Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Mol Ther ; 32(3): 734-748, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38243600

RESUMO

Despite the revolutionary success of chimeric antigen receptor (CAR)-T therapy for hematological malignancies, successful CAR-T therapies for solid tumors remain limited. One major obstacle is the scarcity of tumor-specific cell-surface molecules. One potential solution to overcome this barrier is to utilize antibodies that recognize peptide/major histocompatibility complex (MHCs) in a T cell receptor (TCR)-like fashion, allowing CAR-T cells to recognize intracellular tumor antigens. This study reports a highly specific single-chain variable fragment (scFv) antibody against the MAGE-A4p230-239/human leukocyte antigen (HLA)-A∗02:01 complex (MAGE-A4 pMHC), screened from a human scFv phage display library. Indeed, retroviral vectors encoding CAR, utilizing this scFv antibody as a recognition component, efficiently recognized and lysed MAGA-A4+ tumor cells in an HLA-A∗02:01-restricted manner. Additionally, the adoptive transfer of T cells modified by the CAR-containing glucocorticoid-induced tumor necrosis factor receptor (TNFR)-related receptor (GITR) intracellular domain (ICD), but not CD28 or 4-1BB ICD, significantly suppressed the growth of MAGE-A4+ HLA-A∗02:01+ tumors in an immunocompromised mouse model. Of note, a comprehensive analysis revealed that a broad range of amino acid sequences of the MAGE-A4p230-239 peptide were critical for the recognition of MAGE-A4 pMHC by these CAR-T cells, and no cross-reactivity to analogous peptides was observed. Thus, MAGE-A4-targeted CAR-T therapy using this scFv antibody may be a promising and safe treatment for solid tumors.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Anticorpos de Cadeia Única , Camundongos , Animais , Humanos , Anticorpos de Cadeia Única/genética , Receptores de Antígenos Quiméricos/genética , Linfócitos T , Antígenos HLA-A , Imunoterapia Adotiva
2.
Int J Cancer ; 152(12): 2554-2566, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-36727538

RESUMO

The efficacy of immune checkpoint inhibitors is limited in refractory solid tumors. T-cell receptor gene-modified T (TCR-T)-cell therapy has attracted attention as a new immunotherapy for refractory cold tumors. We first investigated the preclinical efficacy and mode of action of TCR-T cells combined with the pullulan nanogel:long peptide antigen (LPA) vaccine in a mouse sarcoma model that is resistant to immune checkpoint inhibition. Without lymphodepletion, the pullulan nanogel:LPA vaccine markedly increased the number of TCR-T cells in the draining lymph node and tumor tissue. This change was associated with enhanced CXCR3 expression in TCR-T cells in the draining lymph node. In the phase 1 trial, autologous New York esophageal squamous cell carcinoma 1 (NY-ESO-1)-specific TCR-T cells were infused twice into HLA-matched patients with NY-ESO-1+ soft tissue sarcoma (STS). The pullulan nanogel:LPA vaccine contains an epitope recognized by TCR-T cells, and it was subcutaneously injected 1 day before and 7 days after the infusion of TCR-T cells. Lymphodepletion was not performed. Three patients with refractory synovial sarcoma (SS) were treated. Two out of the three patients developed cytokine release syndrome (CRS) with low-to-moderate cytokine level elevation. We found obvious tumor shrinkage lasting for more than 2 years by tumor imaging and long-term persistence of TCR-T cells in one patient. In conclusion, NY-ESO-1-specific TCR-T-cell therapy plus vaccination with the pullulan nanogel carrying an LPA containing the NY-ESO-1 epitope without lymphodepletion is feasible and can induce promising long-lasting therapeutic effects in refractory SS (Registration ID: JMA-IIA00346).


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Sarcoma Sinovial , Neoplasias de Tecidos Moles , Vacinas , Animais , Camundongos , Nanogéis , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Antígenos de Neoplasias , Sarcoma Sinovial/terapia , Epitopos , Terapia Baseada em Transplante de Células e Tecidos
3.
Cancer Sci ; 114(11): 4172-4183, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37675556

RESUMO

Adoptive immunotherapy using genetically engineered patient-derived lymphocytes to express tumor-reactive receptors is a promising treatment for malignancy. However, utilization of autologous T cells in this therapy limits the quality of gene-engineered T cells, thereby inhibiting the timely infusion of the cells into patients. In this study, we evaluated the anti-tumor efficacy and the potential to induce graft-versus-host disease (GVHD) in T cell receptor (TCR) gene-engineered allogeneic T cells that downregulate the endogenous TCR and HLA class I molecules with the aim of developing an "off-the-shelf" cell product with expanded application of genetically engineered T cells. We transduced human lymphocytes with a high-affinity TCR specific to the cancer/testis antigen NY-ESO-1 using a novel retrovirus vector with siRNAs specific to the endogenous TCR (siTCR vector). These T cells showed reduced expression of endogenous TCR and minimized reactivity to allogeneic cells in vitro. In non-obese diabetic/SCID/γcnull mice, TCR gene-transduced T cells induced tumor regression without development of GVHD. A lentivirus-based CRISPR/Cas9 system targeting ß-2 microglobulin in TCR gene-modified T cells silenced the HLA class I expression and prevented allogeneic CD8+ T cell stimulation without disrupting their anti-tumor capacity. This report is the first demonstration that siTCR technology is effective in preventing GVHD. Adoptive cell therapy with allogeneic T cells engineered with siTCR vector may be useful in developing an "off-the-shelf" therapy for patients with malignancy.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Neoplasias , Camundongos , Animais , Humanos , RNA Interferente Pequeno/genética , Células Alógenas/metabolismo , Camundongos SCID , Receptores de Antígenos de Linfócitos T , Genes Codificadores dos Receptores de Linfócitos T , Imunoterapia Adotiva , Neoplasias/genética , Doença Enxerto-Hospedeiro/prevenção & controle
4.
Int J Mol Sci ; 24(13)2023 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-37446055

RESUMO

The benefits of CAR-T therapy could be expanded to the treatment of solid tumors through the use of derived autologous αß T cell, but clinical trials of CAR-T therapy for patients with solid tumors have so far been disappointing. CAR-T therapy also faces hurdles due to the time and cost intensive preparation of CAR-T cell products derived from patients as such CAR-T cells are often poor in quality and low in quantity. These inadequacies may be mitigated through the use of third-party donor derived CAR-T cell products which have a potent anti-tumor function but a constrained GVHD property. Vγ9Vδ2 TCR have been shown to exhibit potent antitumor activity but not alloreactivity. Therefore, in this study, CAR-T cells were prepared from Vγ9Vδ2 T (CAR-γδ T) cells which were expanded by using a novel prodrug PTA. CAR-γδ T cells suppressed tumor growth in an antigen specific manner but only during a limited time window. Provision of GITR co-stimulation enhanced anti-tumor function of CAR-γδ T cells. Our present results indicate that, while further optimization of CAR-γδ T cells is necessary, the present results demonstrate that Vγ9Vδ2 T cells are potential source of 'off-the-shelf' CAR-T cell products for successful allogeneic adoptive immunotherapy.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Neoplasias , Pró-Fármacos , Receptores de Antígenos Quiméricos , Humanos , Imunoterapia Adotiva/métodos , Difosfonatos , Receptores de Antígenos de Linfócitos T gama-delta , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Linfócitos T , Imunoterapia
5.
Blood ; 132(11): 1134-1145, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30045840

RESUMO

The recent success of chimeric antigen receptor (CAR)-T cell therapy for treatment of hematologic malignancies supports further development of treatments for both liquid and solid tumors. However, expansion of CAR-T cell therapy is limited by the availability of surface antigens specific for the tumor while sparing normal cells. There is a rich diversity of tumor antigens from intracellularly expressed proteins that current and conventional CAR-T cells are unable to target. Furthermore, adoptively transferred T cells often suffer from exhaustion and insufficient expansion, in part, because of the immunosuppressive mechanisms operating in tumor-bearing hosts. Therefore, it is necessary to develop means to further activate and expand those CAR-T cells in vivo. The Wilms tumor 1 (WT1) is an intracellular oncogenic transcription factor that is an attractive target for cancer immunotherapy because of its overexpression in a wide range of leukemias and solid tumors, and a low level of expression in normal adult tissues. In the present study, we developed CAR-T cells consisting of a single chain variable fragment (scFv) specific to the WT1235-243/HLA-A*2402 complex. The therapeutic efficacy of our CAR-T cells was demonstrated in a xenograft model, which was further enhanced by vaccination with dendritic cells (DCs) loaded with the corresponding antigen. This enhanced efficacy was mediated, at least partly, by the expansion and activation of CAR-T cells. CAR-T cells shown in the present study not only demonstrate the potential to expand the range of targets available to CAR-T cells, but also provide a proof of concept that efficacy of CAR-T cells targeting peptide/major histocompatibility complex can be boosted by vaccination.


Assuntos
Imunidade Celular , Imunoterapia Adotiva , Neoplasias/terapia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Vacinação , Proteínas WT1/imunologia , Animais , Linhagem Celular Tumoral , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Linfócitos T/patologia , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cancer Sci ; 109(10): 2998-3004, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29999574

RESUMO

Exosomes are representative extracellular vesicles (EV) derived from multivesicular endosomes (MVE) and have been described as new particles in the communication of neighborhood and/or distant cells by serving as vehicles for transfer between cells of membrane and cytosolic proteins, lipids, and nucleotides including micro (mi) RNAs. Exosomes from immune cells and tumor cells act in part as a regulator in tumor immunology. CD8+ T cells that show potent cytotoxic activity against tumor cells reside as an inactive naïve form in the T-cell zone of secondary lymphoid organs. Once receiving tumor-specific antigenic stimulation by dendritic cells (DC), CD8+ T cells are activated and differentiated into effector CTL. Subsequently, CTL circulate systemically, infiltrate into tumor lesions through the stromal neovasculature where mesenchymal stromal cells, for example, mesenchymal stem cells (MSC) and cancer-associated fibroblasts (CAF), abundantly exist, destroy mesenchymal tumor stroma in an exosome-mediated way, go into tumor parenchyma, and attack tumor cells by specific interaction. DC-derived and regulatory T (Treg) cell-derived exosomes, respectively, promote and inhibit CTL generation in this setting. In this review, we describe the roles of exosomes from immune cells and tumor cells on the regulation of tumor progression.


Assuntos
Citotoxicidade Imunológica , Exossomos/imunologia , Imunidade Celular , Neoplasias/imunologia , Antígenos de Neoplasias/imunologia , Progressão da Doença , Humanos , Invasividade Neoplásica/imunologia , Neoplasias/patologia , Transdução de Sinais/imunologia
7.
Gan To Kagaku Ryoho ; 44(5): 373-377, 2017 May.
Artigo em Japonês | MEDLINE | ID: mdl-28536330

RESUMO

In addition to the cytokines and cytotoxic granules, exosomes have been known as the intercellular communicator and cytotoxic missile of immune cells for the past decade. It has been well known that mature dendritic cell(DC)-derived exosomes participate in the T cell and natural killer(NK)cell activation, while immature DCs secrete tolerogenic exosomes for regulatory T(Treg)cell generation. Treg cell-derived EVs act as a suppressor against pathogenic type-1 T helper(Th1)cell responses. CD8+ T cells produce tumoricidal exosomes for preventing tumor invasion and metastasis transiently after T cell receptor(TCR)-mediated stimulation. Thus, immune cells produce functional exosomes in the activation state- and/or differentiation stage-dependent manner. In this review, the role of immune cell-derived exosomes will be introduced, focusing mainly on immune reaction against tumor.


Assuntos
Células Dendríticas/imunologia , Exossomos , Células Matadoras Naturais/imunologia , Linfócitos T/imunologia , Humanos , Neoplasias/imunologia
8.
Small Methods ; 6(2): e2100785, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35174988

RESUMO

Extracellular vesicles (EVs) are released by all types of mammalian cells for cell-cell communication. In this study, surface glycans on EVs are compared in terms of their cell type, size, and isolation method to examine whether EV glycan profiles by lectin microarray can be used to define EV subpopulations. Moreover, EVs are glycoengineered with four distinctive surface glycan patterns and evaluated their cellular uptake efficiencies for potential drug delivery applications. Both similarities and differences in glycan patterns are identified on EVs obtained under each experimental condition. EV size- and isolation method-dependent lectin-binding patterns are observed. Moreover, cellular uptake behaviors of EVs are affected by EV glycan profiles and acceptor cells. The in vivo biodistribution of EVs is also dependent on their glycan profile. These results suggest that EV surface glycans are a potential novel indicator of EV heterogeneity, and glycoengineering is a useful approach to regulate cell-EV interactions for biomedical applications.


Assuntos
Vesículas Extracelulares/transplante , Lectinas/metabolismo , Análise em Microsséries/métodos , Polissacarídeos/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Vesículas Extracelulares/metabolismo , Células HCT116 , Células HT29 , Humanos , Injeções Intravenosas , Camundongos , Células PC-3 , Distribuição Tecidual
9.
J Extracell Vesicles ; 11(3): e12205, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35289089

RESUMO

The development of a new large-scale purification protocol is required for research on the reliable bioactivity and drug discovery of extracellular vesicles (EVs). To address this issue, herein, we propose an effective method for preparing high-performance exosomes (EXOs) by using an anion-exchange method. Cytotoxic T-lymphocyte (CTL) EVs from 4 L of culture supernatant through a 220 nm cut-off filter are divided into two populations at a deproteinization rate of over 99.97%, which are eluted at low (0.15 M-0.3 M) and high (0.3 M-0.5 M) NaCl concentrations (approximately 2 × 1012 and 1.5 × 1012 particles, respectively) through the anion-exchange column chromatography. The former are abundant in EXO proteins, including late endosome-associated proteins and rab-family and integrin-family proteins, and functional micro (mi) RNAs, and have bioactivity for preventing tumour metastasis by depleting mesenchymal cell populations in the primary tumour lesions. By contrast, the latter is microvesicle (MV)-like particles including DNA, core histone and ribosomal proteins, and GC-rich miRNAs with unknown function, and are easily phagocytosed by mannose receptor+ Kupffer cells. Thus, the anion-exchange method is suitable for the large-scale separation of bioactive EXOs and MV-like EVs as a cargo for dangerous nucleic acids at high-purity.


Assuntos
Exossomos , Vesículas Extracelulares , Neoplasias , Ácidos Nucleicos , Ânions/análise , Exossomos/genética , Vesículas Extracelulares/química , Humanos , Neoplasias/diagnóstico , Ácidos Nucleicos/análise
10.
Cancer Immunol Immunother ; 60(7): 1029-38, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21519830

RESUMO

Staphylococcal enterotoxins A (SEA) and B (SEB) are classical models of superantigens (SAg), which induce potent T-cell-stimulating activity by forming complexes with MHC class II molecules on antigen-presenting cells. This large-scale activation of T-cells is accompanied by increased production of cytokines such as interferon-γ (IFN-γ). Additionally, as we previously reported, IFN-γ-producing CD8(+) T cells act as "helper cells," supporting the ability of dendritic cells to produce interleukin-12 (IL-12)p70. Here, we show that DC pulsed with SAg promote the enhancement of anti-tumor immunity. Murine bone marrow-derived dendritic cells (DC) were pulsed with OVA(257-264) (SIINFEKL), which is an H-2Kb target epitope of EG7 [ovalbumin (OVA)-expressing EL4] cell lines, in the presence of SEA and SEB and were subcutaneously injected into naïve C57BL/6 mice. SAg plus OVA(257-264)-pulsed DC vaccine strongly enhanced peptide-specific CD8(+) T cells exhibiting OVA(257-264)-specific cytotoxic activity and IFN-γ production, leading to the induction of protective immunity against EG7 tumors. Furthermore, cyclophosphamide (CY) added to SAg plus tumor-antigens (OVA(257-264), tumor lysate, or TRP-2) pulsed DC immunization markedly enhanced tumor-specific T-cell expansion and had a significant therapeutic effect against various tumors (EG7, 2LL, and B16). Superantigens are potential candidates for enhancing tumor immunity in DC vaccines.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Carcinoma Pulmonar de Lewis/imunologia , Células Dendríticas/imunologia , Linfoma/imunologia , Melanoma Experimental/imunologia , Superantígenos/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antineoplásicos Alquilantes/uso terapêutico , Linfócitos T CD8-Positivos , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Ciclofosfamida/uso terapêutico , Citocinas/metabolismo , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/metabolismo , Interferon gama/metabolismo , Interleucina-12/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Ativação Linfocitária , Linfoma/tratamento farmacológico , Linfoma/metabolismo , Masculino , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Taxa de Sobrevida , Linfócitos T Auxiliares-Indutores/imunologia , Células Tumorais Cultivadas , Vacinas de Subunidades Antigênicas/uso terapêutico
11.
J Immunol ; 181(11): 8096-102, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19018002

RESUMO

Gold compounds clinically used as immunomodulators have high potential to evoke hypersensitivity reactions as an adverse effect. To explore the mechanism of gold allergy, we immunologically characterized T cells infiltrating skin rashes and generated 44 gold-specific T cell clones and lines from a rheumatoid arthritis patient who developed skin rashes and systemic symptoms after gold treatment. CD4(+) and CD8(+) cells predominantly infiltrating the skin rashes and some of the T cell clones and lines shared common Vbetas. These cells exhibited Th0-like, Th2-like, and Tc1-like cytokine profiles, and showed chemotactic activities for thymus and activation-regulated chemokine and IFN-gamma-inducible protein-10 corresponding to the cytokine profiles. T cell recognition of gold consisted of MHC-restricted and MHC-independent pathways. Blocking studies with anti-MHC Abs indicated that the groove of MHC in APCs, where Ags should ordinarily be settled, did not serve as a conjugating site of gold for these T cells in certain cases. These observations raise the possibility that gold-specific CD4(+) and CD8(+) T cells and APCs promiscuously interact under stimulation with gold, resulting in various clinical manifestations in gold allergy.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Antirreumáticos/efeitos adversos , Comunicação Celular/imunologia , Hipersensibilidade a Drogas/imunologia , Tiomalato Sódico de Ouro/efeitos adversos , Pele/imunologia , Linfócitos T Citotóxicos/imunologia , Células Th2/imunologia , Idoso , Células Apresentadoras de Antígenos/patologia , Antirreumáticos/administração & dosagem , Comunicação Celular/efeitos dos fármacos , Quimiocina CXCL10/imunologia , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/imunologia , Hipersensibilidade a Drogas/patologia , Feminino , Ouro , Tiomalato Sódico de Ouro/administração & dosagem , Antígenos de Histocompatibilidade/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Pele/patologia , Linfócitos T Citotóxicos/patologia , Células Th2/patologia , Timo/imunologia , Timo/patologia
12.
Biomedicines ; 8(9)2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32825298

RESUMO

The medical comorbidities including skin diseases are associated with male infertility. The most common cause of male infertility is the inability of testes to produce sperm; however, the influence of persistent dermatitis on testicular function has not been elucidated so far. We investigated the relationship between skin inflammation and impaired sperm production using a spontaneous dermatitis mouse model. We examined the breeding records of dermatitis mice and their wild-type littermates. Sperm count, motility, and viability were analyzed by direct microscopic observation and flow cytometry. In addition, testis and epididymis were histologically examined. Finally, sperm viability was evaluated in another dermatitis mouse model and in wild-type mice in which inflammatory cytokines were intraperitoneally administered. Compared to wild-type littermate mice, the number of children born was lower in mice with dermatitis. The body weight and testis size were decreased age-dependently. In the skin disease group, the sperm count and movement ratio were clearly decreased, and reduced sperm viability was observed. Histological examination revealed the detachment of Sertoli cells and reduced spermatogenesis. The fibrosis of epididymal stroma was severe, and it might affect defective sperm maturation in the epididymis. In addition, this phenomena was reproduced by a hapten applied dermatitis mouse model and the intraperitoneal administration of inflammatory cytokines. Once the skin is inflamed, inflammatory cytokines are produced and released, which may affect testicular and sperm function. Additional studies are needed to determine the relationship between male infertility and severe dermatitis in human.

13.
Anticancer Res ; 29(4): 1015-22, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19414340

RESUMO

BACKGROUND: A drawback of cancer gene therapy is the failure of toxic gene introduction into a proportion of the tumor cells, resulting in re-progression of the disease. Cancer cell-specificity of the gene introduction has also been problematic. MATERIALS AND METHODS: Previously defined promoter/enhancer DNA of Q5 tumor antigen gene was used for the purpose of transcriptional targeting. A replication defective adenovirus carrying H-2D(d) cDNA placed downstream of the Q5 promoter/enhancer (Ad/Q5-H-2D(d)) was constructed and administered systemically to C57Bl/6 (H-2(b)) mice bearing pre-established hepatic metastasis of the syngeneic M5076 tumor. RESULTS: 5 of the 10 mice showed complete regression of the tumor. Combination of the therapy with low dose cyclophosphamide (CY) administration augmented the therapeutic effect. The effect was brought about by tumor cell-specific allograft rejection reactions and by tumor antigen-specific reactions induced as "bystander effect" by the former reactions. CONCLUSION: The results provide an experimental basis of a highly efficient novel strategy of cancer gene therapy.


Assuntos
Adenoviridae/genética , Genes Neoplásicos/genética , Terapia Genética , Antígenos H-2/genética , Neoplasias Hepáticas/terapia , Transcrição Gênica , Transgenes/fisiologia , Animais , Antineoplásicos Alquilantes/administração & dosagem , Terapia Combinada , Ciclofosfamida/administração & dosagem , DNA Complementar/genética , Vetores Genéticos , Rejeição de Enxerto , Antígenos H-2/metabolismo , Antígeno de Histocompatibilidade H-2D , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundário , Complexo Principal de Histocompatibilidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Taxa de Sobrevida , Transplante Homólogo , Replicação Viral
14.
J Clin Invest ; 129(3): 1278-1294, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30628894

RESUMO

Immune checkpoint inhibitors and adoptive transfer of gene-engineered T cells have emerged as novel therapeutic modalities for hard-to-treat solid tumors; however, many patients are refractory to these immunotherapies, and the mechanisms underlying tumor immune resistance have not been fully elucidated. By comparing the tumor microenvironment of checkpoint inhibition-sensitive and -resistant murine solid tumors, we observed that the resistant tumors had low immunogenicity. We identified antigen presentation by CD11b+F4/80+ tumor-associated macrophages (TAMs) as a key factor correlated with immune resistance. In the resistant tumors, TAMs remained inactive and did not exert antigen-presenting activity. Targeted delivery of a long peptide antigen to TAMs by using a nano-sized hydrogel (nanogel) in the presence of a TLR agonist activated TAMs, induced their antigen-presenting activity, and thereby transformed the resistant tumors into tumors sensitive to adaptive immune responses such as adoptive transfer of tumor-specific T cell receptor-engineered T cells. These results indicate that the status and function of TAMs have a significant impact on tumor immune sensitivity and that manipulation of TAM functions would be an effective approach for improving the efficacy of immunotherapies.


Assuntos
Apresentação de Antígeno , Antígenos de Neoplasias/farmacologia , Sistemas de Liberação de Medicamentos , Hidrogéis/farmacologia , Macrófagos/imunologia , Nanopartículas/química , Neoplasias Experimentais/terapia , Microambiente Tumoral/efeitos dos fármacos , Transferência Adotiva , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Feminino , Hidrogéis/química , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Linfócitos T/transplante , Microambiente Tumoral/genética
15.
Curr Med Chem ; 15(10): 991-6, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18393856

RESUMO

Aggressive immunity characterized by the motion of cytotoxic T lymphocytes (CTLs), T helper (Th) 1 cells, and natural killer (NK) cells is the first line of defense against intracellular microorganism invasion and tumor formation. In patients with infectious diseases and tumors, aggressive immunity is often attenuated by immune suppressive effects provided by regulatory T (Treg) cells including CD4(+) CD25(+) forkhead-box (fox) p3(+) T cells, T regulatory (Tr) 1, Th3, and a subpopulation of gammadelta-type of T cell receptor-expressing T (gammadelta T) cells. It has been demonstrated that Treg cells down-regulate aggressive immunity by direct cell interactions and suppressive cytokines (e.g., interleukin (IL)-10, transforming growth factor (TGF)-beta). Today, instead of synthesizing chemical agents with serious side effects, protein agents, catalytic oligonucleotides, and natural medicines involved in the elimination of Treg cell-mediated suppressive responses for the restoration of aggressive immunity are expected to be alternatives as a mild clinical remedy against microorganism invasion and tumors.


Assuntos
Anti-Infecciosos/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Pareamento de Bases , Sequência de Bases , Humanos , Terapia de Imunossupressão , Subpopulações de Linfócitos/imunologia , Dados de Sequência Molecular , Neoplasias/imunologia , Linfócitos T Reguladores/efeitos dos fármacos
16.
Cancer Res ; 66(20): 10136-44, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17047078

RESUMO

Percutaneous peptide immunization (PPI) is a simple and noninvasive immunization approach to induce potent CTL responses by peptide delivery via skin with the stratum corneum removed. After such a barrier disruption in human skin, epidermal Langerhans cells, although functionally matured through the up-regulation of HLA expression and costimulatory molecules, were found to emigrate with a reduced number of dendrites. CD8(+) populations binding to MHC-peptide tetramers/pentamers and producing IFN-gamma appeared in the blood after PPI with HLA class I-restricted antigenic peptides. PPI with melanoma-associated peptides reduced the lesion size and suppressed further development of tumors in four of seven patients with advanced melanoma. These beneficial effects were accompanied by the generation of circulating CTLs with in vitro cytolytic activity and extensive infiltration of tetramer/pentamer-binding cells into regressing lesions. PPI elicited neither local nor systemic toxicity or autoimmunity, except for vitiligo, in patients with melanoma. Therefore, PPI represents a novel therapeutic intervention for cancer in the clinical setting.


Assuntos
Melanoma/imunologia , Melanoma/terapia , Peptídeos/administração & dosagem , Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Administração Cutânea , Adulto , Idoso , Antígenos de Neoplasias/imunologia , Movimento Celular/imunologia , Epitopos de Linfócito T/imunologia , Seguimentos , Humanos , Interferon gama/biossíntese , Interferon gama/imunologia , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Estadiamento de Neoplasias
17.
Nat Commun ; 9(1): 435, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29382847

RESUMO

Fibroblastic tumour stroma comprising mesenchymal stem cells (MSCs) and cancer-associated fibroblasts (CAFs) promotes the invasive and metastatic properties of tumour cells. Here we show that activated CD8+ T cell-derived extracellular vesicles (EVs) interrupt fibroblastic stroma-mediated tumour progression. Activated CD8+ T cells from healthy mice transiently release cytotoxic EVs causing marked attenuation of tumour invasion and metastasis by apoptotic depletion of mesenchymal tumour stromal cells. Infiltration of EV-producing CD8+ T cells is observed in neovascular areas with high mesenchymal cell density, and tumour MSC depletion is associated with preferential engulfment of CD8+ T cell EVs in this setting. Thus, CD8+ T cells have the capacity to protect tumour progression by EV-mediated depletion of mesenchymal tumour stromal cells in addition to their conventional direct cytotoxicity against tumour cells.


Assuntos
Linfócitos T CD8-Positivos/citologia , Vesículas Extracelulares/fisiologia , Células-Tronco Mesenquimais/patologia , Neoplasias Experimentais/terapia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Células Cultivadas , Vesículas Extracelulares/transplante , Feminino , Células-Tronco Mesenquimais/imunologia , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/patologia
18.
J Dermatol Sci ; 48(2): 77-85, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17719748

RESUMO

Dendritic cell (DC)-based tumor immunotherapy is widely known to elicit protective anti-tumor immune responses, although the safety and effectiveness have yet to be thoroughly explored. We reported that a disruption in the stratum corneum barrier resulted in enhanced permeability and alterations in the skin immune system in such a way that epidermal Langerhans cells (LCs) functioned as vigorous antigen presenters for T helper (Th) cells and cytotoxic T lymphocytes (CTLs). In both human and murine models, topical application of melanoma-associated antigen peptides onto stratum corneum barrier-disrupted skin, specifically induced tumoricidal immune responses in vivo and in vitro accompanying an increased expression of MHC and co-stimulatory molecules on LCs. In addition, for reasons of simplicity, safety and effectiveness, percutaneous peptide application has demonstrated a certain degree of feasibility in clinical approach in patients with melanoma. In the future, resolution of some of the outstanding issues concerning the selection of the most effective adjuvants in combination with barrier disruption and depletion of regulatory T (Treg) cell-mediated immune suppression would appear as essential to improve percutaneous melanoma immunotherapy.


Assuntos
Antígenos de Neoplasias/administração & dosagem , Imunoterapia Ativa/métodos , Células de Langerhans/fisiologia , Melanoma/terapia , Linfócitos T Citotóxicos/fisiologia , Administração Cutânea , Epiderme/imunologia , Humanos , Imunoterapia Ativa/tendências , Melanoma/imunologia , Peptídeos/administração & dosagem
19.
Oncotarget ; 8(19): 30766-30780, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28430604

RESUMO

Long-surviving memory CD8+ T cells generated by stimulation with appropriate tumor-associated antigens are the most aggressive and persistent tumoricidal effectors. In this event of memory CD8+ T cell development, the signal transducer and activator of transcription (STAT) proteins function as the crucial intracellular signaling molecules, but the regulatory mechanism of STATs in CD8+ T cells is not fully understood. In this study, we report for the first time, by using murine vaccination models, that signal-transducing adaptor protein-2 (STAP2) maintains the cytotoxicity of long-lived memory CD8+ T cells by controlling a STAT3/suppressor of cytokine signaling 3 (SOCS3) cascade. Following T cell activation, STAP2 expression was transiently reduced but was subsequently recovered and augmented. Analysis using small-interfering RNA (siRNA) demonstrated that restored STAP2 expression was associated with the activation of STAT3/SOCS3 signals and maintenance of cytotoxic T lymphocytes (CTLs) secondary responses by preventing their differentiation into terminal effector cells. Notably, this STAP2-dependent memory differentiation was observed in the spleen, but not in the lymph nodes (LNs). These findings indicate an essential role for STAP2 in the generation of a high-quality memory CD8+ CTLs periphery, and suggest the therapeutic potential of STAP2 in cancer patients.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Epitopos de Linfócito T/imunologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Ativação Linfocitária/imunologia , Proteínas de Membrana/imunologia , Camundongos , Fator de Transcrição STAT3/metabolismo , Baço/imunologia , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Vacinas de DNA/imunologia , Vacinas de Subunidades Antigênicas/imunologia
20.
Am J Surg Pathol ; 30(9): 1111-9, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16931956

RESUMO

Interactions between chemokines and chemokine receptors are involved in migration and invasion of lymphoma cells. We investigated expression profiles of CXCR3 and CCR4 by immunohistochemistry and flow cytometry, and their biologic behaviors by real-time horizontal chemotaxis assay in cutaneous T cell and NK/T-cell lymphomas (TCLs). Tumor cells in mycosis fungoides (MF) constantly expressed CXCR3 at the patch stage, and expressed CCR4 at the tumor stage and in the folliculotropic variant of MF. Neoplastic cells at the plaque stage expressed CXCR3 and/or CCR4. Sezary cells in the dermis and circulation were positive for CCR4. Epidermotropic atypical cells in pagetoid reticulosis expressed CXCR3. CD30 cells exclusively expressed CCR4 in anaplastic large-cell lymphoma, and CXCR3 and/or CCR4 in lymphomatoid papulosis. In CD8TCL and extranodal NK/TCL characterized by extensive epidermotropism, tumor cells were positive for CXCR3. These data demonstrated preferential expression of CXCR3 in epidermotropic tumor cells, and of CCR4 in dermis-based lymphomas. In chemotaxis assays, CCR4 tumor cells in MF and CXCR3 tumor cells in CD8TCL migrated to thymus and activation-regulated chemokine and inducible protein-10, respectively. Therefore, spatial and temporal interactions between chemokine receptors and their ligands seem to dictate recruitment and retention of lymphoma cells in the skin.


Assuntos
Células Matadoras Naturais , Linfoma Cutâneo de Células T/química , Receptores de Quimiocinas/análise , Neoplasias Cutâneas/química , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Quimiotaxia/fisiologia , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Doenças Linfáticas/metabolismo , Linfoma Difuso de Grandes Células B/química , Masculino , Pessoa de Meia-Idade , Micose Fungoide/química , Receptores CCR4 , Receptores CXCR3
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA