Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Front Immunol ; 15: 1373497, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38720889

RESUMO

Introduction: Intraoperative radiation therapy (IORT) delivers a single accelerated radiation dose to the breast tumor bed during breast-conserving surgery (BCS). The synergistic biologic effects of simultaneous surgery and radiation remain unclear. This study explores the cellular and molecular changes induced by IORT in the tumor microenvironment and its impact on the immune response modulation. Methods: Patients with hormone receptor (HR)-positive/HER2-negative, ductal carcinoma in situ (DCIS), or early-stage invasive breast carcinoma undergoing BCS with margin re-excision were included. Histopathological evaluation and RNA-sequencing in the re-excision tissue were compared between patients with IORT (n=11) vs. non-IORT (n=11). Results: Squamous metaplasia with atypia was exclusively identified in IORT specimens (63.6%, p=0.004), mimicking DCIS. We then identified 1,662 differentially expressed genes (875 upregulated and 787 downregulated) between IORT and non-IORT samples. Gene ontology analyses showed that IORT was associated with the enrichment of several immune response pathways, such as inflammatory response, granulocyte activation, and T-cell activation (p<0.001). When only considering normal tissue from both cohorts, IORT was associated with intrinsic apoptotic signaling, response to gamma radiation, and positive regulation of programmed cell death (p<0.001). Using the xCell algorithm, we inferred a higher abundance of γδ T-cells, dendritic cells, and monocytes in the IORT samples. Conclusion: IORT induces histological changes, including squamous metaplasia with atypia, and elicits molecular alterations associated with immune response and intrinsic apoptotic pathways. The increased abundance of immune-related components in breast tissue exposed to IORT suggests a potential shift towards active immunogenicity, particularly immune-desert tumors like HR-positive/HER2-negative breast cancer.


Assuntos
Neoplasias da Mama , Imunomodulação , Cuidados Intraoperatórios , Mastectomia Segmentar , Microambiente Tumoral , Humanos , Feminino , Neoplasias da Mama/radioterapia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Pessoa de Meia-Idade , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação , Imunomodulação/efeitos da radiação , Idoso , Adulto , Terapia Combinada
2.
Front Immunol ; 12: 754436, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34733287

RESUMO

Cancer patients with low or absent pre-existing anti-tumour immunity ("cold" tumours) respond poorly to treatment with immune checkpoint inhibitors (ICPI). In order to render these patients susceptible to ICPI, initiation of de novo tumour-targeted immune responses is required. This involves triggering of inflammatory signalling, innate immune activation including recruitment and stimulation of dendritic cells (DCs), and ultimately priming of tumour-specific T cells. The ability of tumour localised therapies to trigger these pathways and act as in situ tumour vaccines is being increasingly explored, with the aspiration of developing combination strategies with ICPI that could generate long-lasting responses. In this effort, it is crucial to consider how therapy-induced changes in the tumour microenvironment (TME) act both as immune stimulants but also, in some cases, exacerbate immune resistance mechanisms. Increasingly refined immune monitoring in pre-clinical studies and analysis of on-treatment biopsies from clinical trials have provided insight into therapy-induced biomarkers of response, as well as actionable targets for optimal synergy between localised therapies and ICB. Here, we review studies on the immunomodulatory effects of novel and experimental localised therapies, as well as the re-evaluation of established therapies, such as radiotherapy, as immune adjuvants with a focus on ICPI combinations.


Assuntos
Inibidores de Checkpoint Imunológico/uso terapêutico , Imunomodulação , Imunoterapia/métodos , Neoplasias/terapia , Adjuvantes Imunológicos/uso terapêutico , Animais , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/efeitos da radiação , Vacinas Anticâncer/uso terapêutico , Terapia Combinada , Células Dendríticas/imunologia , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Herpesvirus Humano 1/fisiologia , Humanos , Imunidade Inata/efeitos dos fármacos , Agentes de Imunomodulação/uso terapêutico , Imunomodulação/efeitos dos fármacos , Imunomodulação/efeitos da radiação , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/radioterapia , Terapia Viral Oncolítica , Subpopulações de Linfócitos T/imunologia , Microambiente Tumoral/imunologia
3.
J Photochem Photobiol B ; 221: 112247, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34175580

RESUMO

A need exists for further research elucidating the benefits of environmentally safe photoprotective agents against ultraviolet (UV) exposure, and plant extracts represent a human-friendly alternative formulation. This study was designed to evaluate the potential use of Bellis perennis extract (BPE), from the Asteraceae family, known as the common daisy or the English daisy, in cosmeceuticals as a photoprotective factor, using an in vitro model of UVA-induced keratinocyte damage. Human skin keratinocytes (HaCaT cell line) were incubated with BPE at 0.01, 0.1, or 1% in Dulbecco's Modified Eagle Medium (DMEM), and after 15 min they were submitted to UVA radiation at 5, 10, and 15 J/cm2 doses, respectively. For comparative purposes, Polypodium leucotomos extract (PLE), known as the fern, was used as a positive control in assessing the photoprotective effect. After 24 h of UVA exposure, cell viability (MTT and LDH assays), levels of cleaved caspase-3, cyclooxygenase-2, IL-6, reactive oxygen species (ROS) and antioxidant enzyme (catalase, SOD, and glutathione peroxidase) activity were determined. UVA radiation at 5, 10, and 15 J/cm2 doses reduced cell viability to 63%, 43%, and 23%, respectively; we selected 10 J/cm2 for our purposes. After 24 h of UVA exposure, treatment with 1% BPE and 1% PLE significantly recovered cell viability (p < 0.05). Furthermore, treatment was associated with lower cleaved caspase-3 and ROS levels, higher catalase activity, and lower IL-6 levels in the treated UVA keratinocytes compared with the untreated UVA group (p < 0.01). Our results demonstrate photoprotective and immunomodulatory effects of BPE in skin keratinocytes and support its use as a bioactive agent in cosmetic formulations to prevent skin damage caused by exposure to the UV light.


Assuntos
Asteraceae/química , Imunomodulação/efeitos dos fármacos , Extratos Vegetais/farmacologia , Protetores contra Radiação/farmacologia , Raios Ultravioleta , Asteraceae/metabolismo , Caspase 3/metabolismo , Catalase/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Humanos , Imunomodulação/efeitos da radiação , Queratinócitos/citologia , Queratinócitos/metabolismo , Extratos Vegetais/química , Protetores contra Radiação/química , Espécies Reativas de Oxigênio/metabolismo
4.
Front Immunol ; 12: 617365, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33936033

RESUMO

In situ tumor ablation techniques, like radiotherapy, cryo- and heat-based thermal ablation are successfully applied in oncology for local destruction of tumor masses. Although diverse in technology and mechanism of inducing cell death, ablative techniques share one key feature: they generate tumor debris which remains in situ. This tumor debris functions as an unbiased source of tumor antigens available to the immune system and has led to the concept of in situ cancer vaccination. Most studies, however, report generally modest tumor-directed immune responses following local tumor ablation as stand-alone treatment. Tumors have evolved mechanisms to create an immunosuppressive tumor microenvironment (TME), parts of which may admix with the antigen depot. Provision of immune stimuli, as well as approaches that counteract the immunosuppressive TME, have shown to be key to boost ablation-induced anti-tumor immunity. Recent advances in protein engineering have yielded novel multifunctional antibody formats. These multifunctional antibodies can provide a combination of distinct effector functions or allow for delivery of immunomodulators specifically to the relevant locations, thereby mitigating potential toxic side effects. This review provides an update on immune activation strategies that have been tested to act in concert with tumor debris to achieve in situ cancer vaccination. We further provide a rationale for multifunctional antibody formats to be applied together with in situ ablation to boost anti-tumor immunity for local and systemic tumor control.


Assuntos
Técnicas de Ablação , Imunomodulação/efeitos dos fármacos , Imunomodulação/efeitos da radiação , Neoplasias/imunologia , Neoplasias/terapia , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos de Neoplasias/imunologia , Terapia Combinada , Humanos , Terapia de Imunossupressão , Imunoterapia/métodos , Neoplasias/patologia , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação
5.
J Immunol Res ; 2021: 6664453, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33628851

RESUMO

BACKGROUND: The immune mechanisms underlying low-intensity ultrasound- (LIUS-) mediated suppression of inflammation and tumorigenesis remain poorly determined. METHODS: We used microarray datasets from the NCBI GEO DataSet repository and conducted comprehensive data-mining analyses, where we examined the gene expression of 1376 innate immune regulators (innatome genes (IGs) in cells treated with LIUS. RESULTS: We made the following findings: (1) LIUS upregulates proinflammatory IGs and downregulates metastasis genes in cancer cells, and LIUS upregulates adaptive immunity pathways but inhibits danger-sensing and inflammation pathways and promote tolerogenic differentiation in bone marrow (BM) cells. (2) LIUS upregulates IGs encoded for proteins localized in the cytoplasm, extracellular space, and others, but downregulates IG proteins localized in nuclear and plasma membranes, and LIUS downregulates phosphatases. (3) LIUS-modulated IGs act partially via several important pathways of reactive oxygen species (ROS), reverse signaling of immune checkpoint receptors B7-H4 and BTNL2, inflammatory cytokines, and static or oscillatory shear stress and heat generation, among which ROS is a dominant mechanism. (4) LIUS upregulates trained immunity enzymes in lymphoma cells and downregulates trained immunity enzymes and presumably establishes trained tolerance in BM cells. (5) LIUS modulates chromatin long-range interactions to differentially regulate IGs expression in cancer cells and noncancer cells. CONCLUSIONS: Our analysis suggests novel molecular mechanisms that are utilized by LIUS to induce tumor suppression and inflammation inhibition. Our findings may lead to development of new treatment protocols for cancers and chronic inflammation.


Assuntos
Citocinas/metabolismo , Proteínas de Checkpoint Imunológico/metabolismo , Neoplasias/etiologia , Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Evasão Tumoral/imunologia , Ondas Ultrassônicas , Imunidade Adaptativa , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Hipertermia Induzida/métodos , Proteínas de Checkpoint Imunológico/genética , Imunidade Inata , Imunomodulação/efeitos da radiação , Modelos Biológicos , Neoplasias/patologia , Neoplasias/terapia , Transdução de Sinais/efeitos da radiação
7.
Int J Radiat Oncol Biol Phys ; 109(4): 867-879, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33340603

RESUMO

PURPOSE: Phase 1 clinical trials have established low-dose, whole-lung radiation therapy (LD-RT) as safe for patients with coronavirus disease 2019 (COVID-19)-related pneumonia. By focally dampening cytokine hyperactivation, LD-RT may improve disease outcomes through immunomodulation. METHODS AND MATERIALS: Patients with COVID-19-related pneumonia were treated with 1.5 Gy whole-lung LD-RT, followed for 28 days or until hospital discharge, and compared with age- and comorbidity-matched controls meeting identical disease severity criteria. Eligible patients were hospitalized, severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2) positive, had radiographic consolidations, and required supplemental oxygen but had not rapidly declined on admission or before drug therapy or LD-RT. Efficacy endpoints were time to clinical recovery, radiographic improvement, and biomarker response. RESULTS: Ten patients received whole-lung LD-RT between April 24 and May 24, 2020 and were compared with 10 control patients blindly matched by age and comorbidity. Six controls received COVID-19 drug therapies. Median time to clinical recovery was 12 days in the control cohort compared with 3 days in the LD-RT cohort (hazard ratio 2.9, P = .05). Median time to hospital discharge (20 vs 12 days, P = .19) and intubation rates (40% vs 10%, P = .12) in the control and LD-RT cohorts were compared. Median time from admission to recovery was 10 versus 13 days (P = .13). Hospital duration average was 19 versus 22.6 days (P = .53). Average hospital days on supplemental oxygen of any duration was 13.1 versus 14.7 days (P = .69). Average days with a documented fever was 1 versus 4.3 days (P = .12). Twenty-eight-day overall survival was 90% for both cohorts. The LD-RT cohort trended toward superior rates of improved radiographs (P = .12) and delirium (P < .01). Statistically significant reductions were observed in numerous hematologic, cardiac, hepatic, and inflammatory markers. CONCLUSIONS: A prospective cohort of predominantly elderly hospitalized patients with COVID-19-related pneumonia were recovered to room air quicker than age- and comorbidity-matched controls, with trending or significant improvements in delirium, radiographs, and biomarkers, and no significant acute toxicity. Low-dose, whole-lung radiation for patients with COVID-19-related pneumonia appears safe and may be an effective immunomodulatory treatment. Larger prospective randomized trials are needed to define the efficacy of LD-RT for COVID-19.


Assuntos
COVID-19/imunologia , COVID-19/radioterapia , Imunomodulação/efeitos da radiação , Pulmão/efeitos da radiação , Doses de Radiação , Adulto , Idoso , Idoso de 80 Anos ou mais , COVID-19/sangue , COVID-19/diagnóstico por imagem , Feminino , Hospitalização , Humanos , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Masculino , Pessoa de Meia-Idade , Dosagem Radioterapêutica , Segurança , Tomografia Computadorizada por Raios X , Resultado do Tratamento
8.
Commun Biol ; 3(1): 783, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33335270

RESUMO

Thermal ablation is a standard therapy for patients with hepatocellular carcinoma (HCC). Contemporary ablation devices are imperfect, as they lack tumor specificity. An ideal ablation modality would generate thermal energy only within tumoral tissue. Furthermore, as hyperthermia is known to influence tumor immunity, such a tumor-specific ablation modality may have the ability to favorably modulate the tumor immune landscape. Here we show a clinically relevant thermal ablation modality that generates tumor-specific hyperthermia, termed molecularly targeted photothermal ablation (MTPA), that is based upon the excellent localization of indocyanine green to HCC. In a syngeneic rat model, we demonstrate the tumor-specific hyperthermia generated by MTPA. We also show through spatial and transcriptomic profiling techniques that MTPA favorably modulates the intratumoral myeloid population towards tumor immunogenicity and diminishes the systemic release of oncogenic cytokines relative to conventional ablation modalities.


Assuntos
Carcinoma Hepatocelular/etiologia , Imunomodulação/efeitos da radiação , Neoplasias Hepáticas/etiologia , Terapia Fototérmica/métodos , Animais , Biomarcadores Tumorais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Citocinas/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Expressão Gênica , Hipertermia Induzida , Imunomodulação/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Terapia de Alvo Molecular , Ratos , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação
9.
Sci Rep ; 10(1): 21517, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33299018

RESUMO

While for photon radiation hypofractionation has been reported to induce enhanced immunomodulatory effects, little is known about the immunomodulatory potential of carbon ion radiotherapy (CIRT). We thus compared the radio-immunogenic effects of photon and carbon ion irradiation on two murine cancer cell lines of different tumor entities. We first calculated the biological equivalent doses of carbon ions corresponding to photon doses of 1, 3, 5, and 10 Gy of the murine breast cancer cell line EO771 and the OVA-expressing pancreatic cancer cell line PDA30364/OVA by clonogenic survival assays. We compared the potential of photon and carbon ion radiation to induce cell cycle arrest, altered surface expression of immunomodulatory molecules and changes in the susceptibility of cancer cells to cytotoxic T cell (CTL) mediated killing. Irradiation induced a dose-dependent G2/M arrest in both cell lines irrespective from the irradiation source applied. Likewise, surface expression of the immunomodulatory molecules PD-L1, CD73, H2-Db and H2-Kb was increased in a dose-dependent manner. Both radiation modalities enhanced the susceptibility of tumor cells to CTL lysis, which was more pronounced in EO771/Luci/OVA cells than in PDA30364/OVA cells. Overall, compared to photon radiation, the effects of carbon ion radiation appeared to be enhanced at higher dose range for EO771 cells and extenuated at lower dose range for PDA30364/OVA cells. Our data show for the first time that equivalent doses of carbon ion and photon irradiation exert similar immunomodulating effects on the cell lines of both tumor entities, highlighted by an enhanced susceptibility to CTL mediated cytolysis in vitro.


Assuntos
Radioterapia com Íons Pesados/métodos , Imunomodulação/efeitos da radiação , Fótons/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Carbono/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Humanos , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas
10.
Nat Immunol ; 21(6): 649-659, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32424359

RESUMO

Efficient generation of germinal center (GC) responses requires directed movement of B cells between distinct microenvironments underpinned by specialized B cell-interacting reticular cells (BRCs). How BRCs are reprogrammed to cater to the developing GC remains unclear, and studying this process is largely hindered by incomplete resolution of the cellular composition of the B cell follicle. Here we used genetic targeting of Cxcl13-expressing cells to define the molecular identity of the BRC landscape. Single-cell transcriptomic analysis revealed that BRC subset specification was predetermined in the primary B cell follicle. Further topological remodeling of light and dark zone follicular dendritic cells required CXCL12-dependent crosstalk with B cells and dictated GC output by retaining B cells in the follicle and steering their interaction with follicular helper T cells. Together, our results reveal that poised BRC-defined microenvironments establish a feed-forward system that determines the efficacy of the GC reaction.


Assuntos
Escuridão , Células Dendríticas Foliculares/imunologia , Células Dendríticas Foliculares/metabolismo , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Imunomodulação/efeitos da radiação , Luz , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores , Comunicação Celular , Quimiocina CXCL12/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Análise de Célula Única , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
11.
Semin Radiat Oncol ; 30(2): 181-186, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32381297

RESUMO

Recent preclinical and clinical studies have elucidated mechanisms whereby radiation therapy influences the anti-tumor immune response. Immunogenic cell death and phenotypic changes in tumor cells surviving radiation may underlie this effect and contribute to the capacity of radiation to elicit an in situ tumor vaccine effect. In situ vaccination is a therapeutic strategy that seeks to convert a patient's own tumor into a source of enhanced antigen recognition for the purpose of augmenting a systemic anti-tumor immune response. Capitalizing on the in situ vaccine effect of radiation, several groups have demonstrated anti-tumor efficacy in preclinical models by combining radiation with immune checkpoint blockade. Local delivery of immune adjuvants and/or immune stimulatory cytokines via direct injection into the radiated tumor microenvironment may further increase the in situ vaccine capacity of radiation therapy. However, recent studies suggest that in some contexts this effect is antagonized by the presence of distant untreated sites of disease that may dampen the systemic immune response generated by in situ vaccination through a phenomenon termed concomitant immune tolerance. Concomitant immune tolerance may be overcome by delivering radiation to all sites of metastatic disease, however this is often not possible to safely achieve using external beam radiation therapy without considerable risk of lymphopenia that would negate the immune effects of in situ vaccination. For patients with widespread metastatic disease, alternative strategies may include systemic treatment with targeted radionuclide therapies alone or in combination with an external beam radiation therapy-based in situ vaccine approach.


Assuntos
Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/imunologia , Terapia Combinada , Citocinas/imunologia , Citocinas/farmacologia , Modelos Animais de Doenças , Humanos , Inibidores de Checkpoint Imunológico/imunologia , Inibidores de Checkpoint Imunológico/farmacologia , Imunomodulação/imunologia , Imunomodulação/efeitos da radiação , Hipofracionamento da Dose de Radiação , Vacinação
12.
Eur J Immunol ; 50(5): 725-735, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32012249

RESUMO

Extracorporeal photochemotherapy (ECP) that takes advantage of the immunomodulatory effects of UV light has been extensively used for many years for the treatment of several T cell-mediated diseases, including graft-versus-host disease (GvHD) and systemic scleroderma. Immune mechanisms that lead to the establishment of T cell tolerance in ECP-treated patients remain poorly known. In this study, we have tested the effect of UV/psoralen-treated BM-derived dendritic cells, referred to as ECP-BMDCs on the outcome of an antigen-specific T cell-mediated reaction, that is, contact hypersensitivity (CHS), which is mediated by CD8+ effector T cells (CD8+ Teff ). The intravenous (i.v.) injection of antigen-pulsed ECP-BMDCs in recipient C57BL/6 mice induced specific CD8+ T cells endowed with immunomodulatory properties (referred to as CD8+ TECP ), which prevented the priming of CD8+ Teff and the development of CHS, independently of conventional CD4+ regulatory T cells. CD8+ TECP mediated tolerance by inhibiting the migration and functions of skin DC and subsequently the priming of CD8+ Teff . CD8+ TECP displayed none of the phenotypes of the usual CD8+ T regulatory cells described so far. Our results reveal an underestimated participation of CD8+ T cells to ECP-induced immunomodulation that could explain the therapeutic effects of ECP in T cell-mediated diseases.


Assuntos
Células Dendríticas/imunologia , Dermatite de Contato/terapia , Tolerância Imunológica , Imunomodulação/efeitos da radiação , Linfócitos T Citotóxicos/efeitos da radiação , Linfócitos T Reguladores/efeitos da radiação , Alérgenos/administração & dosagem , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células Dendríticas/citologia , Células Dendríticas/transplante , Dermatite de Contato/imunologia , Dermatite de Contato/fisiopatologia , Dinitrofluorbenzeno/administração & dosagem , Modelos Animais de Doenças , Feminino , Ficusina/administração & dosagem , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fotoferese/métodos , Fármacos Fotossensibilizantes/administração & dosagem , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Raios Ultravioleta
13.
Cancer Immunol Immunother ; 69(2): 293-306, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31953578

RESUMO

Cancer immunotherapies are promising treatments for many forms of cancer. Nevertheless, the response rates to, e.g., immune checkpoint inhibitors (ICI), are still in low double-digit percentage. This calls for further therapy optimization that should take into account combination of immunotherapies with classical tumor therapies such as radiotherapy. By designing multimodal approaches, immune modulatory properties of certain radiation schemes, additional immune modulation by immunotherapy with ICI and hyperthermia, as well as patient stratification based on genetic and immune constitutions have to be considered. In this context, both the tumor and its microenvironment including cells of the innate and adaptive immune system have to be viewed in synopsis. Knowledge of immune activation and immune suppression by radiation is the basis for well-elaborated addition of certain immunotherapies. In this review, the focus is set on additional immune stimulation by hyperthermia and restoration of an immune response by ICI. The impact of radiation dose and fractionation on immune modulation in multimodal settings has to be considered, as the dynamics of the immune response and the timing between radiotherapy and immunotherapy. Another big challenge is the patient stratification that should be based on matrices of biomarkers, taking into account genetics, proteomics, radiomics, and "immunomics". One key aim is to turn immunological "cold" tumors into "hot" tumors, and to eliminate barriers of immune-suppressed or immune-excluded tumors. Comprehensive knowledge of immune alterations induced by radiation and immunotherapy when being applied together should be utilized for patient-adapted treatment planning and testing of innovative tumor therapies within clinical trials.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Desenho de Fármacos , Imunomodulação/efeitos dos fármacos , Neoplasias/etiologia , Neoplasias/terapia , Animais , Antineoplásicos Imunológicos/farmacologia , Biomarcadores Tumorais , Terapia Combinada , Humanos , Hipertermia Induzida/métodos , Imunidade , Fatores Imunológicos/farmacologia , Imunomodulação/efeitos da radiação , Imunoterapia , Neoplasias/patologia , Projetos de Pesquisa , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação
14.
Oncogene ; 39(1): 187-203, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31467431

RESUMO

Radiotherapy induces immune-related responses in cancer patients by various mechanisms. Here, we investigate the immunomodulatory role of tumor-derived microparticles (TMPs)-extracellular vesicles shed from tumor cells-following radiotherapy. We demonstrate that breast carcinoma cells exposed to radiation shed TMPs containing elevated levels of immune-modulating proteins, one of which is programmed death-ligand 1 (PD-L1). These TMPs inhibit cytotoxic T lymphocyte (CTL) activity both in vitro and in vivo, and thus promote tumor growth. Evidently, adoptive transfer of CTLs pre-cultured with TMPs from irradiated breast carcinoma cells increases tumor growth rates in mice recipients in comparison with control mice receiving CTLs pre-cultured with TMPs from untreated tumor cells. In addition, blocking the PD-1-PD-L1 axis, either genetically or pharmacologically, partially alleviates TMP-mediated inhibition of CTL activity, suggesting that the immunomodulatory effects of TMPs in response to radiotherapy is mediated, in part, by PD-L1. Overall, our findings provide mechanistic insights into the tumor immune surveillance state in response to radiotherapy and suggest a therapeutic synergy between radiotherapy and immune checkpoint inhibitors.


Assuntos
Antígeno B7-H1/genética , Neoplasias da Mama/radioterapia , Micropartículas Derivadas de Células/imunologia , Imunomodulação/imunologia , Animais , Antígeno B7-H1/imunologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Micropartículas Derivadas de Células/genética , Micropartículas Derivadas de Células/efeitos da radiação , Feminino , Xenoenxertos , Humanos , Evasão da Resposta Imune/imunologia , Evasão da Resposta Imune/efeitos da radiação , Imunomodulação/efeitos da radiação , Camundongos , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Transdução de Sinais/efeitos da radiação , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/efeitos da radiação
15.
Clin Cancer Res ; 26(4): 910-921, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31757878

RESUMO

PURPOSE: Radiotherapy (RT) is frequently used for local control of solid tumors using equal dose per fraction. Recently, single high-dose radiation has been used for ablation of solid tumors. In this report, we provide a novel immunological basis for radiation dose fractionation consisting of a single high-dose radiotherapy, followed by postablation modulation (PAM) with four daily low-dose fractions (22 Gy + 0.5 Gy × 4) to reprogram the tumor microenvironment by diminishing immune suppression, enabling infiltration of effector cells and increasing efficacy of tumor control. EXPERIMENTAL DESIGN: Palpable 3LL and 4T1 tumors in C57Bl/6 and Balb/c mice were irradiated with the Small-Animal Radiation Research Platform irradiator, and tumor growth and survival were monitored. Immunomodulation of tumor and immune cells in vitro and in vivo characterization of tumor-infiltrating immune effector cells were performed by FACS. For systemic application of PAM-RT, whole-lung irradiation was administered in 4T1-bearing Balb/c mice. RESULTS: We report significant tumor growth delays and increased survival in 3LL tumor-bearing mice with PAM. Primary tumor PAM-RT increased infiltration of immune effector cells and decreased Treg in irradiated tumors and secondary lymphoid organs. In a model of murine metastatic breast cancer (4T1), we demonstrated that systemic PAM-RT to the whole lung, 12 days after primary tumor ablative radiotherapy, increased survival with suppression of pulmonary metastases. CONCLUSIONS: We provide a novel immunologic basis for radiation dose fractionation consisting of a single high dose of radiotherapy followed by daily low-dose PAM-RT fractionation to improve the immunogenic potential of ablative radiotherapy.


Assuntos
Carcinoma Pulmonar de Lewis/radioterapia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Mamárias Experimentais/radioterapia , Microambiente Tumoral/imunologia , Técnicas de Ablação/métodos , Animais , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/cirurgia , Linhagem Celular Tumoral , Fracionamento da Dose de Radiação , Feminino , Imunomodulação/efeitos da radiação , Linfócitos do Interstício Tumoral/efeitos da radiação , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/cirurgia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Taxa de Sobrevida , Microambiente Tumoral/efeitos da radiação
16.
PLoS One ; 14(9): e0222022, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31479484

RESUMO

In the United States, breast cancer is one of the most common and the second leading cause of cancer-related death in women. Treatment modalities for mammary tumor are surgical removal of the tumor tissue followed by either chemotherapy or radiotherapy or both. Radiation therapy is a whole body irradiation regimen that suppresses the immune system leaving hosts susceptible to infection or secondary tumors. Boron neutron capture therapy (BNCT) in that regard is more selective, the cells that are mostly affected are those that are loaded with 109 or more 10B atoms. Previously, we have described that liposomal encapsulation of boron-rich compounds such as TAC and MAC deliver a high payload to the tumor tissue when injected intravenously. Here we report that liposome-mediated boron delivery to the tumor is inversely proportional to the size of the murine mammary (EMT-6) tumors. The plausible reason for the inverse ratio of boron and EMT-6 tumor size is the necrosis in these tumors, which is more prominent in the large tumors. The large tumors also have receding blood vessels contributing further to poor boron delivery to these tumors. We next report that the presence of boron in blood is essential for the effects of BNCT on EMT-6 tumor inhibition as direct injection of boron-rich liposomes did not provide any added advantage in inhibition of EMT-6 tumor in BALB/c mice following irradiation despite having a significantly higher amount of boron in the tumor tissue. BNCT reaction in PBMCs resulted in the modification of these cells to anti-tumor phenotype. In this study, we report the immunomodulatory effects of BNCT when boron-rich compounds are delivered systemically.


Assuntos
Terapia por Captura de Nêutron de Boro/métodos , Imunomodulação/efeitos da radiação , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/radioterapia , Animais , Boro/administração & dosagem , Boro/sangue , Boro/farmacocinética , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Humanos , Isótopos/administração & dosagem , Isótopos/sangue , Isótopos/farmacocinética , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/efeitos da radiação , Lipossomos , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Necrose , Distribuição Tecidual
17.
Int J Biol Macromol ; 140: 505-514, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31437508

RESUMO

In order to investigate the impacts of ultrasound-assisted extraction (UAE) on Flammulina velutipes polysaccharides (FVPs), the differences between FVPs extracted by ultrasound-assisted extraction (FVPU) and FVPs extracted by hot water extraction (FVPH) were compared in terms of yield, primary compositions, surface microstructure, helix-coil transition structure, molecular weight distribution, antioxidant activity, and bidirectional immunomodulatory activity. Results indicated that UAE changed the above properties of FVPs. Compared with FVPH, higher yield, protein content, and uronic acid content but lower polysaccharide and polyphenol contents were observed in FVPU. UAE changed the surface microstructure, destroyed the triple helix structure, and increased the proportion of low molecular weight polysaccharide components of FVPU. Compared with FVPH, FVPU showed a stronger reducing power and scavenging activities on DPPH radical, hydroxyl radical, and superoxide anion radical. FVPU was a better inhibitor of inflammation compared with FVPH. However, FVPH had a better immunity enhancing effect compared with FVPU. These results were attributed to the cavitation effect of ultrasonic waves on the structure of polysaccharides during the extraction process of UAE. These findings suggested that UAE was an efficient and environmentally friendly method to produce new polysaccharides from F. velutipes for the development of functional foods or nutraceuticals.


Assuntos
Antioxidantes/química , Flammulina/química , Polissacarídeos/química , Animais , Antioxidantes/farmacologia , Fracionamento Químico , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Radical Hidroxila/química , Imunomodulação/efeitos dos fármacos , Imunomodulação/efeitos da radiação , Inflamação/tratamento farmacológico , Inflamação/imunologia , Camundongos , Polissacarídeos/isolamento & purificação , Polissacarídeos/farmacologia , Células RAW 264.7 , Superóxidos/química , Temperatura , Ondas Ultrassônicas , Água/química
18.
Radiother Oncol ; 140: 116-124, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31271996

RESUMO

BACKGROUND: Traditional concepts of radiobiology model the direct radiation-induced cellular cytotoxicity but are not focused on late and sustained effects of radiation. Recent experimental data show the close involvement of immunological processes. METHODS: Based on systematic PubMed searches, experimental data on immunological radiation effects are summarized and analyzed in a non-quantitative descriptive manner to provide a translational perspective on the immuno-modulatory impact of radiation in cancer. RESULTS: Novel experimental findings document that sustained radiation effects are ultimately mediated through systemic factors such as cytotoxic CD8+ T cells and involve a local immuno-stimulation. Increased tumor infiltration of CD8+ T cell is a prerequisite for long-term radiation effects. CD8+ T cell depletion induces radio-resistance in experimental tumors. The proposed sequence of events involves radiation-damaged cells that release HMGB1, which activates macrophages via TLR4 to a local immuno-stimulation via TNF, which contributes to maturation of DCs. The mature DCs migrate to lymph nodes where they trigger effective CD8+ T cell responses. Radiation effects are boosted, when the physiological self-terminating negative feedback of immune reactions is antagonised via blocking of TGF-ß or via checkpoint inhibition with involvement of CD8+ T cells as common denominator. CONCLUSION: The concept of immuno-radiobiology emphasizes the necessity for a functional integrity of APCs and T cells for the long-term effects of radiotherapy. Local irradiation at higher doses induces tumor infiltration of CD8+ T cells, which can be boosted by immunotherapy. More systematic research is warranted to better understand the immunological effects of escalating radiation doses.


Assuntos
Linfócitos T CD8-Positivos/efeitos da radiação , Imunomodulação/efeitos da radiação , Neoplasias/radioterapia , Animais , Linfócitos T CD8-Positivos/imunologia , Humanos , Imunoterapia , Neoplasias/imunologia
19.
Hematol Oncol Clin North Am ; 33(2): 233-248, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30832997

RESUMO

Radiotherapy has known immunomodulatory effects and there exists a strong preclinical rationale for combining radiotherapy with immunotherapies. Broadly, the concurrent administration of immunotherapies and radiotherapy does not seem to result in undue toxicity, even when radiotherapy is administered to definitive doses. Recently reported results from prospective clinical trials evaluating radiotherapy/ICB combinations, such as the PACIFIC trial, provide important information on safety and efficacy in the definitive setting and identify potential abscopal effects. This review details the preclinical foundation for the combination of radiotherapy and immunotherapies, summarizes the most recent clinical data available, and highlights active and future areas of study.


Assuntos
Imunomodulação/efeitos da radiação , Neoplasias/imunologia , Neoplasias/radioterapia , Radioimunoterapia , Ensaios Clínicos como Assunto , Humanos
20.
Front Immunol ; 10: 193, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30828330

RESUMO

In recent decades, there has been substantial growth in our understanding of the immune system and its role in tumor growth and overall survival. A central finding has been the cross-talk between tumor cells and the surrounding environment or stroma. This tumor stroma, comprised of various cells, and extracellular matrix (ECM), has been shown to aid in suppressing host immune responses against tumor cells. Through immunosuppressive cytokine secretion, metabolic alterations, and other mechanisms, the tumor stroma provides a complex network of safeguards for tumor proliferation. With recent advances in more effective, localized treatment, radiation therapy (XRT) has allowed for strategies that can effectively alter and ablate tumor stromal tissue. This includes promoting immunogenic cell death through tumor antigen release to increasing immune cell trafficking, XRT has a unique advantage against the tumoral immune evasion mechanisms that are orchestrated by stromal cells. Current studies are underway to elucidate pathways within the tumor stroma as potential targets for immunotherapy and chemoradiation. This review summarizes the effects of tumor stroma in tumor immune evasion, explains how XRT may help overcome these effects, with potential combinatorial approaches for future treatment modalities.


Assuntos
Neoplasias/patologia , Neoplasias/radioterapia , Células Estromais/efeitos da radiação , Microambiente Tumoral/efeitos da radiação , Animais , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/efeitos da radiação , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Matriz Extracelular/efeitos da radiação , Humanos , Imunidade , Imunomodulação/efeitos da radiação , Neoplasias/imunologia , Tolerância a Radiação/imunologia , Tolerância a Radiação/efeitos da radiação , Radioterapia , Células Estromais/imunologia , Microambiente Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...