Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.000
Filtrar
1.
Front Immunol ; 14: 1212641, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37388741

RESUMO

Tolerogenic dendritic cells (tDC) arrest the progression of autoimmune-driven dysglycemia into clinical, insulin-requiring type 1 diabetes (T1D) and preserve a critical mass of ß cells able to restore some degree of normoglycemia in new-onset clinical disease. The safety of tDC, generated ex vivo from peripheral blood leukocytes, has been demonstrated in phase I clinical studies. Accumulating evidence shows that tDC act via multiple layers of immune regulation arresting the action of pancreatic ß cell-targeting effector lymphocytes. tDC share a number of phenotypes and mechanisms of action, independent of the method by which they are generated ex vivo. In the context of safety, this yields confidence that the time has come to test the best characterized tDC in phase II clinical trials in T1D, especially given that tDC are already being tested for other autoimmune conditions. The time is also now to refine purity markers and to "universalize" the methods by which tDC are generated. This review summarizes the current state of tDC therapy for T1D, presents points of intersection of the mechanisms of action that the different embodiments use to induce tolerance, and offers insights into outstanding matters to address as phase II studies are imminent. Finally, we present a proposal for co-administration and serially-alternating administration of tDC and T-regulatory cells (Tregs) as a synergistic and complementary approach to prevent and treat T1D.


Assuntos
Doenças Autoimunes , Diabetes Mellitus Tipo 1 , Humanos , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Terapia Baseada em Transplante de Células e Tecidos , Células Dendríticas/transplante , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Tolerância Imunológica
2.
J Immunol ; 208(5): 1066-1075, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35140132

RESUMO

BATF3-deficient mice that lack CD8+ dendritic cells (DCs) showed an exacerbation of chronic graft-versus-host disease (cGVHD), including T follicular helper (Tfh) cell and autoantibody responses, whereas mice carrying the Sle2c2 lupus-suppressive locus with a mutation in the G-CSFR showed an expansion of CD8+ DCs and a poor mobilization of plasmacytoid DCs (pDCs) and responded poorly to cGVHD induction. Here, we investigated the contribution of CD8+ DCs and pDCs to the humoral response to protein immunization, where CD8neg DCs are thought to represent the major inducers. Both BATF3-/- and Sle2c2 mice had reduced humoral and germinal center (GC) responses compared with C57BL/6 (B6) controls. We showed that B6-derived CD4+ DCs are the major early producers of IL-6, followed by CD4-CD8- DCs. Surprisingly, IL-6 production and CD80 expression also increased in CD8+ DCs after immunization, and B6-derived CD8+ DCs rescued Ag-specific adaptive responses in BATF3-/- mice. In addition, inflammatory pDCs (ipDCs) produced more IL-6 than all conventional DCs combined. Interestingly, G-CSFR is highly expressed on pDCs. G-CSF expanded pDC and CD8+ DC numbers and IL-6 production by ipDCs and CD4+ DCs, and it improved the quality of Ab response, increasing the localization of Ag-specific T cells to the GC. Finally, G-CSF activated STAT3 in early G-CSFR+ common lymphoid progenitors of cDCs/pDCs but not in mature cells. In conclusion, we showed a multilayered role of DC subsets in priming Tfh cells in protein immunization, and we unveiled the importance of G-CSFR signaling in the development and function pDCs.


Assuntos
Células Dendríticas/imunologia , Doença Enxerto-Hospedeiro/imunologia , Células Progenitoras Linfoides/citologia , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Células T Auxiliares Foliculares/imunologia , Transferência Adotiva , Animais , Autoanticorpos/imunologia , Antígeno B7-1/biossíntese , Fatores de Transcrição de Zíper de Leucina Básica/genética , Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Diferenciação Celular/imunologia , Células Dendríticas/transplante , Feminino , Fator Estimulador de Colônias de Granulócitos/metabolismo , Interleucina-6/biossíntese , Ativação Linfocitária/imunologia , Células Progenitoras Linfoides/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Proteínas Repressoras/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/imunologia
3.
Pharm Res ; 39(2): 353-367, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35166995

RESUMO

PURPOSE: The invention and application of new immunotherapeutic methods can compensate for the inefficiency of conventional cancer treatment approaches, partly due to the inhibitory microenvironment of the tumor. In this study, we tried to inhibit the growth of cancer cells and induce anti-tumor immune responses by silencing the expression of the ß-catenin in the tumor microenvironment and transmitting interleukin (IL)-15 cytokine to provide optimal conditions for the dendritic cell (DC) vaccine. METHODS: For this purpose, we used folic acid (FA)-conjugated SPION-carboxymethyl dextran (CMD) chitosan (C) nanoparticles (NPs) to deliver anti-ß-catenin siRNA and IL-15 to cancer cells. RESULTS: The results showed that the codelivery of ß-catenin siRNA and IL-15 significantly reduced the growth of cancer cells and increased the immune response. The treatment also considerably stimulated the performance of the DC vaccine in triggering anti-tumor immunity, which inhibited tumor development and increased survival in mice in two different cancer models. CONCLUSIONS: These findings suggest that the use of new nanocarriers such as SPION-C-CMD-FA could be an effective way to use as a novel combination therapy consisting of ß-catenin siRNA, IL-15, and DC vaccine to treat cancer.


Assuntos
Antineoplásicos/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Células Dendríticas/transplante , Portadores de Fármacos , Interleucina-15/administração & dosagem , Nanopartículas Magnéticas de Óxido de Ferro , Melanoma Experimental/terapia , RNA Interferente Pequeno/administração & dosagem , Terapêutica com RNAi , Neoplasias Cutâneas/terapia , beta Catenina/genética , Animais , Antineoplásicos/química , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/imunologia , Composição de Medicamentos , Feminino , Regulação Neoplásica da Expressão Gênica , Interleucina-15/química , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos BALB C , RNA Interferente Pequeno/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral
4.
J Immunol Res ; 2022: 1178874, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35155685

RESUMO

Dendritic cell- (DC-) based vaccination has emerged as a promising antitumour immunotherapy. However, overcoming immune tolerance and immunosuppression in the tumour microenvironment (TME) is still a great challenge. Recent studies have shown that Rose Bengal (RB) can effectively induce immunogenic cell death (ICD) in cancer cells, presenting whole tumour antigens for DC processing and presentation. However, the synergistic antitumour effect of combining intralesional RB with immature DCs (RB-iDCs) remains unclear. In the present study, we investigated whether RB-iDCs have superior antitumour effects compared with either single agent and evaluated the immunological mechanism of RB-iDCs in a murine lung cancer model. The results showed that intralesional RB-iDCs suppressed subcutaneous tumour growth and lung metastasis, which resulted in 100% mouse survival and significantly increased TNF-α production by CD8+ T cells. These effects were closely related to the induction of the expression of distinct ICD hallmarks by RB in both bulk cancer cells and cancer stem cells (CSCs), especially calreticulin (CRT), thus enhancing immune effector cell (i.e., CD4+, CD8+, and memory T cells) infiltration and attenuating the accumulation of immunosuppressive cells (i.e., Tregs, macrophages, and myeloid-derived suppressor cells (MDSCs)) in the TME. This study reveals that the RB-iDC vaccine can synergistically destroy the primary tumour, inhibit distant metastasis, and prevent tumour relapse in a lung cancer mouse model, which provides important preclinical data for the development of a novel combinatorial immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Imunoterapia/métodos , Neoplasias Pulmonares/imunologia , Linfócitos do Interstício Tumoral/imunologia , Melanoma/imunologia , Imunidade Adaptativa , Animais , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Diferenciação Celular , Células Dendríticas/transplante , Humanos , Imunização , Neoplasias Pulmonares/secundário , Melanoma/patologia , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Rosa Bengala/metabolismo
5.
Scand J Immunol ; 95(2): e13131, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34936112

RESUMO

The role of the immune system against coronavirus disease 2019 (COVID-19) is unknown in many aspects, and the protective or pathologic mechanisms of the immune response are poorly understood. Pro-inflammatory cytokine release and a consequent cytokine storm can lead to acute respiratory distress syndrome (ARDS) and result in multi-organ failure. There are many T cell subsets during anti-viral immunity. The Th17-associated response, as a pro-inflammatory pathway, and its consequent outcomes in many autoimmune disorders play a fundamental role in progression of systemic hyper-inflammation during COVID-19. Therapeutic strategies based on immunomodulation therapy could be helpful for targeting hyper-inflammatory immune responses in COVID-19, especially Th17-related inflammation and hyper-cytokinemia. Cell-based immunotherapeutic approaches including mesenchymal stem cells (MSCs), tolerogenic dendritic cells (tolDCs) and regulatory T cells (Tregs) seem to be promising strategies as orchestrators of the immune response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this review, we highlight Th17-related immunopathology of SARS-CoV-2 infection and discuss cell-based immunomodulatory strategies and their mechanisms for regulation of the hyper-inflammation during COVID-19.


Assuntos
COVID-19/patologia , COVID-19/terapia , Síndrome da Liberação de Citocina/patologia , Imunomodulação/imunologia , Células Th17/imunologia , Transferência Adotiva/métodos , COVID-19/imunologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Citocinas/sangue , Células Dendríticas/transplante , Humanos , Transplante de Células-Tronco Mesenquimais , SARS-CoV-2/imunologia , Linfócitos T Reguladores/transplante
6.
Life Sci ; 288: 120166, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-34813798

RESUMO

Following various immunotherapies, lack of proper anti-tumor immune responses is considered a significant problem in novel cancer therapeutic approaches. The expression of inhibitory checkpoint molecules on tumor-infiltrating T cells is one of the main reasons for the ineffectiveness of various immunotherapies. Therefore, we decided to inhibit two of the most important immune checkpoints expressed on tumor-associated T cells, PD-1 and A2aR. Ligation of PD-1 with PD-L1 and A2aR with adenosine significantly suppress T cell responses against tumor cells. Whitin tumors, specific inhibition of these molecules on T cells is of particular importance for successful immunotherapy as well as the elimination of treatment-associated side-effects. Thus, in this study, superparamagnetic iron oxide (SPION) nanoparticles (NPs) were covered by chitosan lactate (CL), functionalized with TAT peptide, and loaded with siRNA molecules against PD-1 and A2aR. Appropriate physicochemical properties of the prepared NPs resulted in efficient delivery of siRNA to tumor-derived T cells and suppressed the expression of A2aR and PD-1, ex vivo. T cell functions such as cytokine secretion and proliferation were considerably enhanced by the downregulation of these molecules which led to an increase in their survival time. Interestingly, treatment of CT26 and 4T1 mouse tumors with siRNA-loaded NPs not only inhibited tumor growth but also markedly increased anti-tumor immune responses and survival time. The results strongly support the efficacy of SPION-CL-TAT NPs loaded with anti-PD-1/A2aR siRNAs in cancer therapy and their further development for cancer patients in the near future.


Assuntos
Neoplasias da Mama/terapia , Neoplasias Colorretais/terapia , Nanopartículas/administração & dosagem , Receptor de Morte Celular Programada 1/antagonistas & inibidores , RNA Interferente Pequeno/genética , Receptor A2A de Adenosina/química , Vacinas/administração & dosagem , Animais , Apoptose , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Proliferação de Células , Quitosana/química , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Terapia Combinada , Células Dendríticas/imunologia , Células Dendríticas/transplante , Feminino , Humanos , Imunoterapia , Ácido Láctico/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , Receptor de Morte Celular Programada 1/imunologia , Receptor A2A de Adenosina/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Leukoc Biol ; 110(6): 1023-1031, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34643294

RESUMO

The 78-kDa glucose-regulated protein (GRP78) has extracellular, anti-inflammatory properties that can aid resolving inflammation. It has been established previously that GRP78 induced myeloid CD11c+ cell differentiation into distinct tolerogenic cells. This tolerance induction makes GRP78 a potential therapeutic agent for transplanted allogeneic grafts and autoimmune diseases, such as type 1 diabetes. In this research, it is revealed that rmGRP78-treated NOD mice bone marrow-derived CD11c+ cells (GRP78-DCs) highly expressed B7-H4 but down-regulated CD86 and CD40, and retained a tolerogenic signature even after stimulation by LPS. In the assessment of in vivo therapeutic efficacy after the adoptive transfer of GRP78-DCs into NOD mice, fluorescent imaging analyses revealed that the transfer specifically homed in inflamed pancreases, promoting ß-cell survival and alleviating insulitis in NOD mice. The adoptive transfer of GRP78-DCs also helped reduce Th1, Th17, and CTL, suppressing inflammatory cytokine production in vivo. The findings suggest that adoptive GRP78-DC transfer is critical to resolving inflammation in NOD mice and may have relevance in a clinical setting.


Assuntos
Células Dendríticas/transplante , Diabetes Mellitus Tipo 1/imunologia , Chaperona BiP do Retículo Endoplasmático , Tolerância Imunológica/imunologia , Ilhotas Pancreáticas , Transferência Adotiva , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Chaperona BiP do Retículo Endoplasmático/imunologia , Chaperona BiP do Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático/farmacologia , Feminino , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Pancreatite/imunologia
8.
Front Immunol ; 12: 669965, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34489928

RESUMO

Introduction: Animal studies and preclinical studies in cancer patients suggest that the induction of immunogenic cell death (ICD) by neoadjuvant chemotherapy with doxorubicin and cyclophosphamide (NAC-AC) recovers the functional performance of the immune system. This could favor immunotherapy schemes such as the administration of antigen-free autologous dendritic cells (DCs) in combination with NAC-AC to profit as cryptic vaccine immunogenicity of treated tumors. Objective: To explore the safety and immunogenicity of autologous antigen-free DCs administered to breast cancer patients (BCPs) in combination with NAC-AC. Materials and Methods: A phase I/II cohort clinical trial was performed with 20 BCPs treated with NAC-AC [nine who received DCs and 11 who did not (control group)]. The occurrence of adverse effects and the functional performance of lymphocytes from BCPs before and after four cycles of NAC-AC receiving DCs or not were assessed using flow cytometry and compared with that from healthy donors (HDs). Flow cytometry analysis using manual and automated algorithms led us to examine functional performance and frequency of different lymphocyte compartments in response to a stimulus in vitro. This study was registered at clinicaltrials.gov (NCT03450044). Results: No grade II or higher adverse effects were observed associated with the transfer of DCs to patients during NAC-AC. Interestingly, in response to the in vitro stimulation, deficient phosphorylation of Zap70 and AKT proteins observed before chemotherapy in most patients' CD4 T cells significantly recovered after NAC-AC only in patients who received DCs. Conclusions: The transfer of autologous DCs in combination with NAC-AC in BCPs is a safe procedure. That, in BCPs, the administration of DCs in combination with NAC-AC favors the recovery of the functional capacity of T cells suggests that this combination may potentiate the adjuvant effect of ICD induced by NAC-AC on T cells and, hence, potentiate the immunogenicity of tumors as cryptic vaccines.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/terapia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/transplante , Imunoterapia Adotiva , Linfócitos do Interstício Tumoral/imunologia , Terapia Neoadjuvante , Linfócitos T/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Quimioterapia Adjuvante , Colômbia , Ciclofosfamida/uso terapêutico , Células Dendríticas/imunologia , Doxorrubicina/uso terapêutico , Feminino , Humanos , Imunoterapia Adotiva/efeitos adversos , Pessoa de Meia-Idade , Fatores de Tempo , Resultado do Tratamento , Microambiente Tumoral
9.
J Immunol Methods ; 498: 113133, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34480950

RESUMO

The study of the effect of cryopreservation on the functionality of monocyte-derived dendritic cells (MDDCs) and dendritic cells (DCs) is essential for their use in different clinical applications such as DCs-based vaccines. Its full maturation and its optimal functionality are crucial for DCs based immunotherapy. In this study, we compared MDDCs derived from fresh and cryopreserved PBMCs in the aspects of phenotype and its effect on T cells at the level of proliferation and cytokine secretion. We pulsed MDDCs obtained from fresh and cryopreserved PBMCs with two different stimuli, CEF and SEA, and the expression maturation markers and cytokine secretion were analyzed. Our results showed that the cryopreservation had no effects in the phenotype of the MDDCs obtained, cell viability, maturation markers expression and/or cytokines secretion, independently whether MDDCs had been generated from fresh or cryopreserved PBMCs. Thus, this study suggests that the use of cryopreserved cells is a good method to keep the cells before use in immunotherapy, avoiding the variability within same individual due to severe blood draws. Even so, the interpretation and comparison of different results should be done considering the different cryopreservation techniques and assays, and their effects on PBMCs, specifically on MDDC and DC cells.


Assuntos
Diferenciação Celular , Criopreservação , Células Dendríticas/imunologia , Monócitos/imunologia , Vacinas/imunologia , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/transplante , Estudos de Viabilidade , Citometria de Fluxo , Humanos , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Fenótipo , Linfócitos T/imunologia , Linfócitos T/metabolismo
10.
Cancer Lett ; 522: 184-197, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34562519

RESUMO

Despite the many successes and opportunities presented by PD-1 blockade in cancer therapies, anti-PD-1 monoclonal antibodies still face multiple challenges. Herein we report a strategy based on a nanobody (Nb) to circumvent these obstacles. A new PD-1-blocking Nb (PD-1 Nb20) in combination with tumor-specific dendritic cell (DC)/tumor-fusion cell (FC) vaccine that aims to improve the activation, proliferation, cytokine secretion, and tumor cell cytotoxicity of CD8+ T-cells. This combination was found to effectively enhance the in vitro cytotoxicity of CD8+ T-cells to kill human non-small cell lung cancer (NSCLC) HCC827 cells, hepatocellular carcinoma (HCC) HepG2 cells, and tongue squamous cell carcinoma (TSCC) Tca8113 cells. Moreover, CD8+ T-cells pre-treated with PD-1 Nb20 and tumor-specific DC/tumor-FCs significantly suppressed the growth of NSCLC-, HCC- and TSCC-derived xenograft tumors and prolonged the survival of tumor-bearing mice, through promoting T-cell infiltration to kill tumor cells and inhibiting tumor angiogenesis. These data demonstrate that PD-1 Nb20 in synergy with DC/tumor-FC vaccine augment the broad spectrum of antitumor activity of CD8+ T-cells, providing an alternative and promising immunotherapeutic strategy for tumor patients who are T-cell-dysfunctional or not sensitive to anti-PD-1 therapy.


Assuntos
Vacinas Anticâncer/farmacologia , Células Dendríticas/transplante , Receptor de Morte Celular Programada 1/imunologia , Anticorpos de Domínio Único/farmacologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/patologia , Células Hep G2 , Xenoenxertos , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Camundongos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Anticorpos de Domínio Único/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Neoplasias da Língua/tratamento farmacológico , Neoplasias da Língua/genética , Neoplasias da Língua/imunologia , Neoplasias da Língua/patologia
11.
Exp Oncol ; 43(3): 217-223, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34591429

RESUMO

BACKGROUND: One of the major factors restricting in vivo efficacy of dendritic cells (DCs) based immunotherapy is the inefficient migration of these cells to the lymphoid tissue, wherein DCs activate antigen-specific T cells. A fundamentally new approach for the possibility of enhancing the antitumor effects of DC-based immunotherapy may be the use of magnetically sensitive nanocomplexes to increase the target delivery of DCs to the lymph nodes of the recipient. AIM: To study the antitumor and immunomodulatory effects of the DC-nanovaccine with magnetosensitive properties and its influence on the immunosuppressive tumor microenvironment in mice with sarcoma 37. MATERIALS AND METHODS: The antitumor, antimetastatic and immunomodulatory effects of DCs loaded with magnetic nanocomplex under magnetic field (MF) control in mice with sarcoma 37 have been investigated. RESULTS: Combined therapy contributed to a significant reduction in tumor volume and weight compared to the control group of mice and mice that received the DC vaccine without MF. Therapy with magnetically sensitive DC nanovaccine with and without the addition of the MF was accompanied by a significant down-regulation of the level of FoxP3, transforming growth factor ß, interleukin (IL)-10 and vascular endothelial growth factors, mRNA expression in tumor tissues. A significant increase in interferon-γ and IL-4 mRNA expression was found in mice treated with the magnetically sensitive DC nanovaccine under MF control. CONCLUSION: A significant increase in the antitumor efficacy of the DC vaccine can be achieved using magnetosensitive nanocarriers of tumor antigens under MF control.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/transplante , Imunoterapia/métodos , Magnetismo/métodos , Nanopartículas/administração & dosagem , Neoplasias Experimentais/terapia , Animais , Apoptose , Proliferação de Células , Feminino , Linfonodos/imunologia , Linfonodos/patologia , Masculino , Camundongos , Camundongos Endogâmicos CBA , Camundongos Nus , Nanopartículas/química , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Células Tumorais Cultivadas
12.
Int J Mol Sci ; 22(15)2021 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-34360736

RESUMO

Myeloid regulatory cell-based therapy has been shown to be a promising cell-based medicinal approach in organ transplantation and for the treatment of autoimmune diseases, such as type 1 diabetes, rheumatoid arthritis, Crohn's disease and multiple sclerosis. Dendritic cells (DCs) are the most efficient antigen-presenting cells and can naturally acquire tolerogenic properties through a variety of differentiation signals and stimuli. Several subtypes of DCs have been generated using additional agents, including vitamin D3, rapamycin and dexamethasone, or immunosuppressive cytokines, such as interleukin-10 (IL-10) and transforming growth factor-beta (TGF-ß). These cells have been extensively studied in animals and humans to develop clinical-grade tolerogenic (tol)DCs. Regulatory macrophages (Mregs) are another type of protective myeloid cell that provide a tolerogenic environment, and have mainly been studied within the context of research on organ transplantation. This review aims to thoroughly describe the ex vivo generation of tolDCs and Mregs, their mechanism of action, as well as their therapeutic application and assessment in human clinical trials.


Assuntos
Artrite Reumatoide , Terapia Baseada em Transplante de Células e Tecidos , Células Dendríticas , Diabetes Mellitus Tipo 1 , Tolerância Imunológica , Macrófagos , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/terapia , Colecalciferol/farmacologia , Células Dendríticas/imunologia , Células Dendríticas/transplante , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Humanos , Interleucina-10/farmacologia , Macrófagos/imunologia , Macrófagos/transplante , Fator de Crescimento Transformador beta/farmacologia
13.
Front Immunol ; 12: 676337, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34421892

RESUMO

Dendritic cells (DCs) are key mediators of transplant rejection. Numerous factors have been identified that regulate transplant immunopathology by modulating the function of DCs. Among these, microRNAs (miRNAs), small non-coding RNA molecules, have received much attention. The miRNA miR-223 is very highly expressed and tightly regulated in hematopoietic cells. It plays an important role in modulating the immune response by regulating neutrophils and macrophages, and its dysregulation contributes to multiple types of immune diseases. However, the role of miR-223 in immune rejection is unclear. Here, we observed expression of miR-223 in patients and mice who had undergone heart transplantation and found that it increased in the serum of both, and also in DCs from the spleens of recipient mice, although it was unchanged in splenic T cells. We also found that miR-223 expression decreased in lipopolysaccharide-stimulated DCs. Increasing the level of miR-223 in DCs promoted polarization of DCs toward a tolerogenic phenotype, which indicates that miR-223 can attenuate activation and maturation of DCs. MiR-223 effectively induced regulatory T cells (Tregs) by inhibiting the function of antigen-presenting DCs. In addition, we identified Irak1 as a miR-223 target gene and an essential regulator of DC maturation. In mouse allogeneic heterotopic heart transplantation models, grafts survived longer and suffered less immune cell infiltration in mice with miR-223-overexpressing immature (im)DCs. In the miR-223-overexpressing imDC recipients, T cells from spleen differentiated into Tregs, and the level of IL-10 in heart grafts was markedly higher than that in the control group. In conclusion, miR-223 regulates the function of DCs via Irak1, differentiation of T cells into Tregs, and secretion of IL-10, thereby suppressing allogeneic heart graft rejection.


Assuntos
Células Dendríticas/imunologia , Rejeição de Enxerto/sangue , Sobrevivência de Enxerto/genética , Transplante de Coração , Quinases Associadas a Receptores de Interleucina-1/metabolismo , MicroRNAs/sangue , Transdução de Sinais/genética , Tolerância ao Transplante/genética , Animais , Transplante de Células/métodos , Células Cultivadas , Células Dendríticas/transplante , Rejeição de Enxerto/terapia , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Interleucina-10/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Modelos Animais , Linfócitos T Reguladores/imunologia , Transfecção , Transplante Homólogo
14.
Toxicol Appl Pharmacol ; 429: 115699, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34437932

RESUMO

PURPOSE: Cancer stem cells (CSC) are the most common causes of lung cancer relapse and mouse resistance to chemotherapy. CD166 was identified as CSC marker for lung cancer. Our study aimed to detect the effect of dendritic cell vaccine loaded with tumor cell lysate (TCL-DCV) on percentage of CD166+ CSC in lung of mice exposed to Benzo(a)Pyrene (BP). METHODS: Female albino mice were divided into 5 groups (22 mice per group): normal control (NC), lung cancer control (LCC) (50 mg/kg BP orally, twice weekly for four weeks), dendritic cell (DC), TCL-DCV and cisplatin. Cisplatin (6 mg/kg, intraperitoneal) was given in two doses (18th and 20th week). 1 × 106 cells of each of DC and TCL-DCV was given subcutaneously as cisplatin. At the end of experiment (22 weeks), lung tissue was used for evaluation of cytotoxic T lymphocyte antigen-4 (Ctla-4), transforming growth factor-ß (Tgf-ß), forkhead box protein P3 (Foxp3), programmed death ligand 1 (Pd-l1) and interleukin 12 (Il-12) gene expression using quantitative RT-PCR. The percentage of CD83+, CD8+ and CD166+ cells in lung tissue were measured using flow cytometry. RESULTS: The results revealed that TCL-DCV reversed the tumorigenic effect of BP in the lung as evidenced by histopathological examination. Compared to cisplatin, dendritic cell vaccination (TCL-DCV) significantly decreased percentage of CD166+ CSC. This anticancer stemness effect was attributed to the immune-stimulatory effect as indicated by increased percentage of CD83+ and CD8+ cells, upregulation of Il-12, and downregulation of Tgf-ß, Ctla-4, Pd-l1 and Foxp3 gene expression compared to LCC group. CONCLUSIONS: TCL-DCV ameliorated cancer stemness through modulating tumor immune archetypes which make it a potent therapeutic alternative to chemotherapy resistant cases.


Assuntos
Antígenos CD/metabolismo , Antineoplásicos/farmacologia , Vacinas Anticâncer , Moléculas de Adesão Celular Neuronais/metabolismo , Cisplatino/farmacologia , Células Dendríticas/transplante , Proteínas Fetais/metabolismo , Neoplasias Pulmonares/terapia , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Camundongos , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Microambiente Tumoral
15.
Int Immunopharmacol ; 98: 107833, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34352472

RESUMO

Dendritic cell (DC) vaccination can be achieved via straight loading of vaccine into DCs ex vivo or administration to DCs in vivo. However, there is no certain consensus on which approach is preferable, and each strategy has its advantages and disadvantages, which affect the efficacy and safety of vaccines. It will also be more complicated when a vaccine delivery system is included. In this study, the efficacy of ex vivo pulsed DC-based vaccine compared with in vivo subcutaneous administration of a cationic liposomes (CLs) formulation containing gp100 antigen (gp100-CLs) was evaluated in a murine melanoma model. In combination with an anti-PD-1 antibody, the ex vivo approach of gp100-CLs yielded a significant (P < 0.01) increase in the number of antigen-specific tumors infiltrated lymphocytes (TILs) with a significant upregulation of IFN-γ (P < 0.0001) and PD-1 (P < 0.0001) expression level. They also dampened the function of immunosuppressive regulatory T cells (Tregs) via significant downregulation of IL-10 and TGF-ß (P < 0.0001) expression level compared to in vivo approach in the tumor microenvironment (TME). Furthermore, prophylactic immunization with gp100-CLs pulsed DCs ex vivo delayed tumor growth and induced the survival benefit over in vivo immunization. Collectively, the ex vivo DC-based vaccination pulsed with gp100 encapsulated in liposomes synergizes with anti-PD-1 antibody and represents a preferable approach against melanoma.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Imunoterapia Adotiva/métodos , Lipossomos/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Melanoma/terapia , Neoplasias Cutâneas/terapia , Animais , Apresentação de Antígeno , Antineoplásicos Imunológicos/farmacologia , Terapia Combinada , Células Dendríticas/transplante , Modelos Animais de Doenças , Vias de Administração de Medicamentos , Humanos , Lipossomos/síntese química , Melanoma/imunologia , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Neoplasias Cutâneas/imunologia , Linfócitos T Reguladores/imunologia , Vacinação , Antígeno gp100 de Melanoma/metabolismo
16.
Gynecol Oncol ; 162(3): 652-660, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34294416

RESUMO

OBJECTIVE: DCVAC/OvCa is an active cellular immunotherapy designed to stimulate an immune response against ovarian cancer. We explored the safety and efficacy of DCVAC/OvCa plus carboplatin and gemcitabine in platinum-sensitive ovarian cancer. METHODS: In this open-label, parallel-group, phase 2 trial (ClinicalTrials.gov number NCT02107950), patients with platinum-sensitive ovarian cancer relapsing after first-line chemotherapy were randomized to DCVAC/OvCa and chemotherapy or chemotherapy alone. DCVAC/OvCa was administered every 3-6 weeks (10 doses). Endpoints included safety, progression-free survival (PFS; primary efficacy endpoint) and overall survival (OS; secondary efficacy endpoint). RESULTS: Between November 2013 and May 2015, 71 patients were randomized to chemotherapy in combination with DCVAC/OvCa or to chemotherapy alone. Treatment-emergent adverse events related to DCVAC/OvCa, leukapheresis and chemotherapy occurred in six (16.2%), two (5.4%), and 35 (94.6%) patients in the DCVAC/OvCa group. Chemotherapy-related events occurred in all patients in the chemotherapy group. Seven patients in the DCVAC/OvCa group were excluded from primary efficacy analyses due to failure to receive ≥1 dose of DCVAC/OvCa. PFS was not improved (hazard ratio [HR] 0.73, 95% confidence interval [CI] 0.42-1.28, P = 0.274, data maturity 78.1%). Median OS was significantly prolonged (by 13.4 months) in the DCVAC/OvCa group (HR 0.38, 95% CI 0.20-0.74, P = 0.003; data maturity 56.3%). A signal for enhanced surrogate antigen-specific T-cell activity was seen with DCVAC/OvCa. CONCLUSIONS: DCVAC/OvCa combined with chemotherapy had a favorable safety profile in patients with platinum-sensitive ovarian cancer. DCVAC/OvCa did not improve PFS, but the exploratory analyses revealed OS prolongation and enhanced surrogate antigen-specific T-cell activity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Epitelial do Ovário/terapia , Células Dendríticas/imunologia , Imunoterapia Adotiva/métodos , Neoplasias Ovarianas/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carboplatina/administração & dosagem , Terapia Combinada , Células Dendríticas/transplante , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Feminino , Humanos , Imunoterapia Adotiva/efeitos adversos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Gencitabina
17.
Signal Transduct Target Ther ; 6(1): 263, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34248142

RESUMO

Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-ß), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.


Assuntos
Imunidade Celular/genética , Inflamação/tratamento farmacológico , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Células Dendríticas/transplante , Humanos , Inflamassomos/efeitos dos fármacos , Inflamação/genética , Interferons/genética , Interleucinas/genética , Janus Quinases/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , NF-kappa B/genética , Neoplasias/genética , Fatores de Transcrição STAT/genética , Transdução de Sinais/genética , Receptores Toll-Like/genética , Fator de Crescimento Transformador beta/genética
18.
Front Immunol ; 12: 704776, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34262573

RESUMO

Current treatment for patients with non-small-cell lung cancer (NSCLC) is suboptimal since therapy is only effective in a minority of patients and does not always induce a long-lasting response. This highlights the importance of exploring new treatment options. The clinical success of immunotherapy relies on the ability of the immune system to mount an adequate anti-tumor response. The activation of cytotoxic T cells, the effector immune cells responsible for tumor cell killing, is of paramount importance for the immunotherapy success. These cytotoxic T cells are primarily instructed by dendritic cells (DCs). DCs are the most potent antigen-presenting cells (APCs) and are capable of orchestrating a strong anti-cancer immune response. DC function is often suppressed in NSCLC. Therefore, resurrection of DC function is an interesting approach to enhance anti-cancer immune response. Recent data from DC-based treatment studies has given rise to the impression that DC-based treatment cannot induce clinical benefit in NSCLC by itself. However, these are all early-phase studies that were mainly designed to study safety and were not powered to study clinical benefit. The fact that these studies do show that DC-based therapies were well-tolerated and could induce the desired immune responses, indicates that DC-based therapy is still a promising option. Especially combination with other treatment modalities might enhance immunological response and clinical outcome. In this review, we will identify the possibilities from current DC-based treatment trials that could open up new venues to improve future treatment.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Células Dendríticas , Imunoterapia , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Células Dendríticas/imunologia , Células Dendríticas/transplante , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia
19.
Sci Immunol ; 6(61)2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-34244314

RESUMO

Asthma is a common inflammatory lung disease with no known cure. Previously, we uncovered a lung TNFR2+ conventional DC2 subset (cDC2s) that induces regulatory T cells (Tregs) maintaining lung tolerance at steady state but promotes TH2 response during house dust mite (HDM)-induced asthma. Lung IFNß is essential for TNFR2+ cDC2s-mediated lung tolerance. Here, we showed that exogenous IFNß reprogrammed TH2-promoting pathogenic TNFR2+ cDC2s back to tolerogenic DCs, alleviating eosinophilic asthma and preventing asthma exacerbation. Mechanistically, inhaled IFNß, not IFNα, activated ERK2 signaling in pathogenic lung TNFR2+ cDC2s, leading to enhanced fatty acid oxidation (FAO) and lung Treg induction. Last, human IFNß reprogrammed pathogenic human lung TNFR2+ cDC2s from patients with emphysema ex vivo. Thus, we identified an IFNß-specific ERK2-FAO pathway that might be harnessed for DC therapy.


Assuntos
Asma/imunologia , Células Dendríticas/transplante , Interferon beta/uso terapêutico , Receptores Tipo II do Fator de Necrose Tumoral/genética , Células Th2/imunologia , Transferência Adotiva , Animais , Asma/patologia , Células Cultivadas , Células Dendríticas/imunologia , Dermatophagoides pteronyssinus/imunologia , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Humanos , Tolerância Imunológica/imunologia , Fatores Imunológicos/uso terapêutico , Interferon-alfa/farmacologia , Pulmão/citologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Oxirredução , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia
20.
Int Immunopharmacol ; 99: 107979, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34293711

RESUMO

The strong genetic association between autoimmune regulator (AIRE) and autoimmune diseases indicates its critical role in immune tolerance. AIRE deficiency is thought to promote the development of follicular helper T (TFH) cells, which are considered to be essential in B cell proliferation. Excessive TFH cell generation is a key step towards the development of autoimmune diseases, including type 1 diabetes. However, the potential mechanism by which AIRE contributes to the generation and function of the TFH cell population has remained elusive. We show that AIRE reduced TFH cell generation by inhibiting the expression of inducible costimulatory ligand (ICOSL), interleukin (IL)-6 and IL-27 in dendritic cells (DCs). To understand the precise impact of AIRE-overexpressing bone marrow-derived DCs (AIRE-BMDCs) on type 1 diabetes progression and the associated molecular mechanisms, we transferred AIRE-BMDCs to recipient NOD mice and found that transplantation of AIRE-BMDCs can prevent or delay the onset of diabetes, attenuate diabetes after the establishment of overt hyperglycaemia, and lead to the inhibition of autoreactive pathological TFH cells and germinal centre (GC) B cells. To further determine the potential mechanism underlying this TFH cell depletion, BMDCs were cotransferred with recombinant mouse ICOSL (ICOSLG protein). We demonstrated that NOD mice were more susceptible to diabetes when they received AIRE-BMDCs and ICOSLG than when they received only mock-vehicle BMDCs (GFP-BMDCs). In addition, we did not observe the reversal of diabetes in any mice subjected to this cotransfer system. A single cycle of ICOSLG treatment temporarily promoted TFH cell proliferation and GC development. Our results reveal a mechanistic role of AIRE-BMDCs in the initiation of TFH cell differentiation, and the AIRE-mediated decrease in ICOSL expression in BMDCs plays a critical role. The effect of decreased ICOSL expression in type 1 diabetes will guide the design and evaluation of parallel studies in patients.


Assuntos
Doenças Autoimunes/prevenção & controle , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 1/prevenção & controle , Ligante Coestimulador de Linfócitos T Induzíveis/antagonistas & inibidores , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Fatores de Transcrição/biossíntese , Animais , Células Dendríticas/transplante , Feminino , Centro Germinativo , Ligante Coestimulador de Linfócitos T Induzíveis/biossíntese , Interleucina-6/antagonistas & inibidores , Interleucinas/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...