma patients.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/farmacologia , Glioma/imunologia , Glioma/terapia , Linfócitos T/imunologia , Idoso , Animais , Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Glioma/genética , Glioma/metabolismo , Humanos , Imunoterapia Adotiva , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteínas Mitocondriais/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/imunologia , Serina Endopeptidases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cancer Immunol Res ; 8(12): 1485-1495, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33008840

RESUMO

Chimeric antigen receptor (CAR) T-cell therapy has shown remarkable successes in fighting B-cell leukemias/lymphomas. Promising response rates are reported in patients treated with B-cell maturation antigen (BCMA) CAR T cells for multiple myeloma. However, responses appear to be nondurable, highlighting the need to expand the repertoire of multiple myeloma-specific targets for immunotherapy and to generate new CAR T cells. Here, we developed a "dual-CAR" targeting two multiple myeloma-associated antigens and explored its safety and efficacy. To reduce the "off-target" toxicity, we used the recognition of paired antigens that were coexpressed by the tumor to induce efficient CAR T-cell activation. The dual-CAR construct presented here was carefully designed to target the multiple myeloma-associated antigens, taking into consideration the distribution of both antigens on normal human tissues. Our results showed that the CD138/CD38-targeted dual CAR (dCAR138-38) elicited a potent anti-multiple myeloma response both in vitro and in vivo NSG mice transplanted with a multiple myeloma cell line and treated with dCAR138-38 showed median survival of 97 days compared with 31 days in the control group treated with mock-lymphocytes. The dCAR138-38 showed increased specificity toward cells expressing both targeted antigens compared with single-antigen-expressing cells and low activity toward primary cells from healthy tissues. Our findings indicated that the dCAR138-38 may provide a potent and safe alternative therapy for patients with multiple myeloma.


Assuntos
Antígeno de Maturação de Linfócitos B/imunologia , Mieloma Múltiplo/imunologia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Antígeno de Maturação de Linfócitos B/metabolismo , Linhagem Celular Tumoral , Citotoxicidade Imunológica/imunologia , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Imunoterapia , Imunoterapia Adotiva/métodos , Camundongos , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/metabolismo
6.
Cancer Immunol Immunother ; 69(7): 1165-1175, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32130452

RESUMO

Chimeric antigen receptor T cell (CAR-T) therapy is a novel approved treatment for hematological malignancies, still under development for solid tumors. Here, we use a rate equation-based mathematical model to discover regimens and schedules that maintain efficacy while potentially reducing toxicity by decreasing the amount of CAR-T infused. Tested on an in vivo murine model of spontaneous breast cancer, we show that our mathematical model accurately recapitulates in vivo tumor growth results achieved in the previous experiments. Moreover, we use the mathematical model to predict results of new therapy schedules and successfully prospectively validated these predictions in the in vivo. We conclude that using one tenth and even one percent of a full CAR-T dose used in preclinical trials can achieve efficacious results similar to full dose treatment.


Assuntos
Modelos Animais de Doenças , Imunoterapia Adotiva/métodos , Imunoterapia Adotiva/normas , Neoplasias Mamárias Experimentais/terapia , Modelos Teóricos , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Feminino , Neoplasias Mamárias Experimentais/imunologia , Camundongos
7.
Nanomedicine ; 14(3): 835-847, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29306001

RESUMO

Nanoparticulate vaccines are promising tools to overcome cancer immune evasion. However, a deeper understanding on nanoparticle-immune cell interactions and treatments regime is required for optimal efficacy. We provide a comprehensive study of treatment schedules and mode of antigen-association to nanovaccines on the modulation of T cell immunity in vivo, under steady-state and tumor-bearing mice. The coordinated delivery of antigen and two adjuvants (Monophosphoryl lipid A, oligodeoxynucleotide cytosine-phosphate-guanine motifs (CpG)) by nanoparticles was crucial for dendritic cell activation. A single vaccination dictated a 3-fold increase on cytotoxic memory-T cells and raised antigen-specific immune responses against B16.M05 melanoma. It generated at least a 5-fold increase on IFN-γ cytokine production, and presented over 50% higher lymphocyte count in the tumor microenvironment, compared to the control. The number of lymphocytes at the tumor site doubled with triple immunization. This lymphocyte infiltration pattern was confirmed in mammary huHER2 carcinoma, with significant tumor reduction.


Assuntos
Neoplasias da Mama/prevenção & controle , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Carcinogênese/efeitos dos fármacos , Nanopartículas/administração & dosagem , Linfócitos T Citotóxicos/imunologia , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Vacinas Anticâncer/química , Carcinogênese/metabolismo , Carcinogênese/patologia , Feminino , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química , Células Tumorais Cultivadas
8.
Mol Ther ; 22(5): 1018-28, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24686242

RESUMO

The adoptive transfer of regulatory T cells (Tregs) offers a promising strategy to combat pathologies that are characterized by aberrant immune activation, including graft rejection and autoinflammatory diseases. Expression of a chimeric antigen receptor (CAR) gene in Tregs redirects them to the site of autoimmune activity, thereby increasing their suppressive efficiency while avoiding systemic immunosuppression. Since carcinoembryonic antigen (CEA) has been shown to be overexpressed in both human colitis and colorectal cancer, we treated CEA-transgenic mice that were induced to develop colitis with CEA-specific CAR Tregs. Two disease models were employed: T-cell-transfer colitis as well as the azoxymethane-dextran sodium sulfate model for colitis-associated colorectal cancer. Systemically administered CEA-specific (but not control) CAR Tregs accumulated in the colons of diseased mice. In both model systems, CEA-specific CAR Tregs suppressed the severity of colitis compared to control Tregs. Moreover, in the azoxymethane-dextran sodium sulfate model, CEA-specific CAR Tregs significantly decreased the subsequent colorectal tumor burden. Our data demonstrate that CEA-specific CAR Tregs exhibit a promising potential in ameliorating ulcerative colitis and in hindering colorectal cancer development. Collectively, this study provides a proof of concept for the therapeutic potential of CAR Tregs in colitis patients as well as in other autoimmune inflammatory disorders.


Assuntos
Antígeno Carcinoembrionário/biossíntese , Colite/terapia , Neoplasias Colorretais/terapia , Linfócitos T Reguladores/transplante , Animais , Colite/complicações , Colite/genética , Neoplasias Colorretais/complicações , Neoplasias Colorretais/genética , Sulfato de Dextrana/administração & dosagem , Modelos Animais de Doenças , Humanos , Imunoterapia Adotiva/métodos , Camundongos , Linfócitos T Reguladores/metabolismo
9.
Cancer J ; 20(2): 123-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24667957

RESUMO

The nickname "T-body" is used to denote a T cell expressing an antigen-specific or antibody-based chimeric receptor that combines antibody specificity with T-cell effector or regulatory function. Initially, we designed and constructed chimeric antibody-based receptors and expressed them in T cells to study the role of major histocompatibility complex in triggering T-cell activation. To this end, we replaced both variable domains (Vα and Vß of the native T-cell receptor chains) with antibody-derived VH and VL sequences. After transfection into T cells, the 2 chimeric chains paired, associated with the CD3 complex, and endowed transfectants with non-major histocompatibility complex-restricted antibody type specificity. In subsequent studies, we developed next generation of chimeric antibody-based receptors by fusing an antibody single-chain variable fragment to T-cell activation motifs. This modular configuration simplified gene transfer, avoided mixed pairing with endogenous T-cell receptor chains, and enabled simultaneous insertion of various domains as costimulatory moieties to generate T-bodies with efficient antitumor reactivity.


Assuntos
Imunoterapia Adotiva , Neoplasias/enzimologia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Linfócitos T/imunologia , Complexo CD3/imunologia , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Complexo Principal de Histocompatibilidade/genética , Complexo Principal de Histocompatibilidade/imunologia , Neoplasias/genética , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
10.
Mol Ther ; 22(5): 1029-38, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24572294

RESUMO

Continuous oncogenic processes that generate cancer require an on-going treatment approach to eliminate the transformed cells, and prevent their further development. Here, we studied the ability of T cells expressing a chimeric antibody-based receptor (CAR) to offer a therapeutic benefit for breast cancer induced by erbB-2. We tested CAR-modified T cells (T-bodies) specific to erbB-2 for their antitumor potential in a mouse model overexpressing a human erbB-2 transgene that develops mammary tumors. Comparing the antitumor reactivity of CAR-modified T cells under various therapeutic settings, either prophylactic, prior to tumor development, or therapeutically. We found that repeated administration of CAR-modified T cells is required to eliminate spontaneously developing mammary cancer. Systemic, as well as intratumoral administered CAR-modified T cells accumulated at tumor sites and eventually eliminated the malignant cells. Interestingly, within a few weeks after a single CAR T cells' administration, and rejection of primary lesion, tumors usually relapsed both in treated mammary gland and at remote sites; however, repeated injections of CAR-modified T cells were able to control the secondary tumors. Since spontaneous tumors can arise repeatedly, especially in the case of syndromes characterized by specific susceptibility to cancer, multiple administrations of CAR-modified T cells can serve to control relapsing disease.


Assuntos
Neoplasias da Mama/terapia , Receptor ErbB-2/imunologia , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/transplante , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Imunoterapia Adotiva , Camundongos , Receptor ErbB-2/genética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nano Lett ; 12(9): 4992-6, 2012 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-22900991

RESUMO

Nanoscale organization of surface ligands often has a critical effect on cell-surface interactions. We have developed an experimental system that allows a high degree of control over the 2-D spatial distribution of ligands. As a proof of concept, we used the developed system to study how T-cell activation is independently affected by antigen density and antigen amount per cell. Arrays of submicrometer gold islands at varying surface coverage were defined on silicon by electron beam lithography (EBL). The gold islands were functionalized with alkanethiol self-assembled monolayers (SAMs) containing a small antigen, 2,4,6-trinotrophenyl (TNP), at various densities. Genetically engineered T-cell hybridomas expressing TNP-specific chimeric T-cell antigen receptor (CAR) were cultured on the SAMs, and their activation was assessed by IL-2 secretion and CD69 expression. It was found that, at constant antigen density, activation increased monotonically with the amount of antigen, while at constant antigen amount activation was maximal at an intermediate antigen density, whose value was independent of the amount of antigen.


Assuntos
Alcanos/química , Ouro/química , Imunoensaio/métodos , Nanopartículas/química , Análise Serial de Proteínas/métodos , Mapeamento de Interação de Proteínas/métodos , Compostos de Sulfidrila/química , Teste de Materiais , Impressão Molecular/métodos , Nanopartículas/ultraestrutura
12.
Gastroenterology ; 143(5): 1375-1384.e5, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22819865

RESUMO

BACKGROUND & AIMS: Pancreatic adenocarcinoma (PAC) is often diagnosed at an advanced and inoperable stage, and standard systemic treatments are generally ineffective. We investigated the effects of adoptive transfer of tumor-specific T cells that express chimeric antibody-based receptors (CAR) to mice with primary and metastatic PAC xenografts. METHODS: Human effector T cells were genetically modified to express CAR against Her2/neu or CD24, a putative PAC stem cell antigen. The antitumor reactivity of the engineered T cells (T-bodies) was evaluated in SCID mice with different PAC xenografts. A total of 1 × 10(7) T-bodies were injected via the tail vein or directly administered to the subcutaneous tumor on 3 or 4 alternating days. Mice were then given twice-daily intraperitoneal injections of interleukin-2 for 10 days. RESULTS: Intratumor injection of human CD24 and Her2/neu-specific T-bodies completely eliminated the tumors from most animals. Intravenous injection of T-bodies reduced tumor size and prolonged survival of mice with orthotopically transplanted tumors; more than 50% of animals appeared to be disease-free more than 2 months later. Additional systemic administration of T-bodies 8 weeks after the initial injection eliminated primary tumors, along with liver and draining lymph node metastases. A single administration of the Her2/neu-specific T-bodies prolonged the survival of mice with tumors in which most of the cells expressed the target antigen. In contrast, the CD24-specific T-bodies prolonged survival of mice in which only a subpopulation of the tumor cells expressed the antigen. CONCLUSIONS: CAR-redirected T cells stop growth and metastasis of PAC xenografts in mice. T-bodies specific to CD24, a putative cancer stem cell antigen, were effective against PAC xenografts that had only a subset of antigen-expressing cells.


Assuntos
Adenocarcinoma/terapia , Imunoterapia Adotiva , Neoplasias Pulmonares/terapia , Neoplasias Pancreáticas/terapia , Linfócitos T/transplante , Adenocarcinoma/imunologia , Adenocarcinoma/secundário , Animais , Antígeno CD24/imunologia , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/imunologia , Metástase Linfática , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Receptor ErbB-2/imunologia , Análise de Sobrevida , Linfócitos T/metabolismo
13.
Nano Lett ; 11(11): 4997-5001, 2011 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-21985491

RESUMO

Seamless embedment of electronic devices in biological systems is expected to add the outstanding computing power, memory, and speed of electronics to the biochemical toolbox of nature. Such amalgamation requires transduction of electronic signals into biochemical cues that affect cells. Inspired by biology, where pathways are directed by molecular recognition, we propose and demonstrate a generic electrical-to-biological transducer comprising a two-state electronic antigen and a chimeric cell receptor engineered to bind the antigen exclusively in its "on" state. T-cells expressing these receptors remain inactivated with the antigen in its "off" state. Switching the antigen to its "on" state by an electrical signal leads to its recognition by the T-cells and correspondingly to cell activation.


Assuntos
Receptores de Antígenos de Linfócitos T/efeitos da radiação , Anticorpos de Cadeia Única/efeitos da radiação , Linfócitos T/efeitos da radiação , Células Cultivadas , Campos Eletromagnéticos , Humanos , Receptores de Antígenos de Linfócitos T/química , Anticorpos de Cadeia Única/química , Linfócitos T/química
14.
Blood ; 118(4): 975-83, 2011 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-21653325

RESUMO

Adoptive cell transfer of allogeneic tumor-specific T cells could potentially be used as a universal treatment for cancer. We present a novel approach for adoptive immunotherapy using fully MHC-mismatched allogeneic T cells redirected with tumor-specific, non-MHC-restricted antibody-based chimeric antigen receptor (T-bodies) in the absence of GVHD. Mice bearing systemic metastatic disease were lymphodepleted by irradiation and treated with Her2/neu re-directed T cells. Lymphodepletion created a 'therapeutic window', which allowed the allo-T-bodies to attack the tumor before their rejection. A single split dose administration of allogeneic T-bodies extended the survival of tumor-bearing mice similarly to syngeneic T-bodies, and to a significantly greater extent than nonspecific allogeneic T cells. Blocking egress of lymphocytes from lymphoid organs using the sphingosine-1-phosphate agonist, FTY720, extended the persistence of allogeneic T cells such that allogeneic T-bodies provided superior therapeutic benefit relative to syngeneic ones, and dramatically extended the median survival time of the treated mice for more than a year. Therefore, we suggest that ex-vivo generated MHC-mismatched T-bodies can be used universally for off-the-shelf cancer immunotherapy and that their graft-versus-host reactivity can be safely harnessed to potentiate adoptive cell therapy.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias/terapia , Linfócitos T/imunologia , Linfócitos T/transplante , Animais , Antígenos de Neoplasias/imunologia , Separação Celular , Cloridrato de Fingolimode , Citometria de Fluxo , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Imunossupressores/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Transplante Homólogo
15.
Prostate ; 69(10): 1034-44, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19343735

RESUMO

Prostate cancer (PC) is a heterogeneous disease whose aggressive phenotype is the second leading cause of cancer-related death in men. The identification of key molecules and pathways that play a pivotal role in PC progression towards an aggressive form is crucial. A major effort towards this end has been taken by global analyses of gene expression profiles. However, the large body of data did not provide a definitive idea about the genes which are associated with the aggressive growth of PC. In order to identify such genes, we performed an interspecies comparison between several human data sets and high quality microarray data that we generated from the transgenic adenocarcinoma of mouse prostate (TRAMP) strain. The TRAMP PC mimics the histological and pathological appearance as well as the aggressive phenotype of human PC (huPC). Analysis of the microarray data, derived from microdissected TRAMP specimens removed at different stages of the disease yielded genetic signatures delineating the TRAMP PC development and progression. Comparison of the TRAMP data with a set of genes representing the core expression signature of huPC yielded a limited set genes. Some of these genes are known predictors of poor prognosis in huPC. Interestingly, the modulation of genes responsible for the invasive phenotype of huPC occurs in TRAMP already during the transition to prostate intraepithelial neoplasia (PIN) and onwards to localized tumors. We therefore suggest that critical oncogenic events leading to an aggressive phenotype of huPC can be studied in the PIN stage of TRAMP.


Assuntos
Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Animais , Progressão da Doença , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estadiamento de Neoplasias , Fenótipo , Neoplasias da Próstata/metabolismo , Membro 25 de Receptores de Fatores de Necrose Tumoral/biossíntese , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Especificidade da Espécie
16.
Gastroenterology ; 136(5): 1721-31, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19208357

RESUMO

BACKGROUND & AIMS: The therapeutic application of regulatory T cells (Tregs) for the treatment of inflammatory diseases is limited by the scarcity of antigen-specific Tregs. A preferred approach to endow effector T cells (Teff) with a desired specificity uses chimeric immune receptors with antibody-type specificity. Accordingly, employing such chimeric immune receptors to redirect Tregs to sites of inflammation may be a useful therapeutic approach to alleviate a broad scope of diseases in which an uncontrolled inflammatory response plays a major role. METHODS: To enable application of the approach in clinical setting, which requires the genetic modification of the patient's own Tregs, we describe here a novel protocol that allows the efficient retroviral transduction and 2,4,6-trinitrophenol-specific expansion of murine naturally occurring regulatory T cells (nTregs), with a 2,4,6-trinitrophenol-specific tripartite chimeric receptor. RESULTS: Transduced Tregs maintained their Foxp3 level, could undergo repeated expansion upon ex vivo encounter with their cognate antigen in a major histocompatibility complex-independent, costimulation-independent, and contact-dependent manner and specifically suppressed Teff cells. Adoptive transfer of small numbers of the transduced nTregs was associated with antigen-specific, dose-dependent amelioration of trinitrobenzenesulphonic acid colitis. CONCLUSIONS: This study demonstrates that nTregs can be efficiently transduced to express functional, antigen-specific chimeric receptors that enable the specific suppression of effector T cells both in vitro and in vivo. This approach may enable future cell-based therapeutic application in inflammatory bowel disease, as well as other inflammatory disorders.


Assuntos
Especificidade de Anticorpos , Colite/terapia , Engenharia Genética , Receptores de Antígenos/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Colite/imunologia , Feminino , Produtos do Gene env/imunologia , Produtos do Gene gag/imunologia , Produtos do Gene pol/imunologia , Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/imunologia , Retroviridae/genética
17.
Gastroenterology ; 134(7): 2014-24, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18424268

RESUMO

BACKGROUND & AIMS: Treatment with ex vivo expanded regulatory T cells (Tregs) is regarded as a promising therapeutic approach in inflammatory bowel disease but is hampered by impaired Treg accumulation and function at inflammatory sites. We aim to study whether antigen-specific redirected Tregs can overcome these limitations. METHODS: We developed transgenic mice whose T cells, including Tregs, express chimeric receptor (CR) made of antibody variable region as recognition unit and T-cell stimulatory and costimulatory domains to activate specifically in response to the predetermined model antigen 2,4,6-trinitrophenol (TNP). RESULTS: TNP-specific CR-bearing Tregs were potently and specifically activated by exogenous TNP and suppressed effector T cells in the absence of costimulatory B7-CD28 interaction. TNP-specific transgenic (Tg) mice were resistant to 2,4,6-trinitrobenzene sulphonic acid (TNBS) colitis but not to other hapten-mediated colitis. Adoptive transfer of CR-bearing Tregs to wild-type mice with TNBS colitis was associated with significant amelioration of colitis and improved survival. Although TNP-specific CR-bearing Tregs did not suppress oxazolone colitis, they cured it after addition of traces of TNBS to oxazolone-inflamed colons, demonstrating a "bystander" effect. In vivo imaging of adoptively transferred CR-bearing Tregs demonstrated that they preferentially migrate to TNBS-induced colonic mucosal lesions within hours of induction of colitis. CONCLUSIONS: Tregs can be redirected with specificity distinct from that of pathogenic lymphocytes, accumulate at colonic inflammatory lesions, and suppress effector T cells in a specific, nonmajor histocompatibility complex-restricted, and noncostimulatory-dependent manner, resulting in significant amelioration of colitis. Hopefully, this approach will lead to a novel therapy for inflammatory bowel disease, as well as other inflammatory diseases.


Assuntos
Colite/terapia , Colo/imunologia , Imunoterapia Adotiva , Ativação Linfocitária , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Reguladores/transplante , Animais , Efeito Espectador , Antígenos CD28/imunologia , Linhagem Celular , Movimento Celular , Colite/induzido quimicamente , Colite/imunologia , Colite/patologia , Colite/prevenção & controle , Colo/patologia , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Antígenos de Histocompatibilidade/imunologia , Imunidade nas Mucosas , Região Variável de Imunoglobulina/imunologia , Mucosa Intestinal/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Oxazolona , Picratos/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de IgG/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Reguladores/imunologia , Fatores de Tempo , Ácido Trinitrobenzenossulfônico
18.
Prostate ; 68(5): 530-9, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18247403

RESUMO

BACKGROUND: Small cell carcinoma of the prostate (SCCP) is a rare subset of prostate cancer (0.5-2% of all prostatic carcinomas), predominantly composed of neuroendocrine (NE) cells, with a very poor prognosis. Irradiation is one of the mainstay options for SCCP local treatment, yet, little is known about the clinical response of these aggressive tumors to radiotherapy. METHODS: Using SCID mice, the response to fractionated ionizing radiation (IR) of two unique human NE xenografts of SCCP (WISH-PC2 and WM-4A) was investigated. RESULTS: Fractionated irradiation of WISH-PC2 xenografts using total doses of >24 Gy induced a delay in tumor growth, while total doses of >36 Gy led to local tumor eradication. However, most of the irradiated mice suffered from disseminated metastases. Similarly, in the WM-4A xenograft, a total dose of 20 Gy led to tumor growth delay and some of the mice also developed metastases. Non-irradiated local xenografts failed to disseminate, even following surgical excision of the main tumor mass; however, tumor cells administered intravenously did form metastases. Metastases of both xenografts were located in the adrenal/kidney and inter-scapular regions, areas rich in brown adipose tissue. A correlation was found between the appearance of irradiation-induced metastases and activation of the gelatinase activity of matrix metalloproteinase-9. CONCLUSIONS: Clinically, this study raises the possibility that radiation to SCCP may promote metastatic disease. For patients in whom prostate biopsy shows a predominance of small cell cancer, it may be necessary to deliver systemic therapy together with the radiotherapy in order to prevent the development of metastases.


Assuntos
Carcinoma de Células Pequenas/patologia , Carcinoma de Células Pequenas/radioterapia , Proliferação de Células/efeitos da radiação , Neoplasias da Próstata/patologia , Neoplasias da Próstata/radioterapia , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias das Glândulas Suprarrenais/secundário , Animais , Carcinoma de Células Pequenas/metabolismo , Modelos Animais de Doenças , Humanos , Neoplasias Renais/secundário , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos SCID , Metástase Neoplásica/radioterapia , Neoplasias da Próstata/metabolismo
19.
Blood ; 105(8): 3087-93, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15626734

RESUMO

Chimeric receptor (CR)-redirected lymphocytes (T bodies) have great potential in the eradication of tumor cells. To extend this approach to target cells that do not express surface ligands to costimulatory receptors (eg, cancer cells), we have generated an antibody-based tripartite chimeric receptor (TPCR) that contains scFv linked to the costimulatory molecule, CD28 without its ligand-binding domain, and to the cytoplasmic moiety of the FcRgamma subunit. In this study, we tested the ability of 2,4,6-trinitrophenyl (TNP)-specific TPCR to drive primary, naive T cells derived from CR-transgenic (Tg) mice to undergo full activation. As a control, we used Tg mice expressing a similar transgene but lacking the signaling region of CD28 (Tg-TPCRDeltaCD28). Only T cells from the TPCR-Tg and not the CD28-truncated TPCR-Tg mice could undergo activation following stimulation on hapten-modified target cells not expressing B7. Moreover, when stimulated with TNP protein displayed on plastic, the TPCR-Tg T cells expressing the entire TPCR gene became fully activated for proliferation, interleukin 2 production, protection from apoptosis, and killing of TNP-modified target cells. Finally, TPCR-Tg mice manifested a delayed-type hypersensitivity response following skin challenge in the absence of priming. Taken together, our results suggest that the TPCR is the receptor configuration of choice for clinical applications using primary T or stem cells.


Assuntos
Antígenos CD28/genética , Ativação Linfocitária/imunologia , Linfoma de Células B/imunologia , Proteínas Recombinantes de Fusão/genética , Linfócitos T/imunologia , Animais , Anticorpos/imunologia , Apoptose/imunologia , Antígeno B7-1/metabolismo , Antígenos CD28/metabolismo , Linhagem Celular Tumoral , Imunoterapia/métodos , Interleucina-2/metabolismo , Subunidade alfa de Receptor de Interleucina-2 , Linfoma de Células B/terapia , Camundongos , Camundongos Transgênicos , Receptores de Interleucina/genética , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Linfócitos T/citologia , Linfócitos T/metabolismo
20.
Discov Med ; 5(27): 259-64, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20704885

RESUMO

Extract: Metastasis of the bone is common in two of the major gender-specific malignancies -- breast and prostate cancers. Although both primary breast and prostate cancer are manageable by "classical" therapies such as surgery, irradiation, and chemotherapy, when metastases (secondary cancers) disseminate to the bones these diseases are, by and large, incurable. Metastasis to the bone is implicated in around 70% of prostate and breast cancer deaths. The idea of harnessing the immune system to fight disseminated cancer has proven to be effective in experimental animal models against transplanted tumors. Here, both arms of the immune system, namely, the humoral one characterized by anti-tumor antibodies and the cellular one composed of a type of white blood cell, specifically the cytotoxic T-lymphocytes (CTLs), were found to cause tumor rejection either following immunization of the experimental mice before the tumor inoculations (active-vaccination) or after adoptive transfer of cancer-specific antibodies or CTLs into tumor-bearing mice (passive-vaccination). Encouraged by these results, scientists and clinicians have joined forces in extensive efforts to apply both active and passive vaccination for the immunotherapy of cancer patients. These attempts have flourished over the last fifteen years following the discovery of the first human tumor antigens in melanoma patients.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...