Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Nat Commun ; 13(1): 954, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35177645

RESUMO

Hepatocellular carcinoma (HCC) invariably exhibits inadequate O2 (hypoxia) and nutrient supply. Hypoxia-inducible factor (HIF) mediates cascades of molecular events that enable cancer cells to adapt and propagate. Macropinocytosis is an endocytic process initiated by membrane ruffling, causing the engulfment of extracellular fluids (proteins), protein digestion and subsequent incorporation into the biomass. We show that macropinocytosis occurs universally in HCC under hypoxia. HIF-1 activates the transcription of a membrane ruffling protein, EH domain-containing protein 2 (EHD2), to initiate macropinocytosis. Knockout of HIF-1 or EHD2 represses hypoxia-induced macropinocytosis and prevents hypoxic HCC cells from scavenging protein that support cell growth. Germline or somatic deletion of Ehd2 suppresses macropinocytosis and HCC development in mice. Intriguingly, EHD2 is overexpressed in HCC. Consistently, HIF-1 or macropinocytosis inhibitor suppresses macropinocytosis and HCC development. Thus, we show that hypoxia induces macropinocytosis through the HIF/EHD2 pathway in HCC cells, harnessing extracellular protein as a nutrient to survive.


Assuntos
Carcinoma Hepatocelular/imunologia , Proteínas de Transporte/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Hepáticas/imunologia , Pinocitose/imunologia , Hipóxia Tumoral/genética , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Knockout , Pinocitose/efeitos dos fármacos , Pinocitose/genética , Estudo de Prova de Conceito , Hipóxia Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
BMC Cancer ; 21(1): 1271, 2021 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-34819027

RESUMO

INTRODUCTION: Recent data has demonstrated that hypoxia drives an immunosuppressive tumour microenvironment (TME) via various mechanisms including hypoxia inducible factor (HIF)-dependent upregulation of programmed death ligand 1 (PD-L1). Both hypoxia and an immunosuppressive TME are targetable independent negative prognostic factors for bladder cancer. Therefore we sought to investigate whether hypoxia is associated with upregulation of PD-L1 in the disease. MATERIALS AND METHODS: Three human muscle-invasive bladder cancer cell lines (T24, J82, UMUC3) were cultured in normoxia (20% oxygen) or hypoxia (1 and 0.1% oxygen) for 24 h. Differences in PD-L1 expression were measured using Western blotting, quantitative polymerase chain reaction (qPCR) and flow cytometry (≥3 independent experiments). Statistical tests performed were unpaired t tests and ANOVA. For in silico work an hypoxia signature was used to apply hypoxia scores to muscle-invasive bladder cancers from a clinical trial (BCON; n = 142) and TCGA (n = 404). Analyses were carried out using R and RStudio and statistical tests performed were linear models and one-way ANOVA. RESULTS: When T24 cells were seeded at < 70% confluence, there was decreased PD-L1 protein (p = 0.009) and mRNA (p < 0.001) expression after culture in 0.1% oxygen. PD-L1 protein expression decreased in both 0.1% oxygen and 1% oxygen in a panel of muscle-invasive bladder cancer cells: T24 (p = 0.009 and 0.001), J82 (p = 0.008 and 0.013) and UMUC3 (p = 0.003 and 0.289). Increasing seeding density decreased PD-L1 protein (p < 0.001) and mRNA (p = 0.001) expression in T24 cells grown in both 20 and 1% oxygen. Only when cells were 100% confluent, were PD-L1 protein and mRNA levels higher in 1% versus 20% oxygen (p = 0.056 and p = 0.037). In silico analyses showed a positive correlation between hypoxia signature scores and PD-L1 expression in both BCON (p = 0.003) and TCGA (p < 0.001) cohorts, and between hypoxia and IFNγ signature scores (p < 0.001 for both). CONCLUSION: Tumour hypoxia correlates with increased PD-L1 expression in patient derived bladder cancer tumours. In vitro PD-L1 expression was affected by cell density and decreased PD-L1 expression was observed after culture in hypoxia in muscle-invasive bladder cancer cell lines. As cell density has such an important effect on PD-L1 expression, it should be considered when investigating PD-L1 expression in vitro.


Assuntos
Antígenos de Neoplasias/metabolismo , Antígeno B7-H1/metabolismo , Hipóxia Tumoral , Microambiente Tumoral , Neoplasias da Bexiga Urinária/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Contagem de Células , Linhagem Celular Tumoral , Humanos , RNA Mensageiro/metabolismo , Hipóxia Tumoral/imunologia , Microambiente Tumoral/imunologia , Regulação para Cima , Neoplasias da Bexiga Urinária/imunologia , Neoplasias da Bexiga Urinária/patologia
3.
Cancer Med ; 10(18): 6317-6326, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34363337

RESUMO

BACKGROUND: 2-deoxy-2-[fluorine-18] fluoro-d-glucose (18 F-FDG) positron emission tomography (18 F-FDG-PET) is a convenient modality to assess the metabolic activity within tumor cells. However, there is no consensus regarding the relationship between 18 F-FDG uptake and the immune environment in thymic epithelial tumors (TETs). We conducted a clinicopathological study to elucidate the relationship between 18 F-FDG uptake and programmed death ligands 1 and 2 (PD-L1/PD-L2) expression in patients with TETs. METHODS: A total of 108 patients with histologically confirmed TETs classified as thymomas or thymic carcinomas who underwent surgical resection or biopsy or needle biopsy and 18 F-FDG PET before any treatment between August 2007 and March 2020 were enrolled in this study. Tumor specimens underwent immunohistochemical staining for PD-L1, PD-L2, GLUT1, HIF-1α, VEGFR2, VEGF-C, and ß2 adrenergic receptor. RESULTS: High uptakes of SUVmax , SUVmean , MTV, and TLG were identified in 28 (25.9%), 61 (56.5%), 55 (50.9%), and 55 (50.9%) of 108 patients, respectively. High uptake of SUVmax significantly correlated with PS (performance status) of 1-2, thymic carcinoma, and advanced stage, and SUVmax on 18 F-FDG uptake displayed a close association with PD-L1 and PD-L2 expressions, but not with MTV and TLG. Our analysis revealed that SUVmax was identified as being significant relationship for positive PD-L1/PD-L2 expression. GLUT1, HIF-1α, and VEGFR2 were significantly associated with the expression of PD-L1/PD-L2 from the biological viewpoint. CONCLUSION: 18 F-FDG accumulation was closely associated with the expression of PD-L1/PD-L2, which, in turn, was correlated with glucose metabolism and hypoxia. PD-L1/PD-L2 could affect the glucose metabolism and hypoxia in thymic tumor cells.


Assuntos
Neoplasias Epiteliais e Glandulares/imunologia , Timoma/imunologia , Timo/diagnóstico por imagem , Neoplasias do Timo/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígeno B7-H1/análise , Antígeno B7-H1/metabolismo , Biópsia , Feminino , Fluordesoxiglucose F18/administração & dosagem , Transportador de Glucose Tipo 1/análise , Transportador de Glucose Tipo 1/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/diagnóstico , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Epiteliais e Glandulares/cirurgia , Tomografia por Emissão de Pósitrons/métodos , Tomografia por Emissão de Pósitrons/estatística & dados numéricos , Proteína 2 Ligante de Morte Celular Programada 1/análise , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Estudos Retrospectivos , Timectomia , Timoma/diagnóstico , Timoma/patologia , Timoma/cirurgia , Timo/imunologia , Timo/patologia , Timo/cirurgia , Neoplasias do Timo/diagnóstico , Neoplasias do Timo/patologia , Neoplasias do Timo/cirurgia , Hipóxia Tumoral/imunologia , Efeito Warburg em Oncologia
4.
Cancer Med ; 10(18): 6365-6373, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34387383

RESUMO

Aerobic exercise is an important non-pharmacological means of antitumor intervention, but related mechanisms are poorly understood. In this review, previous studies are summarized from the aspects of tumor oxygenation, autophagy versus apoptosis, and organismal immunity. Current findings on the antitumor effects of aerobic exercise involve AMPK signaling, PI3K/Akt signaling, Th1/Th2 cytokine balance related to immunity, PD-1/PD-L1 immunosuppressive signaling, and related cytokine pathways. Several directions for further research are proposed, including whether newly discovered subgroups of cytokines influence the effects of aerobic exercise on tumors, tailoring corresponding exercise prescriptions based on the bidirectional effects of certain cytokines at different stages, identifying the potential effects of exercise time and intensity, and elucidating details of the unclear mechanisms. Through the discussion of the existing data, we hope to provide new ideas for the future research of exercise therapy.


Assuntos
Terapia por Exercício/métodos , Exercício Físico/imunologia , Neoplasias/terapia , Animais , Apoptose/imunologia , Autofagia/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Condicionamento Físico Animal/métodos , Transdução de Sinais/imunologia , Hipóxia Tumoral/imunologia , Microambiente Tumoral/imunologia
5.
Cell Commun Signal ; 19(1): 76, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34256773

RESUMO

Hypoxia is a pathological condition common to many diseases, although multiple organ injuries induced by hypoxia are often overlooked. There is increasing evidence to suggest that the hypoxic environment may activate innate immune cells and suppress adaptive immunity, further stimulating inflammation and inhibiting immunosurveillance. We found that dysfunctional immune regulation may aggravate hypoxia-induced tissue damage and contribute to secondary injury. Among the diverse mechanisms of hypoxia-induced immune dysfunction identified to date, the role of programmed death-ligand 1 (PD-L1) has recently attracted much attention. Besides leading to tumour immune evasion, PD-L1 has also been found to participate in the progression of the immune dysfunction which mediates hypoxia-induced multiple organ injury. In this review, we aimed to summarise the role of immune dysfunction in hypoxia-induced multiple organ injury, the effects of hypoxia on the cellular expression of PD-L1, and the effects of upregulated PD-L1 expression on immune regulation. Furthermore, we summarise the latest information pertaining to the involvement, diagnostic value, and therapeutic potential of immunosuppression induced by PD-L1 in various types of hypoxia-related diseases, including cancers, ischemic stroke, acute kidney injury, and obstructive sleep apnoea. Video Abstract.


Assuntos
Imunidade Adaptativa/genética , Antígeno B7-H1/imunologia , Inflamação/imunologia , Hipóxia Tumoral/genética , Injúria Renal Aguda/genética , Injúria Renal Aguda/imunologia , Imunidade Adaptativa/imunologia , Antígeno B7-H1/genética , Humanos , Imunidade Inata/genética , Inflamação/genética , AVC Isquêmico/genética , AVC Isquêmico/imunologia , Monitorização Imunológica , Neoplasias/genética , Neoplasias/imunologia , Apneia Obstrutiva do Sono/genética , Apneia Obstrutiva do Sono/imunologia , Hipóxia Tumoral/imunologia
6.
Int Immunopharmacol ; 98: 107895, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34171623

RESUMO

Tumor resistance to therapy modalities is one of the major challenges to the eradication of cancer cells and complete treatment. Tumor includes a wide range of cancer and non-cancer cells that play key roles in the proliferation of cancer cells and suppression of anti-tumor immunity. For overcoming tumor resistance to therapy, it is important to have in-depth knowledge relating to intercellular communications within the tumor microenvironment (TME). TME includes various types of immune cells such as CD4 + T lymphocytes, cytotoxic T lymphocytes (CTLs), natural killer (NK) cells, macrophages, and T regulatory cells (Tregs). Furthermore, some non-immune cells like cancer stem cells (CSCs), mesenchymal stem cells (MSCs), and cancer-associated fibroblasts (CAFs) are involved in the promotion of tumor growth. The interactions between these cells with cancer cells play a key role in tumor growth or inhibition. Resveratrol as a natural agent has shown the ability to modulate the immune system to potentiate anti-tumor immunity and also help to attenuate cancer cells and CSCs resistance. Thus, this review explains how resveratrol can modulate interactions within TME.


Assuntos
Neoplasias/tratamento farmacológico , Resveratrol/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Fibroblastos Associados a Câncer/efeitos dos fármacos , Fibroblastos Associados a Câncer/imunologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/imunologia , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/imunologia , Resveratrol/uso terapêutico , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Hipóxia Tumoral/efeitos dos fármacos , Hipóxia Tumoral/imunologia , Microambiente Tumoral/imunologia
7.
J Exp Clin Cancer Res ; 40(1): 197, 2021 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-34154610

RESUMO

Hypoxia in solid tumors is an important predictor of treatment resistance and poor clinical outcome. The significance of hypoxia in the development of resistance to radiotherapy has been recognized for decades and the search for hypoxia-targeting, radiosensitizing agents continues. This review summarizes the main hypoxia-related processes relevant for radiotherapy on the subcellular, cellular and tissue level and discusses the significance of hypoxia in radiation oncology, especially with regard to the current shift towards hypofractionated treatment regimens. Furthermore, we discuss the strategies to interfere with hypoxia for radiotherapy optimization, and we highlight novel insights into the molecular pathways involved in hypoxia that might be utilized to increase the efficacy of radiotherapy.


Assuntos
Neoplasias/radioterapia , Radioterapia (Especialidade)/métodos , Humanos , Hipóxia Tumoral/imunologia
8.
Clin Transl Oncol ; 23(8): 1678-1687, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33625672

RESUMO

BACKGROUND: The study examines the function of hypoxia-mediated down-regulation of microRNAs (miRNAs) (mir-30c, mir-135a, and mir-27a) in the process of bladder cancer immune escape. METHODS: Quantitative Real-time PCR (qRT-PCR) was carried out to determine gene expression levels of Drosha and Dicer under hypoxia treatment, while western blotting and flow cytometry were used to determine protein expression. Seven reported miRNAs were identified via qRT-PCR assay. Flow cytometry detection of CD3/CD4/CD8-positive expression and statistics. Enzyme-linked immunosorbent assay (ELISA) detected cellular immune factors content. Cell apoptosis was checked via flow cytometry assay. Luciferase report assay and western blot assays were both used to verify the relationship between miRNAs and Casitas B-lineage lymphoma proto-oncogene b (Cbl-b). The animal model was established and Hematoxylin-eosin (HE) staining, TdT-mediated dUTP Nick-End Labeling (TUNEL) staining, and immunohistochemistry (IHC) assays were separately used to verify the conclusions. RESULTS: The CD3 + /CD4 + expression was increased in the hypoxia group, while CD3 + /CD8 + expression, the cellular immune factors content Interleukin-2 (IL-2) and Tumor Necrosis Factor-α (TNFα) along with the cell apoptosis were suppressed. The protein expression of Cbl-b was found to be up-regulated in the hypoxia group. After constructing the overexpression/ knockdown of Cbl-b in peripheral blood mononuclear cell (PBMC), Cbl-b has been found to promote tumor immune escape in bladder cancer. Furthermore, Cbl-b had been identified as the co-targets of mir-30c, mir-135a, and mir-27a and down-regulation of miRNA biogenesis promotes Cbl-b expression and deactivating T cells in vitro/in vivo. CONCLUSION: Hypoxia-mediated down-regulation of miRNAs' biogenesis promotes tumor immune escape in bladder cancer, which could bring much more advance to the medical research on tumors.


Assuntos
Regulação para Baixo/imunologia , MicroRNAs/metabolismo , Evasão Tumoral/imunologia , Hipóxia Tumoral/imunologia , Neoplasias da Bexiga Urinária/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Complexo CD3/metabolismo , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , RNA Helicases DEAD-box/genética , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-2/metabolismo , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/biossíntese , Estudos Prospectivos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-cbl/genética , Distribuição Aleatória , Ribonuclease III/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Neoplasias da Bexiga Urinária/metabolismo
9.
Theranostics ; 10(26): 11998-12010, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33204325

RESUMO

Rationale: Vascular abnormality stemming from the hypoxia-driven elevation of proangiogenic factors is a hallmark for many solid malignant tumors, including colorectal cancer (CRC) and its liver metastasis. We report a hypoxia-triggered liposome-supported metal-polyphenol-gene bio-nanoreactor to tune the proangiogenic factor-mediated immunotolerance and synergize the elicited tumoricidal immunity for CRC treatment. Methods: With the aid of polyphenol gallic acid, Cu2+ ion-based intracellular bio-nanoreactor was synthesized for the delivery of small interfering RNA targeting vascular endothelial growth factor and then cloaked with a hybrid liposomal membrane that harbored a hypoxia-responsive azobenzene derivative. In hypoxic tumor, the liposomal shell disintegrated, and a shrunk-size bio-nanoreactor was burst released. Intracellularly, Cu2+ from the bio-nanoreactor catalyzed a Fenton-like reaction with glutathione, which efficiently converted H2O2 to •OH and enabled a chemodynamic therapy (CDT) in tumor sites. With the alleviation of proangiogenic factor-mediated immunotolerance and high production of CDT-induced tumor-associated antigens, robust tumoricidal immunity was co-stimulated. Results: With colorectal tumor and its liver metastasis models, we determined the underlying mechanism of proangiogenic factor-mediated immunotolerance and highlighted that the liposomal bio-nanoreactor could create positive feedback among the critical players in the vascular endothelium and synergize the elicited tumoricidal immunity. Conclusion: Our work provides an alternative strategy for exerting efficient tumoricidal immunity in the proangiogenic factor-upregulated subpopulation of CRC patients and may have a wide-ranging impact on cancer immune-anti-angiogenic complementary therapy in clinics.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Nanopartículas Metálicas/administração & dosagem , Hipóxia Tumoral/efeitos dos fármacos , Inibidores da Angiogênese/administração & dosagem , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Cobre/química , Sinergismo Farmacológico , Feminino , Ácido Gálico , Humanos , Lipossomos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Nanopartículas Metálicas/química , Camundongos , Polifenóis/química , RNA Interferente Pequeno/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Nanomedicina Teranóstica/métodos , Evasão Tumoral/efeitos dos fármacos , Hipóxia Tumoral/genética , Hipóxia Tumoral/imunologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Med Sci (Paris) ; 36 Hors série n° 1: 56-60, 2020 Oct.
Artigo em Francês | MEDLINE | ID: mdl-33052096

RESUMO

Monoclonal antibody (mAb)-based immunotherapy is booming in oncology. In 2020, more than 40% of FDA (Food and Drug Administration)-approved antibodies (34 out of 84 antibodies, according to The Antibody Society) have an indication for cancer therapy. In contrast to standard chemotherapy, they demonstrate a much better safety profile for patients. Despite this, adverse side effects may occur due to the targeting of the antigen also expressed by healthy tissues. For this reason, emerging strategies aim at optimizing the antibody format and considering the particularities of the tumor microenvironment to confer a more specific action of the antibody at the tumor site.


TITLE: Stratégies de ciblage spécifique de la tumeur fondées sur les caractéristiques des antigènes tumoraux et du microenvironnement tumoral. ABSTRACT: L'immunothérapie à base d'anticorps monoclonaux (AcM) connaît un plein essor en cancérologie. En 2020, plus de 40% des anticorps approuvés par la FDA (Food and Drug Administration) (34 sur 84 anticorps, selon The Antibody Society) ont une indication pour les thérapies anti-cancéreuses. Contrairement à la chimiothérapie standard, ils démontrent un bien meilleur profil de tolérance pour les patients. Malgré cela, des effets indésirables néfastes peuvent survenir en raison du ciblage de l'antigène qui est également exprimé au niveau des tissus sains. C'est pourquoi des stratégies émergentes visent à optimiser le format des anticorps et à tenir compte des particularités du microenvironnement tumoral pour conférer une action encore plus spécifique de l'anticorps au niveau tumoral.


Assuntos
Anticorpos Monoclonais , Antígenos de Neoplasias/isolamento & purificação , Terapia de Alvo Molecular/métodos , Neoplasias/imunologia , Neoplasias/terapia , Microambiente Tumoral/imunologia , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos , Antígenos de Neoplasias/imunologia , Sistemas de Liberação de Medicamentos/métodos , Mapeamento de Epitopos/métodos , Humanos , Pró-Fármacos/uso terapêutico , Hipóxia Tumoral/imunologia
11.
Front Immunol ; 11: 1109, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32625204

RESUMO

Chimeric antigen receptor (CAR) T cell therapies have demonstrated remarkable efficacy for the treatment of hematological malignancies. However, in patients with solid tumors, objective responses to CAR-T cell therapy remain sporadic and transient. A major obstacle for CAR-T cells is the intrinsic ability of tumors to evade immune responses. Advanced solid tumors are largely composed of desmoplastic stroma and immunosuppressive modulators, and characterized by aberrant cell proliferation and vascularization, resulting in hypoxia and altered nutrient availability. To mount a curative response after infusion, CAR-T cells must infiltrate the tumor, recognize their cognate antigen and perform their effector function in this hostile tumor microenvironment, to then differentiate and persist as memory T cells that confer long-term protection. Fortunately, recent advances in synthetic biology provide a wide set of tools to genetically modify CAR-T cells to overcome some of these obstacles. In this review, we provide a comprehensive overview of the key tumor intrinsic mechanisms that prevent an effective CAR-T cell antitumor response and we discuss the most promising strategies to prevent tumor escape to CAR-T cell therapy.


Assuntos
Imunoterapia Adotiva , Neoplasias/imunologia , Neoplasias/terapia , Receptores de Antígenos Quiméricos/imunologia , Evasão Tumoral/imunologia , Microambiente Tumoral/imunologia , Animais , Matriz Extracelular/imunologia , Fibroblastos/imunologia , Humanos , Proteínas de Checkpoint Imunológico/imunologia , Linfócitos do Interstício Tumoral/imunologia , Macrófagos/imunologia , Camundongos , Modelos Imunológicos , Células Supressoras Mieloides/imunologia , Neoplasias/irrigação sanguínea , Linfócitos T Reguladores/imunologia , Hipóxia Tumoral/imunologia
12.
Clin Cancer Res ; 26(17): 4448-4453, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32571789

RESUMO

Frequently, the results generated when testing novel antitumor immunotherapies in vitro do not correlate with data collected in in vivo models and/or in clinical settings. It is our hypothesis that this discrepancy is caused by the use of in vitro conditions, such as normoxia, a two-dimensional surface, optimal growth media, and lack of cell complexity and heterogeneity. These conditions do not accurately reflect the tumor microenvironment (TME) that the tested immunotherapeutic strategies experience in vivo While there are many variables which can have an impact upon the antitumor efficacy of an immunotherapy, the immunosuppressive TME is one in which several of the conditions commonly found in vivo can be mimicked in vitro These conditions, which include hypoxia, low pH, low glucose, presence of adenosine, cell complexity and heterogeneity, as well as the three-dimensional structure of TME, can all affect immune cell-tumor cell interactions. Here, we discuss the impact that these conditions, either individually or in combination, can have on these interactions. Furthermore, we propose that performing in vitro assays under TME-like conditions improves the clinical relevance of the yielded results. This, in turn, contributes to accelerate the speed, reduce the cost, and increase efficiency of screening novel immunotherapies and eventually the development of prospective clinical trials.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Técnicas de Cultura de Células/métodos , Neoplasias/tratamento farmacológico , Microambiente Tumoral/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Técnicas de Cultura de Células/instrumentação , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Concentração de Íons de Hidrogênio , Dispositivos Lab-On-A-Chip , Neoplasias/imunologia , Esferoides Celulares , Hipóxia Tumoral/imunologia , Microambiente Tumoral/efeitos dos fármacos
13.
J Exp Clin Cancer Res ; 39(1): 75, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32357910

RESUMO

The heterogeneous tumor microenvironment (TME) is highly complex and not entirely understood. These complex configurations lead to the generation of oxygen-deprived conditions within the tumor niche, which modulate several intrinsic TME elements to promote immunosuppressive outcomes. Decoding these communications is necessary for designing effective therapeutic strategies that can effectively reduce tumor-associated chemotherapy resistance by employing the inherent potential of the immune system.While classic two-dimensional in vitro research models reveal critical hypoxia-driven biochemical cues, three-dimensional (3D) cell culture models more accurately replicate the TME-immune manifestations. In this study, we review various 3D cell culture models currently being utilized to foster an oxygen-deprived TME, those that assess the dynamics associated with TME-immune cell penetrability within the tumor-like spatial structure, and discuss state of the art 3D systems that attempt recreating hypoxia-driven TME-immune outcomes. We also highlight the importance of integrating various hallmarks, which collectively might influence the functionality of these 3D models.This review strives to supplement perspectives to the quickly-evolving discipline that endeavors to mimic tumor hypoxia and tumor-immune interactions using 3D in vitro models.


Assuntos
Técnicas de Cultura de Células/métodos , Neoplasias/imunologia , Neoplasias/patologia , Hipóxia Tumoral/imunologia , Bioengenharia/métodos , Linhagem Celular Tumoral , Humanos , Neoplasias/metabolismo , Esferoides Celulares/imunologia , Esferoides Celulares/patologia , Células Tumorais Cultivadas , Microambiente Tumoral
14.
Clin Transl Oncol ; 22(12): 2196-2205, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32445035

RESUMO

Radiotherapy is the treatment of choice for many cancer patients. Residual tumor leads to local recurrence after a period of an equilibrium created between proliferating, quiescent and dying cancer cells. The tumor microenvironment is a main obstacle for the efficacy of radiotherapy, as impaired blood flow leads to hypoxia, acidity and reduced accessibility of radiosensitizers. Eradication of remnant disease is an intractable clinical quest. After more than a century of research, anti-tumor immunity has gained a dominant position in oncology research and therapy. Immune cells play a significant role in the eradication of tumors during and after the completion of radiotherapy. The tumor equilibrium reached in the irradiated tumor may shift towards cancer cell eradication if the immune response is appropriately modulated. In the modern immunotherapy era, clinical trials are urged to standardize immunotherapy schemes that could be safely applied to improve clearance of the post-radiotherapy remnant disease.


Assuntos
Imunidade , Neoplasias/radioterapia , Microambiente Tumoral , Humanos , Imunoterapia , Recidiva Local de Neoplasia , Neoplasia Residual/imunologia , Neoplasia Residual/patologia , Neoplasias/imunologia , Neoplasias/patologia , Neovascularização Patológica , Tolerância a Radiação , Radiossensibilizantes/uso terapêutico , Hipóxia Tumoral/imunologia , Microambiente Tumoral/imunologia
15.
Cancer Lett ; 487: 74-84, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32470491

RESUMO

Hypoxia is a universal feature of solid cancers caused by a mismatch between cellular oxygen supply and consumption. To meet the increased demand for oxygen, hypoxic cancer cells (CCs) induce a multifaceted process known as angiogenesis, wherein new vessels are formed by the sprouting of pre-existing ones. In addition to providing oxygen for growth and an exit route for dissemination, angiogenic vessels and factors are co-opted by CCs to enable the generation of an immunotolerant, hypoxic tumor microenvironment, leading to therapeutic failure and mortality. In this review, we discuss how hypoxia-inducible factors (HIFs), the mechanistic target of rapamycin (mTOR), and the unfolded protein response (UPR) control angiogenic factors serving both vascular and immunomodulatory functions in the tumor microenvironment. Possible therapeutic strategies, wherein targeting oxygen sensing might enhance anti-angiogenic and immunologically-mediated anti-cancer responses, are suggested.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias/genética , Serina-Treonina Quinases TOR/genética , Microambiente Tumoral/genética , Resposta a Proteínas não Dobradas/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/patologia , Regulação Neoplásica da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/imunologia , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Hipóxia Tumoral/genética , Hipóxia Tumoral/imunologia , Microambiente Tumoral/imunologia , Resposta a Proteínas não Dobradas/imunologia
16.
Semin Cancer Biol ; 65: 140-154, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31927131

RESUMO

Immunotherapy is poised to become an increasingly utilized therapy in the treatment of cancer. However, several abnormalities in the tumor microenvironment (TME) that can thwart the efficacy of immunotherapies have been established. Microenvironmental hypoxia is a determining factor in shaping aggressiveness, metastatic potential and treatment resistance of solid tumors. The characterization of this phenomenon could prove beneficial for determining a patient's treatment path and for the introduction of novel targetable factors that can enhance therapeutic outcome. Indeed, the ablation of hypoxia has the potential to sensitize tumors to immunotherapy by metabolically remodeling their microenvironment. In this review, we discuss the intrinsic contributions of hypoxia to cellular plasticity, heterogeneity, stemness and genetic instability in the context of immune escape. In addition, we will shed light on how managing hypoxia can ameliorate response to immunotherapy and how integrating hypoxia gene signatures could play a role in this pursuit.


Assuntos
Imunoterapia , Neoplasias/terapia , Hipóxia Tumoral/imunologia , Microambiente Tumoral/imunologia , Humanos , Neoplasias/imunologia , Estresse Fisiológico/genética , Estresse Fisiológico/imunologia
17.
Front Immunol ; 11: 613114, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33552076

RESUMO

The environmental and metabolic pressures in the tumor microenvironment (TME) play a key role in molding tumor development by impacting the stromal and immune cell fractions, TME composition and activation. Hypoxia triggers a cascade of events that promote tumor growth, enhance resistance to the anti-tumor immune response and instigate tumor angiogenesis. During growth, the developing angiogenesis is pathological and gives rise to a haphazardly shaped and leaky tumor vasculature with abnormal properties. Accordingly, aberrantly vascularized TME induces immunosuppression and maintains a continuous hypoxic state. Normalizing the tumor vasculature to restore its vascular integrity, should hence enhance tumor perfusion, relieving hypoxia, and reshaping anti-tumor immunity. Emerging vascular normalization strategies have a great potential in achieving a stable normalization, resulting in mature and functional blood vessels that alleviate tumor hypoxia. Biomarkers enabling the detection and monitoring of tumor hypoxia could be highly advantageous in aiding the translation of novel normalization strategies to clinical application, alone, or in combination with other treatment modalities, such as immunotherapy.


Assuntos
Biomarcadores/metabolismo , Tolerância Imunológica/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Neovascularização Patológica/imunologia , Hipóxia Tumoral/imunologia , Animais , Humanos , Imunoterapia/métodos , Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Microambiente Tumoral/imunologia
19.
Cells ; 8(2)2019 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-30708988

RESUMO

The metabolic reprogramming of tumor cells and immune escape are two major hallmarks of cancer cells. The metabolic changes that occur during tumorigenesis, enabling survival and proliferation, are described for both solid and hematological malignancies. Concurrently, tumor cells have deployed mechanisms to escape immune cell recognition and destruction. Additionally, therapeutic blocking of tumor-mediated immunosuppression has proven to have an unprecedented positive impact in clinical oncology. Increased evidence suggests that cancer metabolism not only plays a crucial role in cancer signaling for sustaining tumorigenesis and survival, but also has wider implications in the regulation of antitumor immune signaling through both the release of signaling molecules and the expression of immune membrane ligands. Here, we review these molecular events to highlight the contribution of cancer cell metabolic reprogramming on the shaping of the antitumor immune response.


Assuntos
Imunidade , Neoplasias/imunologia , Neoplasias/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Modelos Biológicos , Hipóxia Tumoral/imunologia
20.
Cancer Res ; 79(4): 795-806, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30610087

RESUMO

Tumor hypoxia and aerobic glycolysis are well-known resistance factors for anticancer therapies. Here, we demonstrate that tumor-associated macrophages (TAM) enhance tumor hypoxia and aerobic glycolysis in mice subcutaneous tumors and in patients with non-small cell lung cancer (NSCLC). We found a strong correlation between CD68 TAM immunostaining and PET 18fluoro-deoxyglucose (FDG) uptake in 98 matched tumors of patients with NSCLC. We also observed a significant correlation between CD68 and glycolytic gene signatures in 513 patients with NSCLC from The Cancer Genome Atlas database. TAM secreted TNFα to promote tumor cell glycolysis, whereas increased AMP-activated protein kinase and peroxisome proliferator-activated receptor gamma coactivator 1-alpha in TAM facilitated tumor hypoxia. Depletion of TAM by clodronate was sufficient to abrogate aerobic glycolysis and tumor hypoxia, thereby improving tumor response to anticancer therapies. TAM depletion led to a significant increase in programmed death-ligand 1 (PD-L1) expression in aerobic cancer cells as well as T-cell infiltration in tumors, resulting in antitumor efficacy by PD-L1 antibodies, which were otherwise completely ineffective. These data suggest that TAM can significantly alter tumor metabolism, further complicating tumor response to anticancer therapies, including immunotherapy. SIGNIFICANCE: These findings show that tumor-associated macrophages can significantly modulate tumor metabolism, hindering the efficacy of anticancer therapies, including anti-PD-L1 immunotherapy.


Assuntos
Antígeno B7-H1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Glicólise , Neoplasias Pulmonares/patologia , Macrófagos/imunologia , Hipóxia Tumoral/imunologia , Animais , Antígeno B7-H1/imunologia , Carcinoma Pulmonar de Células não Pequenas/etiologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Prognóstico , Linfócitos T/imunologia , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA