Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Theranostics ; 11(14): 7045-7056, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093870

RESUMO

Rationale: Precise treatment of tumors is attracting increasing attention. Molecular probes simultaneously demonstrating the diagnostic signal and pharmacological effect in response to tumor microenvironment are highly desired. γ-glutamyl transpeptidase (GGT) is a biomarker with significantly up-regulated expression in the tumor area. We developed a GGT responsive near-infrared (NIR) nanoassembly for tumor-specific fluorescence imaging-guided photothermal therapy. Methods: The GGT responsive NIR probe was constructed by conjugating GGT-specific substrate γ-glutamic acid (γ-Glu) with cyanine fluorophore (NRh-NH2) via amide reaction. The resulting NRh-G spontaneously assembled into nanoparticles (NRh-G-NPs) around 50 nm. The NPs were characterized and the properties evaluated in the presence or absence of GGT. Subsequently, we studied fluorescence imaging and photothermal therapy of NRh-G-NPs in vitro and in vivo. Results: NRh-G-NPs, upon specific reaction with GGT, turned into NRh-NH2-NPs, showing a ~180-fold fluorescence enhancement and excellent photothermal effect recovery. NRh-G-NPs could selectively light up U87MG tumor cells while their fluorescence was weak in L02 human normal liver cells. The NPs also showed excellent tumor cell ablation upon laser irradiation. After intravenous injection into tumor-bearing mice, NRh-G-NPs could arrive in the tumor area and specifically light up the tumor. Following laser irradiation, the tumor could be completely erased with no tumor reoccurrence for up to 40 days. Conclusions: NRh-G-NPs were specifically responsive to GGT overexpressed in U87MG tumor cells and selectively lit up the tumor for imaging-guided therapy. Besides, the recovery of photothermal property in the tumor area could improve cancer therapy precision and decreased side effects in normal tissues.


Assuntos
Glioma/tratamento farmacológico , Glioma/radioterapia , Hipertermia Induzida/métodos , Nanopartículas/química , Terapia Fototérmica/métodos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos da radiação , gama-Glutamiltransferase/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Feminino , Fluorescência , Corantes Fluorescentes/química , Ácido Glutâmico/química , Humanos , Lasers , Camundongos , Camundongos Nus , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Espectroscopia de Luz Próxima ao Infravermelho , gama-Glutamiltransferase/genética
2.
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
3.
ACS Appl Mater Interfaces ; 12(52): 57768-57781, 2020 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-33326213

RESUMO

Solid tumors inevitably develop radioresistance due to low oxygen partial pressure in the tumor microenvironment. Despite numerous attempts, there are still few effective ways to avoid the hypoxia-induced poor radiotherapeutic effect. To overcome this problem, platinum (Pt) nanodots were fabricated into a mesoporous bismuth (Bi)-based nanomaterial to construct a biodegradable nanocomposite BiPt-folic acid-modified amphiphilic polyethylene glycol (PFA). BiPt-PFA could act as a radiosensitizer to enhance the absorption of X-rays at the tumor site and simultaneously trigger response behaviors related to the tumor microenvironment due to the enrichment of materials in the tumor area. During this process, the Bi-based component consumed glutathione via coordination, thus altering the oxidative stress balance, while Pt nanoparticles catalyzed the decomposition of hydrogen peroxide to generate oxygen, thereby relieving tumor hypoxia. Both Pt and Bi thus co-modulated the tumor microenvironment to improve the radiotherapeutic effect. In addition, Pt dots in BiPt-PFA had strong near-infrared absorption ability and created an intensive photothermal therapeutic effect. Modulation of the tumor microenvironment could thus improve the therapeutic effect in hypoxic tumors by a combination of photothermal therapy and enhanced radiotherapy. BiPt-PFA, as a biodegradable nanocomposite, may thus modulate the tumor microenvironment to enhance the hypoxic tumor therapeutic effect by thermoradiotherapy.


Assuntos
Bismuto/química , Nanocompostos/química , Radiossensibilizantes/química , Hipóxia Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Humanos , Hipertermia Induzida , Nanopartículas Metálicas/química , Camundongos , Platina/química , Polietilenoglicóis/química , Porosidade , Radiossensibilizantes/farmacologia , Segurança , Solubilidade , Hipóxia Tumoral/efeitos da radiação , Microambiente Tumoral/efeitos da radiação , Água/química
4.
J Immunol Res ; 2020: 8459496, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32411806

RESUMO

NK cells are lymphocytes with antitumor properties and can directly lyse tumor cells in a non-MHC-restricted manner. However, the tumor microenvironment affects the immune function of NK cells, which leads to immune evasion. This may be related to the pathogenesis of some diseases. Therefore, great efforts have been made to improve the immunotherapy effect of natural killer cells. NK cells from different sources can meet different clinical needs, in order to minimize the inhibition of NK cells and maximize the response potential of NK cells, for example, modification of NK cells can increase the number of NK cells in tumor target area, change the direction of NK cells, and improve their targeting ability to malignant cells. Checkpoint blocking is also a promising strategy for NK cells to kill tumor cells. Combination therapy is another strategy for improving antitumor ability, especially in combination with oncolytic viruses and nanomaterials. In this paper, the mechanisms affecting the activity of NK cells were reviewed, and the therapeutic potential of different basic NK cell strategies in tumor therapy was focused on. The main strategies for improving the immune function of NK cells were described, and some new strategies were proposed.


Assuntos
Imunoterapia/métodos , Células Matadoras Naturais/transplante , Neoplasias/terapia , Animais , Antígenos de Neoplasias/imunologia , Antineoplásicos Imunológicos/administração & dosagem , Terapia Combinada/métodos , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/métodos , Humanos , Inibidores de Checkpoint Imunológico/administração & dosagem , Evasão da Resposta Imune/efeitos dos fármacos , Memória Imunológica , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/efeitos da radiação , Magnetoterapia , Camundongos , Nanomedicina/métodos , Nanopartículas/administração & dosagem , Neoplasias/imunologia , Vírus Oncolíticos/imunologia , Receptores de Antígenos Quiméricos/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação
5.
Nat Commun ; 11(1): 1126, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-32111847

RESUMO

The efficacy of nano-mediated drug delivery has been impeded by multiple biological barriers such as the mononuclear phagocyte system (MPS), as well as vascular and interstitial barriers. To overcome the abovementioned obstacles, we report a nano-pathogenoid (NPN) system that can in situ hitchhike circulating neutrophils and supplement photothermal therapy (PTT). Cloaked with bacteria-secreted outer membrane vesicles inheriting pathogen-associated molecular patterns of native bacteria, NPNs are effectively recognized and internalized by neutrophils. The neutrophils migrate towards inflamed tumors, extravasate across the blood vessels, and penetrate through the tumors. Then NPNs are rapidly released from neutrophils in response to inflammatory stimuli and subsequently taken up by tumor cells to exert anticancer effects. Strikingly, due to the excellent targeting efficacy, cisplatin-loaded NPNs combined with PTT completely eradicate tumors in all treated mice. Such a nano-platform represents an efficient and generalizable strategy towards in situ cell hitchhiking as well as enhanced tumor targeted delivery.


Assuntos
Quimiotaxia de Leucócito , Sistemas de Liberação de Medicamentos , Nanopartículas/administração & dosagem , Neoplasias/terapia , Neutrófilos/fisiologia , Fototerapia , Animais , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/imunologia , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Cisplatino/administração & dosagem , Cisplatino/química , Cisplatino/farmacocinética , Liberação Controlada de Fármacos , Vesículas Extracelulares/química , Vesículas Extracelulares/imunologia , Imunoterapia Adotiva , Inflamação/etiologia , Camundongos , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Ativação de Neutrófilo , Infiltração de Neutrófilos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Moléculas com Motivos Associados a Patógenos/imunologia , Fototerapia/efeitos adversos , Microambiente Tumoral/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Sci Rep ; 10(1): 1653, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32015396

RESUMO

For multimodality therapies such as the combination of hyperthermia and radiation, quantification of biological effects is key for dose prescription and response prediction. Tumour spheroids have a microenvironment that more closely resembles that of tumours in vivo and may thus be a superior in vitro cancer model than monolayer cultures. Here, the response of tumour spheroids formed from two established human cancer cell lines (HCT116 and CAL27) to single and combination treatments of radiation (0-20 Gy), and hyperthermia at 47 °C (0-780 CEM43) has been evaluated. Response was analysed in terms of spheroid growth, cell viability and the distribution of live/dead cells. Time-lapse imaging was used to evaluate mechanisms of cell death and cell detachment. It was found that sensitivity to heat in spheroids was significantly less than that seen in monolayer cultures. Spheroids showed different patterns of shrinkage and regrowth when exposed to heat or radiation: heated spheroids shed dead cells within four days of heating and displayed faster growth post-exposure than samples that received radiation or no treatment. Irradiated spheroids maintained a dense structure and exhibited a longer growth delay than spheroids receiving hyperthermia or combination treatment at (thermal) doses that yielded equivalent levels of clonogenic cell survival. We suggest that, unlike radiation, which kills dividing cells, hyperthermia-induced cell death affects cells independent of their proliferation status. This induces microenvironmental changes that promote spheroid growth. In conclusion, 3D tumour spheroid growth studies reveal differences in response to heat and/or radiation that were not apparent in 2D clonogenic assays but that may significantly influence treatment efficacy.


Assuntos
Hipertermia Induzida , Neoplasias/radioterapia , Neoplasias/terapia , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Terapia Combinada , Relação Dose-Resposta à Radiação , Células HCT116 , Humanos , Modelos Biológicos , Neoplasias/patologia , Esferoides Celulares/patologia , Esferoides Celulares/efeitos da radiação , Microambiente Tumoral/efeitos da radiação , Ensaio Tumoral de Célula-Tronco
7.
Angew Chem Int Ed Engl ; 59(23): 8833-8838, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-31943602

RESUMO

Continuous irradiation during photodynamic therapy (PDT) inevitably induces tumor hypoxia, thereby weakening the PDT effect. In PDT-induced hypoxia, providing singlet oxygen from stored chemical energy may enhance the cell-killing effect and boost the therapeutic effect. Herein, we present a phototheranostic (DPPTPE@PEG-Py NPs) prepared by using a 2-pyridone-based diblock polymer (PEG-Py) to encapsulate a semiconducting, heavy-atom-free pyrrolopyrrolidone-tetraphenylethylene (DPPTPE) with high singlet-oxygen-generation ability both in dichloromethane and water. The PEG-Py can trap the 1 O2 generated from DPPTPE under laser irradiation and form a stable intermediate of endoperoxide, which can then release 1 O2 in the dark, hypoxic tumor microenvironment. Furthermore, fluorescence-imaging-guided phototherapy demonstrates that this phototheranostic could completely inhibit tumor growth with the help of laser irradiation.


Assuntos
Escuridão , Fototerapia/métodos , Oxigênio Singlete/metabolismo , Hipóxia Tumoral/efeitos da radiação , Microambiente Tumoral/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Humanos , Lasers , Imagem Óptica , Polietilenoglicóis/química , Pirrolidinonas/química , Oxigênio Singlete/química , Estilbenos/química
8.
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
9.
Anal Cell Pathol (Amst) ; 2016: 6146595, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26998418

RESUMO

Cancer originates from genetic mutations accumulation. Cancer stem cells have been depicted as tumorigenic cells that can differentiate and self-renew. Cancer stem cells are thought to be resistant to conventional therapy like chemotherapy and radiation therapy. Radiation therapy and chemotherapy damage carcinomic DNA cells. Because of the ability of cancer stem cells to self-renew and reproduce malignant tumors, they are the subject of intensive research. In this review, CSCs radioresistant mechanisms which include DNA damage response and natural radiosensitizers have been summed up. Reactive oxygen species play an important role in different physiological processes. ROS scavenging is responsible for regulation of reactive oxygen species generation. A researcher has proved that microRNAs regulate tumor radiation resistance. Ionizing radiation does not kill the cancer cells; rather, IR just slows down the signs and symptoms. Ionizing radiation damages DNA directly/indirectly. IR is given mostly in combination with other chemo/radiotherapies. We briefly described here the behavior of cancer stem cells and radioresistance therapies in cancer treatment. To overcome radioresistance in treatment of cancer, strategies like fractionation modification, treatment in combination, inflammation modification, and overcoming hypoxic tumor have been practiced. Natural radiosensitizers, for example, curcumin, genistein, and quercetin, are more beneficial than synthetic compounds.


Assuntos
Neoplasias/terapia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos da radiação , Radiossensibilizantes/uso terapêutico , Radioterapia/métodos , Terapia Combinada , Dano ao DNA , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/efeitos da radiação
10.
J Leukoc Biol ; 97(4): 711-21, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25632046

RESUMO

RT is commonly used to treat malignant tumors. However, tumor regrowth is a major limitation to RT as an antitumor treatment. In the present study, we investigated the tumor-promoting effects of high-dose (or ablative) RT treatments on tumor-bearing mice. We focused on the role of macrophages that interact with IR-CCs in the TME, which cause tumor regrowth. We observed that CT26(H-2(d)) tumor growth was enhanced by i.v. injection of IR-CT26 cells compared with NR control CT26 cells. The levels of iNOS gene expression and NO production from RAW264.7 macrophages (H-2(d)) in response to the interaction with IR-CT26 cells were higher than with NR-CT26 cells. When CT26 tumor-bearing mice were treated i.v. with L-NMMA, a NOS inhibitor, the reduction in in vivo tumor growth was higher in the IR-CT26-injected group compared with the NR-CT26-injected control group. In vivo CT26 tumor growth was decreased after transplanting PEM extracted from L-NMMA-treated, tumor-bearing mice. Although iNOS activity was reduced by inhibiting TLR1 expression with TLR1-siRNA, it was enhanced by TLR1 overexpression. Transcriptional activation and protein expression levels of iNOS were also decreased in the presence of TLR1-siRNA but increased as a result of TLR1 overexpression. These results demonstrate that postradiotherapeutic tumor regrowth may be caused by interaction of IR-CCs with macrophages that induce TLR1-mediated iNOS expression and NO production. Our data suggest that iNOS in macrophages could be a useful target to regulate postradiotherapeutic responses in hosts and subsequently limit tumor regrowth.


Assuntos
Adenocarcinoma/radioterapia , Neoplasias do Colo/radioterapia , Raios gama , Macrófagos/metabolismo , Melanoma Experimental/radioterapia , Proteínas de Neoplasias/fisiologia , Óxido Nítrico Sintase Tipo II/fisiologia , Óxido Nítrico/fisiologia , Receptor 1 Toll-Like/fisiologia , Microambiente Tumoral/efeitos da radiação , Células 3T3 , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Células da Medula Óssea/metabolismo , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Técnicas de Cocultura , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Progressão da Doença , Indução Enzimática , Macrófagos/classificação , Macrófagos Peritoneais/metabolismo , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Recidiva , Receptor 1 Toll-Like/biossíntese , Receptor 1 Toll-Like/genética , ômega-N-Metilarginina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA