Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Br J Haematol ; 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38894496

RESUMO

Chimeric antigen receptor (CAR) T-cell therapies have increased the patients with relapsed/refractory multiple myeloma (RRMM) in whom standard electrophoretic techniques fail to detect the M-protein. Quantitative immunoprecipitation mass spectrometry (QIP-MS) can accurately measure serum M-protein with high sensitivity, and identify interferences caused by therapeutic monoclonal antibodies. Here, we investigate the outcome of QIP-MS in 33 patients treated with the academic BCMA-directed CAR T-cell ARI0002h (Cesnicabtagene Autoleucel). QIP-MS offered more detailed insights than serum immunofixation (sIFE), identifying glycosylated M-proteins and minor additional peaks. Moreover, the potential interferences owing to daratumumab or tocilizumab treatments were successfully detected. When analysing different assay platforms during patient's monitoring after ARI0002h administration, we observed that QIP-MS showed a high global concordance (78.8%) with sIFE, whereas it was only moderate (55.6%) with bone marrow (BM)-based next-generation flow cytometry (NGF). Furthermore, QIP-MS consistently demonstrated the lowest negativity rate across the different timepoints (27.3% vs. 60.0% in months 1 and 12, respectively). Patients with QIP-MS(+)/BM-based NGF(-) showed a non-significant shorter median progression free survival than those with QIP-MS(-)/BM-based NGF(-). In summary, we show the first experience to our knowledge demonstrating that QIP-MS could be particularly useful as a non-invasive technique when evaluating response after CAR T-cell treatment in MM.

2.
Cancer Res ; 83(21): 3562-3576, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37578274

RESUMO

Parkin is an E3 ubiquitin ligase, which plays a key role in the development of Parkinson disease. Parkin defects also occur in numerous cancers, and a growing body of evidence indicates that Parkin functions as a tumor suppressor that impedes a number of cellular processes involved in tumorigenesis. Here, we generated murine and human models that closely mimic the advanced-stage tumors where Parkin deficiencies are found to provide deeper insights into the tumor suppressive functions of Parkin. Loss of Parkin expression led to aggressive tumor growth, which was associated with poor tumor antigen presentation and limited antitumor CD8+ T-cell infiltration and activation. The effect of Parkin deficiency on tumor growth was lost following depletion of CD8+ T cells. In line with previous findings, Parkin deficiency was linked with mitochondria-associated metabolic stress, PTEN degradation, and enhanced Akt activation. Increased Akt signaling led to dysregulation of antigen presentation, and treatment with the Akt inhibitor MK2206-2HCl restored antigen presentation in Parkin-deficient tumors. Analysis of data from patients with clear cell renal cell carcinoma indicated that Parkin expression was downregulated in tumors and that low expression correlated with reduced overall survival. Furthermore, low Parkin expression correlated with reduced patient response to immunotherapy. Overall, these results identify a role for Parkin deficiency in promoting tumor immune evasion that may explain the poor prognosis associated with loss of Parkin across multiple types of cancer. SIGNIFICANCE: Parkin prevents immune evasion by regulating tumor antigen processing and presentation through the PTEN/Akt network, which has important implications for immunotherapy treatments in patients with Parkin-deficient tumors.


Assuntos
Apresentação de Antígeno , Neoplasias , Animais , Humanos , Camundongos , Imunoterapia , Neoplasias/genética , Neoplasias/terapia , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-akt , Evasão Tumoral , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
3.
Front Immunol ; 14: 1083303, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36742300

RESUMO

Adoptive cell therapies, like tumor-infiltrating lymphocytes or chimeric antigen receptor T cells, have become an important immunotherapeutic approach against cancer. One of the main struggles of T cell immunotherapies is how to obtain the most effective T cell phenotype, persistence, and differentiation potential to infuse into patients. Adjusting the T cell ex vivo cell culture conditions is a key factor to increase and improve the efficacy of cellular immunotherapies. In this review, we have summarized the ex vivo impact of short chain fatty acids, a group of gut microbiota derived metabolites, on T cell culture and expansion for immunotherapies. There is a complex gut microbiota-immune system interaction that can affect antitumor immunotherapy efficacy. Indeed, gut microbiota derived metabolites can modulate different biological functions in the immune system local and systemically.


Assuntos
Imunoterapia Adotiva , Neoplasias , Humanos , Imunoterapia , Linfócitos T , Neoplasias/terapia , Ácidos Graxos Voláteis
4.
Mol Ther ; 31(3): 686-700, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36641624

RESUMO

Several studies have shown the influence of commensal microbes on T cell function, specifically in the setting of checkpoint immunotherapy for cancer. In this study, we investigated how vancomycin-induced gut microbiota dysbiosis affects chimeric antigen receptor (CAR) T immunotherapy using multiple preclinical models as well as clinical correlates. In two murine tumor models, hematopoietic CD19+-A20 lymphoma and CD19+-B16 melanoma, mice receiving vancomycin in combination with CD19-directed CAR T cell (CART-19) therapy displayed increased tumor control and tumor-associated antigens (TAAs) cross-presentation compared with CART-19 alone. Fecal microbiota transplant from human healthy donors to pre-conditioned mice recapitulated the results obtained in naive gut microbiota mice. Last, B cell acute lymphoblastic leukemia patients treated with CART-19 and exposed to oral vancomycin showed higher CART-19 peak expansion compared with unexposed patients. These results substantiate the role of the gut microbiota on CAR T cell therapy and suggest that modulation of the gut microbiota using vancomycin may improve outcomes after CAR T cell therapy across tumor types.


Assuntos
Microbioma Gastrointestinal , Receptores de Antígenos Quiméricos , Humanos , Camundongos , Animais , Receptores de Antígenos de Linfócitos T/genética , Apresentação Cruzada , Vancomicina/farmacologia , Imunoterapia , Linfócitos T , Imunoterapia Adotiva/métodos , Receptores de Antígenos Quiméricos/genética , Antígenos CD19
5.
Cancers (Basel) ; 13(18)2021 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-34572890

RESUMO

Allogenic hematopoietic stem cell transplantation (allo-HSCT) is one of the standard treatments for B-cell lymphoproliferative disorders; however, deep relapses are common after an allo-HSCT, and it is associated with poor prognosis. A successful approach to overcome these relapses is to exploit the body's own immune system with chimeric antigen receptor (CAR) T-cells. These two approaches are potentially combinatorial for treating R/R B-cell lymphoproliferative disorders. Several clinical trials have described different scenarios in which allo-HSCT and CAR-T are successively combined. Further, for all transplanted patients, assessment of chimerism is important to evaluate the engraftment success. Nonetheless, for those patients who previously received an allo-HSCT there is no monitorization of chimerism before manufacturing CAR T-cells. In this review, we focus on allo-HSCT and CAR-T treatments and the different sources of T-cells for manufacturing CAR T-cells.

6.
J Immunother Cancer ; 9(2)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33563772

RESUMO

BACKGROUND: Tumor endothelial marker 1 (TEM1) is a protein expressed in the tumor-associated endothelium and/or stroma of various types of cancer. We previously demonstrated that immunization with a plasmid-DNA vaccine targeting TEM1 reduced tumor progression in three murine cancer models. Radiation therapy (RT) is an established cancer modality used in more than 50% of patients with solid tumors. RT can induce tumor-associated vasculature injury, triggering immunogenic cell death and inhibition of the irradiated tumor and distant non-irradiated tumor growth (abscopal effect). Combination treatment of RT with TEM1 immunotherapy may complement and augment established immune checkpoint blockade. METHODS: Mice bearing bilateral subcutaneous CT26 colorectal or TC1 lung tumors were treated with a novel heterologous TEM1-based vaccine, in combination with RT, and anti-programmed death-ligand 1 (PD-L1) antibody or combinations of these therapies, tumor growth of irradiated and abscopal tumors was subsequently assessed. Analysis of tumor blood perfusion was evaluated by CD31 staining and Doppler ultrasound imaging. Immunophenotyping of peripheral and tumor-infiltrating immune cells as well as functional analysis was analyzed by flow cytometry, ELISpot assay and adoptive cell transfer (ACT) experiments. RESULTS: We demonstrate that addition of RT to heterologous TEM1 vaccination reduces progression of CT26 and TC1 irradiated and abscopal distant tumors as compared with either single treatment. Mechanistically, RT increased major histocompatibility complex class I molecule (MHCI) expression on endothelial cells and improved immune recognition of the endothelium by anti-TEM1 T cells with subsequent severe vascular damage as measured by reduced microvascular density and tumor blood perfusion. Heterologous TEM1 vaccine and RT combination therapy boosted tumor-associated antigen (TAA) cross-priming (ie, anti-gp70) and augmented programmed cell death protein 1 (PD-1)/PD-L1 signaling within CT26 tumor. Blocking the PD-1/PD-L1 axis in combination with dual therapy further increased the antitumor effect and gp70-specific immune responses. ACT experiments show that anti-gp70 T cells are required for the antitumor effects of the combination therapy. CONCLUSION: Our findings describe novel cooperative mechanisms between heterologous TEM1 vaccination and RT, highlighting the pivotal role that TAA cross-priming plays for an effective antitumor strategy. Furthermore, we provide rationale for using heterologous TEM1 vaccination and RT as an add-on to immune checkpoint blockade as triple combination therapy into early-phase clinical trials.


Assuntos
Antígenos CD/metabolismo , Neoplasias Colorretais/terapia , Inibidores de Checkpoint Imunológico/administração & dosagem , Neoplasias Pulmonares/terapia , Proteínas de Neoplasias/metabolismo , Vacinas de DNA/administração & dosagem , Adenoviridae/genética , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral , Neoplasias Colorretais/diagnóstico por imagem , Terapia Combinada , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias Pulmonares/diagnóstico por imagem , Camundongos , Hipofracionamento da Dose de Radiação , Resultado do Tratamento , Ultrassonografia Doppler , Vacinas de DNA/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Int J Mol Sci ; 22(3)2021 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-33498529

RESUMO

Hematological malignancies, including multiple myeloma, lymphoma, and leukemia, are a heterogeneous group of neoplasms that affect the blood, bone marrow, and lymph nodes. They originate from uncontrolled growth of hematopoietic and lymphoid cells from different stages in their maturation/differentiation and account for 6.5% of all cancers around the world. During the last decade, it has been proven that the gut microbiota, more specifically the gastrointestinal commensal bacteria, is implicated in the genesis and progression of many diseases. The immune-modulating effects of the human microbiota extend well beyond the gut, mostly through the small molecules they produce. This review aims to summarize the current knowledge of the role of the microbiota in modulating the immune system, its role in hematological malignancies, and its influence on different therapies for these diseases, including autologous and allogeneic stem cell transplantation, chemotherapy, and chimeric antigen receptor T cells.


Assuntos
Microbioma Gastrointestinal , Neoplasias Hematológicas/microbiologia , Animais , Antibacterianos/efeitos adversos , Antibacterianos/uso terapêutico , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/terapia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunoterapia Adotiva , Transplante de Células-Tronco
8.
JCI Insight ; 5(16)2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32814714

RESUMO

Granulosa cell tumors (GCT) are rare ovarian malignancies. Due to the lack of effective treatment in late relapse, there is a clear unmet need for novel therapies. Forkhead Box L2 (FOXL2) is a protein mainly expressed in granulosa cells (GC) and therefore is a rational therapeutic target. Since we identified tumor infiltrating lymphocytes (TILs) as the main immune population within GCT, TILs from 11 GCT patients were expanded, and their phenotypes were interrogated to determine that T cells acquired late antigen-experienced phenotypes and lower levels of PD1 expression. Importantly, TILs maintained their functionality after ex vivo expansion as they vigorously reacted against autologous tumors (100% of patients) and against FOXL2 peptides (57.1% of patients). To validate the relevance of FOXL2 as a target for immune therapy, we developed a plasmid DNA vaccine (FoxL2-tetanus toxin; FoxL2-TT) by fusing Foxl2 cDNA with the immune-enhancing domain of TT. Mice immunization with FoxL2-TT controlled growth of FOXL2-expressing ovarian (BR5) and breast (4T1) cancers in a T cell-mediated manner. Combination of anti-PD-L1 with FoxL2-TT vaccination further reduced tumor progression and improved mouse survival without affecting the female reproductive system and pregnancy. Together, our results suggest that FOXL2 immune targeting can produce substantial long-term clinical benefits. Our study can serve as a foundation for trials testing immunotherapeutic approaches in patients with ovarian GCT.


Assuntos
Vacinas Anticâncer/farmacologia , Proteína Forkhead Box L2/imunologia , Tumor de Células da Granulosa/imunologia , Linfócitos do Interstício Tumoral/patologia , Adulto , Animais , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Epitopos , Feminino , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Tumor de Células da Granulosa/patologia , Tumor de Células da Granulosa/terapia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Tolerância Imunológica/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Camundongos Endogâmicos , Pessoa de Meia-Idade , Gravidez , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Microambiente Tumoral , Vacinas de DNA/imunologia , Vacinas de DNA/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Xenotransplantation ; 27(6): e12635, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32783288

RESUMO

Tumor necrosis factor α (TNFα) and its receptors contribute to rejection of transplanted cells and organs. To elucidate how TNFα affects xenograft rejection, we previously cloned the cDNA of pig TNF-receptor 2 (pTNFR2) and found four isoforms: one comprising the full receptor with four cysteine-rich domains (CRD), a shorter variant (pTNFR2ΔE7-10) encoding for a soluble isoform, another lacking exon 4 (pTNFR2ΔE4) displaying only 3 CRD and poor ligand binding, and the smallest one generated by the two alternative splicings. All isoforms contained the pre-ligand assembly domain (PLAD) responsible for receptor trimerization. We now investigated their roles by structural, expression, and subcellular localization studies. Structural in silico analyses identified four amino acids potentially involved in TNFα binding and lacking in pTNFR2ΔE4. Quantitative RT-PCR determined regulated expression affecting the two pTNFR2 alternative splicings in cytokine-stimulated porcine aortic endothelial cells (PAEC). Particularly, human IL-1α and TNFα produced a strong mRNA upregulation of all isoforms, being the full receptor the predominant one. However, expression of pTNFR2 on PAEC did not correlate with mRNA and decreased after 24-hour exposure to IL-1α or TNFα. Notably, confocal microscopy confirmed the presence of pTNFR2 inside and on the plasma membrane, whereas pTNFR2ΔE4 located only intracellularly. Most interestingly, FRET analyses showed that membrane-bound isoforms pTNFR2 and pTNFR2ΔE4 colocalized intracellularly and associated through the PLAD. Our data show that pTNFR2ΔE4 bind and may retain the full receptor intracellularly. This mechanism has not been described in other species and represents a particularity that may affect the pathophysiology of pig xenografts.


Assuntos
Células Endoteliais , Receptores Tipo II do Fator de Necrose Tumoral , Transplante Heterólogo , Processamento Alternativo , Animais , Rejeição de Enxerto , Isoformas de Proteínas/genética , Receptores Tipo II do Fator de Necrose Tumoral/genética , Suínos , Fator de Necrose Tumoral alfa
10.
Methods Mol Biol ; 2110: 27-45, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32002899

RESUMO

Understanding the molecular bases of xenograft rejection is one of the highest priorities in the xenotransplantation field. This information is needed for the successful development of genetic modifications of the animal source of xenogeneic cells and organs that prevent rejection. Furthermore, the identification of physiological incompatibilities in the xenogeneic setting is also necessary for developing the appropriate strategies that allow long-term xenograft function. As the pig is the species of choice for the development of the majority of xenogeneic applications, the cloning of pig genes or cDNA is a key step to elucidate the interactions of pig and human molecules. In addition, there are currently multiple bioinformatic tools which facilitate the study in silico of protein structures, molecular interactions, and docking sites. Thus, we describe a basic cloning method that comprises total RNA extraction, reverse transcription (RT), and polymerase chain reaction (PCR) for cDNA amplification and include some links for databases and bioinformatics tools available on the Internet for the subsequent analyses and predictions. Finally, some procedures of protein expression and analysis of protein interactions by surface plasmon resonance (SPR) are described for elucidating the molecular mechanisms of xenograft function and rejection.


Assuntos
Técnicas de Diagnóstico Molecular , Pesquisa , Transplante Heterólogo/métodos , Animais , Clonagem Molecular , Biologia Computacional , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/metabolismo , Humanos , Reação em Cadeia da Polimerase , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Suínos
11.
J Clin Invest ; 130(1): 466-479, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31815742

RESUMO

Alterations in gut microbiota impact the pathophysiology of several diseases, including cancer. Radiotherapy (RT), an established curative and palliative cancer treatment, exerts potent immune modulatory effects, inducing tumor-associated antigen (TAA) cross-priming with antitumor CD8+ T cell elicitation and abscopal effects. We tested whether the gut microbiota modulates antitumor immune response following RT distal to the gut. Vancomycin, an antibiotic that acts mainly on gram-positive bacteria and is restricted to the gut, potentiated the RT-induced antitumor immune response and tumor growth inhibition. This synergy was dependent on TAA cross presentation to cytolytic CD8+ T cells and on IFN-γ. Notably, butyrate, a metabolite produced by the vancomycin-depleted gut bacteria, abrogated the vancomycin effect. In conclusion, depletion of vancomycin-sensitive bacteria enhances the antitumor activity of RT, which has important clinical ramifications.


Assuntos
Apresentação de Antígeno/efeitos da radiação , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Microbioma Gastrointestinal , Neoplasias Experimentais , Animais , Apresentação de Antígeno/genética , Antígenos de Neoplasias/genética , Butiratos/imunologia , Linfócitos T CD8-Positivos/patologia , Células Dendríticas/patologia , Feminino , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/efeitos da radiação , Camundongos , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/radioterapia
12.
JCI Insight ; 3(4)2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29467322

RESUMO

Adoptive T cell therapy (ACT) is a promising new modality for malignancies. Here, we report that adoptive T cell efficacy in tumor-bearing mice is significantly affected by differences in the native composition of the gut microbiome or treatment with antibiotics, or by heterologous fecal transfer. Depletion of bacteria with vancomycin decreased the rate of tumor growth in mice from The Jackson Laboratory receiving ACT, whereas treatment with neomycin and metronidazole had no effect, indicating the role of specific bacteria in host response. Vancomycin treatment induced an increase in systemic CD8α+ DCs, which sustained systemic adoptively transferred antitumor T cells in an IL-12-dependent manner. In subjects undergoing allogeneic hematopoietic cell transplantation, we found that oral vancomycin also increased IL-12 levels. Collectively, our findings demonstrate an important role played by the gut microbiota in the antitumor effectiveness of ACT and suggest potentially new avenues to improve response to ACT by altering the gut microbiota.


Assuntos
Células Dendríticas/imunologia , Microbioma Gastrointestinal/imunologia , Transplante de Células-Tronco Hematopoéticas , Interações entre Hospedeiro e Microrganismos/imunologia , Imunoterapia Adotiva/métodos , Interleucina-12/imunologia , Neoplasias/terapia , Adulto , Idoso , Animais , Bactérias/efeitos dos fármacos , Bactérias/genética , Bactérias/imunologia , Bactérias/isolamento & purificação , Antígenos CD8/imunologia , Antígenos CD8/metabolismo , Linhagem Celular Tumoral/transplante , Estudos de Coortes , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Transplante de Microbiota Fecal , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Humanos , Interleucina-12/antagonistas & inibidores , Interleucina-12/genética , Interleucina-12/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Neomicina/administração & dosagem , Neoplasias/imunologia , Neoplasias/microbiologia , Linfócitos T/imunologia , Linfócitos T/transplante , Resultado do Tratamento , Vancomicina/administração & dosagem
13.
JCI Insight ; 3(1)2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29321369

RESUMO

Successful tumor eradication by chimeric antigen receptor-expressing (CAR-expressing) T lymphocytes depends on CAR T cell persistence and effector function. We hypothesized that CD4+ and CD8+ T cells may exhibit distinct persistence and effector phenotypes, depending on the identity of specific intracellular signaling domains (ICDs) used to generate the CAR. First, we demonstrate that the ICOS ICD dramatically enhanced the in vivo persistence of CAR-expressing CD4+ T cells that, in turn, increased the persistence of CD8+ T cells expressing either CD28- or 4-1BB-based CARs. These data indicate that persistence of CD8+ T cells was highly dependent on a helper effect provided by the ICD used to redirect CD4+ T cells. Second, we discovered that combining ICOS and 4-1BB ICDs in a third-generation CAR displayed superior antitumor effects and increased persistence in vivo. Interestingly, we found that the membrane-proximal ICD displayed a dominant effect over the distal domain in third-generation CARs. The optimal antitumor and persistence benefits observed in third-generation ICOSBBz CAR T cells required the ICOS ICD to be positioned proximal to the cell membrane and linked to the ICOS transmembrane domain. Thus, CARs with ICOS and 4-1BB ICD demonstrate increased efficacy in solid tumor models over our current 4-1BB-based CAR and are promising therapeutics for clinical testing.


Assuntos
Ligante 4-1BB/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Adenocarcinoma , Animais , Antineoplásicos/farmacologia , Antígenos CD28/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Membrana Celular , Humanos , Neoplasias Pulmonares , Camundongos , Metástase Neoplásica , Neoplasias Pancreáticas , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
14.
Oncoimmunology ; 6(12): e1398878, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29209575

RESUMO

DNA-based vaccination is a promising approach to cancer immunotherapy. DNA-based vaccines specific for tumor-associated antigens (TAAs) are indeed relatively simple to produce, cost-efficient and well tolerated. However, the clinical efficacy of DNA-based vaccines for cancer therapy is considerably limited by central and peripheral tolerance. During the past decade, considerable efforts have been devoted to the development and characterization of novel DNA-based vaccines that would circumvent this obstacle. In this setting, particular attention has been dedicated to the route of administration, expression of modified TAAs, co-expression of immunostimulatory molecules, and co-delivery of immune checkpoint blockers. Here, we review preclinical and clinical progress on DNA-based vaccines for cancer therapy.

15.
J Immunol ; 195(8): 4020-7, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26378078

RESUMO

Mitochondria provide energy for cells via oxidative phosphorylation. Reactive oxygen species, a byproduct of this mitochondrial respiration, can damage mitochondrial DNA (mtDNA), and somatic mtDNA mutations have been found in all colorectal, ovarian, breast, urinary bladder, kidney, lung, and pancreatic tumors studied. The resulting altered mitochondrial proteins or tumor-associated mitochondrial Ags (TAMAs) are potentially immunogenic, suggesting that they may be targetable Ags for cancer immunotherapy. In this article, we show that the RENCA tumor cell line harbors TAMAs that can drive an antitumor immune response. We generated a cellular tumor vaccine by pulsing dendritic cells with enriched mitochondrial proteins from RENCA cells. Our dendritic cell-based RENCA mitochondrial lysate vaccine elicited a cytotoxic T cell response in vivo and conferred durable protection against challenge with RENCA cells when used in a prophylactic or therapeutic setting. By sequencing mtDNA from RENCA cells, we identified two mutated molecules: COX1 and ND5. Peptide vaccines generated from mitochondrial-encoded COX1 but not from ND5 had therapeutic properties similar to RENCA mitochondrial protein preparation. Thus, TAMAs can elicit effective antitumor immune responses, potentially providing a new immunotherapeutic strategy to treat cancer.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Carcinoma de Células Renais/prevenção & controle , Ciclo-Oxigenase 1/imunologia , Neoplasias Renais/prevenção & controle , Proteínas de Membrana/imunologia , Proteínas Mitocondriais/imunologia , NADH Desidrogenase/imunologia , Neoplasias Experimentais/prevenção & controle , Animais , Antígenos de Neoplasias/farmacologia , Vacinas Anticâncer/farmacologia , Carcinoma de Células Renais/imunologia , Ciclo-Oxigenase 1/farmacologia , Neoplasias Renais/imunologia , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Mitocondriais/farmacologia , NADH Desidrogenase/farmacologia , Neoplasias Experimentais/imunologia
16.
J Invest Dermatol ; 133(6): 1541-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23407395

RESUMO

Munro's microabscesses contain polymorphonuclear leukocytes and form specifically in the epidermis of psoriasis patients. The mechanism whereby the neutrophils are recruited into the epidermis is poorly understood. Using a combination of human and mouse primary keratinocyte cell cultures and the imiquimod-induced psoriasis-like mouse model of skin inflammation, we explored the role of IL-1 signaling in microabscess formation. In vitro imiquimod stimulated production of IL-1α and neutrophil recruiting chemokines. Imiquimod-activated chemokine expression was dependent upon adenosine signaling and independent of IL-1α and IL-1 receptor type 1 (IL-1R1); nevertheless, IL-1α could enhance chemokine expression initiated by imiquimod. Topical application of imiquimod in vivo led to epidermal microabscess formation, acanthosis, and increased IL-1α and chemokine expression in the skin of wild-type mice. However, in IL-1R1-deficient mice these responses were either absent or dramatically reduced. These results demonstrate that IL-1α and IL-1R1 signaling is essential for microabscess formation, neutrophil recruiting chemokine expression, and acanthosis in psoriasis-like skin inflammation induced by imiquimod.


Assuntos
Abscesso/induzido quimicamente , Aminoquinolinas/farmacologia , Toxidermias/imunologia , Queratinócitos/imunologia , Receptores Tipo I de Interleucina-1/imunologia , Receptores Tipo I de Interleucina-1/metabolismo , Abscesso/imunologia , Abscesso/patologia , Adjuvantes Imunológicos/farmacologia , Animais , Animais Recém-Nascidos , Quimiocina CXCL1/imunologia , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/imunologia , Quimiocina CXCL2/metabolismo , Derme/citologia , Toxidermias/patologia , Células Epidérmicas , Humanos , Imiquimode , Interleucina-1alfa/imunologia , Interleucina-1alfa/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/citologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Cultura Primária de Células , Psoríase/induzido quimicamente , Psoríase/imunologia , Psoríase/patologia , Receptores Tipo I de Interleucina-1/genética , Transdução de Sinais/imunologia
17.
Methods Mol Biol ; 885: 17-31, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22565987

RESUMO

Understanding the molecular bases of xenograft rejection is one of the highest priorities in the xenotransplantation field. Furthermore, the identification of physiological incompatibilities in the xenogeneic setting is also necessary for developing the appropriate strategies to have a long-term functioning xenograft. As the pig is the species of choice for the development of xenogeneic applications, the cloning of pig genes or cDNA is a key step to elucidate the interactions of pig and human molecules. It also provides the necessary information for assessing the level of mRNA expression of relevant proteins in tissues and organs of interest for xenotransplantation. In most cases, the cloning of the cDNA is sufficient to attain these goals. Thus, we describe a basic cloning method that comprises total RNA extraction, reverse transcription (RT), and polymerase chain reaction (PCR) amplification. We also include some links for databases and bioinformatic tools available in the Internet for the subsequent analyses and predictions. Finally, we recommend and explain the procedures of northern blotting and quantitative RT-PCR for conducting the mRNA expression studies.


Assuntos
Clonagem Molecular , Expressão Gênica , Suínos/genética , Animais , Biologia Computacional/métodos , DNA Complementar , Humanos , RNA Mensageiro/metabolismo , Transcrição Gênica
18.
Methods Mol Biol ; 885: 33-45, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22565988

RESUMO

Proteins are the focus of numerous xenotransplantation studies because they provide structure and function to the graft. Their presence, absence, or even a functional incompatibility among species can compromise the long-term functioning of the xenograft. In particular, many cell-surface and soluble proteins, such as cytokines and chemokines, are involved in triggering rejection. For this reason, the identification and characterization of key proteins for xenografting, of either pig or human origin, are very important. Understanding their role in the xenogeneic setting can set the bases for the development of genetic engineering approaches that prolong graft survival and ensure function. There are multiple ways of determining and attaining protein expression, as well as studying protein interactions. In this chapter, we describe some basic techniques that allow us to detect and characterize pig and human proteins in order to better understand the molecular bases of rejection and function of pig xenografts.


Assuntos
Proteínas/metabolismo , Proteômica , Transplante Heterólogo , Animais , Anticorpos/imunologia , Anticorpos/metabolismo , Formação de Anticorpos , Citometria de Fluxo , Humanos , Ligação Proteica , Proteínas/genética , Proteínas/imunologia , Proteínas/isolamento & purificação , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Ressonância de Plasmônio de Superfície
19.
Xenotransplantation ; 18(2): 131-46, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21496120

RESUMO

BACKGROUND: TNF and its receptors TNF-Receptor 1 (TNFR1, CD120a) and TNF-Receptor 2 (TNFR2, CD120b) have been implicated in the rejection of transplanted cells and organs. Although pig TNFR1 (pTNFR1) is known to mediate the effects of human TNF in a xenogeneic setting, it is unclear whether pig TNFR2 (pTNFR2) could contribute to xenograft rejection. METHODS: We have cloned the cDNA of various pTNFR2 variants by reverse transcription-polymerase chain reaction (RT-PCR) and rapid amplification of cDNA ends. We have characterized the predicted proteins with bioinformatic tools and conducted expression, affinity, and functional studies to investigate their roles. RESULTS: We have identified four isoforms of pTNFR2: one comprising the four cysteine-rich domains (CRD) conserved between species, a shorter variant (pTNFR2ΔE7-10) encoding for a soluble isoform, another with only three CRD due to the lack of exon 4 (pTNFR2ΔE4), and a fourth variant containing both modifications. Accordingly, multiple mRNA transcripts were observed by northern blotting. Quantitative RT-PCR determined high pTNFR2 expression in lung and immune cells and detected the two alternative splicings in all cells/tissues examined. The full receptor was moderately expressed on the surface of pig cells such as porcine aortic endothelial cells and PK-15 and was regulated by TNF. On the contrary, the membrane-bound pTNFR2ΔE4 was located only intracellularly. Plasmon resonance studies showed that pTNFR2 binds pig and human TNFα with high affinity, but pTNFR2ΔE4 interacts poorly with pig TNFα and does not bind to the human cytokine. Moreover, pull-down experiments with the two recombinant soluble isoforms consistently demonstrated that the two bound together and soluble pTNFR2ΔE4 was able to modulate the TNF inhibitory activity of pTNFR2-GST in a cell-based assay. CONCLUSION: The pTNFR2 may participate in the process of xenograft rejection and other related events, as well as be used in soluble form to block TNF in this setting. In addition, we have discovered other pTNFR2 isoforms that may affect the pig immune responses and have an impact on rejection of xenografts.


Assuntos
Membrana Celular/metabolismo , Rejeição de Enxerto/fisiopatologia , Receptores Tipo II do Fator de Necrose Tumoral/análise , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Transplante Heterólogo/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Rim/citologia , Rim/metabolismo , Camundongos , Dados de Sequência Molecular , Isoformas de Proteínas , RNA Mensageiro/genética , Receptores Tipo II do Fator de Necrose Tumoral/genética , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA