Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35163354

RESUMO

Radiotherapy is a crucial cancer treatment, but its outcome is still far from satisfactory. One of the reasons that cancer cells show resistance to ionizing radiation is hypoxia, defined as a low level of oxygenation, which is typical for solid tumors. In the hypoxic environment, cancer cells are 2-3 times more resistant to ionizing radiation than normoxic cells. To overcome this important impediment, radiosensitizers should be introduced to cancer therapy. When modified with an electrophilic substituent, nucleosides may undergo efficient dissociative electron attachment (DEA) that leaves behind nucleoside radicals, which, in secondary reactions, are able to induce DNA damage, leading to cancer cell death. We report the radiosensitizing effect of one of the best-known DEA-type radiosensitizers-5-bromo-2'-deoxyuridine (BrdU)-on breast (MCF-7) and prostate (PC3) cancer cells under both normoxia and hypoxia. MCF-7 and PC3 cells were treated with BrdU to investigate the effect of hypoxia on cell proliferation, incorporation into DNA and radiosensitivity. While the oxygen concentration did not significantly affect the efficiency of BrdU incorporation into DNA or the proliferation of tumor cells, the radiosensitizing effect of BrdU on hypoxic cells was more evident than on normoxic cells. Further mechanistic studies performed with the use of flow cytometry showed that under hypoxia, BrdU increased the level of histone H2A.X phosphorylation after X-ray exposure to a greater extent than under normal oxygenation conditions. These results confirm that the formation of double-strand breaks in hypoxic BrdU-treated cancer cells is more efficient. In addition, by performing stationary radiolysis of BrdU solution in the presence of an ●OH radical scavenger, we compared the degree of its electron-induced degradation under aerobic and anaerobic conditions. It was determined that radiodegradation under anaerobic conditions was almost twice as high as that under aerobic conditions.


Assuntos
Bromodesoxiuridina/farmacologia , Histonas/metabolismo , Neoplasias/genética , Radiossensibilizantes/farmacologia , Anaerobiose , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Células MCF-7 , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/radioterapia , Células PC-3 , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Hipóxia Tumoral/efeitos da radiação
2.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-35055167

RESUMO

The receptor tyrosine kinase AXL (RTK-AXL) is implicated in therapy resistance and tumor progression in glioblastoma multiforme (GBM). Here, we investigated therapy-induced receptor modifications and how endogenous RTK-AXL expression and RTK-AXL inhibition contribute to therapy resistance in GBM. GBM cell lines U118MG and SF126 were exposed to temozolomide (TMZ) and radiation (RTX). Receptor modifications in response to therapy were investigated on protein and mRNA levels. TMZ-resistant and RTK-AXL overexpressing cell lines were exposed to increasing doses of TMZ and RTX, with and without RTK-AXL tyrosine kinase inhibitor (TKI). Colorimetric microtiter (MTT) assay and colony formation assay (CFA) were used to assess cell viability. Results showed that the RTK-AXL shedding product, C-terminal AXL (CT-AXL), rises in response to repeated TMZ doses and under hypoxia, acts as a surrogate marker for radio-resistance. Endogenous RTX-AXL overexpression leads to therapy resistance, whereas combination therapy of TZM and RTX with TKI R428 significantly increases therapeutic effects. This data proves the role of RTK-AXL in acquired and intrinsic therapy resistance. By demonstrating that therapy resistance may be overcome by combining AXL TKI with standard treatments, we have provided a rationale for future study designs investigating AXL TKIs in GBM.


Assuntos
Benzocicloeptenos/farmacologia , Neoplasias Encefálicas/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Temozolomida/farmacologia , Triazóis/farmacologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Terapia Combinada , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioblastoma/metabolismo , Glioblastoma/terapia , Humanos , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Hipóxia Tumoral/efeitos dos fármacos , Hipóxia Tumoral/efeitos da radiação , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/efeitos da radiação , Receptor Tirosina Quinase Axl
3.
Molecules ; 27(1)2021 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-35011360

RESUMO

Anticancer treatment is largely affected by the hypoxic tumor microenvironment (TME), which causes the resistance of the tumor to radiotherapy. Combining radiosensitizer compounds and O2 self-enriched moieties is an emerging strategy in hypoxic-tumor treatments. Herein, we engineered GdW10@PDA-CAT (K3Na4H2GdW10O36·2H2O, GdW10, polydopamine, PDA, catalase, CAT) composites as a radiosensitizer for the TME-manipulated enhancement of radiotherapy. In the composites, Gd (Z = 64) and W (Z = 74), as the high Z elements, make X-ray gather in tumor cells, thereby enhancing DNA damage induced by radiation. CAT can convert H2O2 to O2 and H2O to enhance the X-ray effect under hypoxic TME. CAT and PDA modification enhances the biocompatibility of the composites. Our results showed that GdW10@PDA-CAT composites increased the efficiency of radiotherapy in HT29 cells in culture. This polyoxometalates and O2 self-supplement composites provide a promising radiosensitizer for the radiotherapy field.


Assuntos
Gadolínio/química , Nanocompostos/química , Radiossensibilizantes/química , Hipóxia Tumoral/efeitos da radiação , Tungstênio/química , Ânions/química , Materiais Biocompatíveis/química , Catalase/metabolismo , Linhagem Celular Tumoral , Células HT29 , Humanos , Peróxido de Hidrogênio/metabolismo , Indóis/química , Oxigênio/metabolismo , Polieletrólitos/química , Polímeros/química , Radiossensibilizantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral
4.
Electromagn Biol Med ; 39(3): 239-249, 2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-32410511

RESUMO

Electrochemotherapy (ECT), the combination of electric pulses (EPs) and an anticancer drug, is a type of cancer treatment method. We investigated the effect of 217-Hz magnetic fields (MFs) similar to that generated by GSM900 mobile phones, as intervening factors, on proposed mechanisms of ECT including permeability, tumor hypoxia and immune system response. The 4T1 cells were exposed to extremely low-frequency (ELF)-MFs at 93, 120 or 159 µT intensities, generated by Helmholtz coils 10 min, and then put in individual groups, comprising no treatment, chemotherapy, EPs or ECT. The cell viability was evaluated. Then, two treatment protocols were selected for in vivo experiments. The mice with 4T1 tumor cells were exposed to ELF-MFs 10 min/day until the day their tumors reached 8 mm in diameter. Then, the tumors were treated to ECT. Tumor hypoxia and immune system response were analyzed through immunohistochemistry assay and enzyme-linked immunosorbent assay technique, respectively. The results in vitro indicated a significant decreased ECT efficacy of 60 V/cm, 5 kHz at the flux density of 93 µT. The results in vivo showed that pre-exposure to ELF-MFs could increase tumor hypoxia induced by ECT. In addition, exposure to ELF-MFs before ECT caused a significant increase in interferon-γ/interleukin-4 in comparison with ECT alone. More studies, including studies on the effect of ELF-MFs emitted from mobile phones on tumor volume changes induced by ECT, are needed to elucidate how the process of ECT is influenced by the MFs.


Assuntos
Telefone Celular , Eletroquimioterapia , Campos Eletromagnéticos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Feminino , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Hipóxia Tumoral/efeitos dos fármacos , Hipóxia Tumoral/efeitos da radiação
5.
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
6.
J Am Chem Soc ; 141(6): 2695-2702, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30652866

RESUMO

Strong oxygen dependence, poor tumor targeting, and limited treatment depth have been considered as the "Achilles' heels" facing the clinical usage of photodynamic therapy (PDT). Different from common approaches, here, we propose an innovative tactic by using photon-initiated dyad cationic superoxide radical (O2-•) generator (ENBOS) featuring "0 + 1 > 1" amplification effect to simultaneously overcome these drawbacks. In particular, by taking advantage of the Förster resonance energy transfer theory, the energy donor successfully endows ENBOS with significantly enhanced NIR absorbance and photon utility, which in turn lead to ENBOS more easily activated and generating more O2-• in deep tissues, that thus dramatically intensifies the type I PDT against hypoxic deep tumors. Moreover, benefiting from the dyad cationic feature, ENBOS achieves superior "structure-inherent targeting" abilities with the signal-to-background ratio as high as 25.2 at 48 h post intravenous injection, offering opportunities for accurate imaging-guided tumor treatment. Meanwhile, the intratumoral accumulation and retention performance are also markedly improved (>120 h). On the basis of these unique merits, ENBOS selectively inhibits the deep-seated hypoxic tumor proliferation at a low light-dose irradiation. Therefore, this delicate design may open new horizons and cause a paradigm change for PDT in future cancer therapy.


Assuntos
Fotoquimioterapia , Superóxidos/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Espaço Intracelular/metabolismo , Espaço Intracelular/efeitos da radiação , Camundongos , Hipóxia Tumoral/efeitos dos fármacos , Hipóxia Tumoral/efeitos da radiação
7.
Clin Exp Pharmacol Physiol ; 46(5): 407-412, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30721527

RESUMO

Methenamine (hexamethylenetetramine, hexamine, urotropine) is a compound discovered in 1859, which is still currently being used as a urinary antiseptic. Methenamine is highly soluble in water and polar solvents, and its molecular constitution is similar to adamantane compounds with tetrahedral cage like structure. In acidic conditions, methenamine decomposes to formaldehyde and ammonia. Recently, methenamine has gained a renewal of interest due to antibiotic-resistant bacteria urinary tract infections; interestingly, bacteria cannot gain resistance to formaldehyde. In 1968, David and Burkitt reported remarkable regression of four Burkitt Lymphoma patients in eight subjects who were treated with septicemine (a solution containing 6.3 g of methenamine iodomethylate and 1 g of methenamine sodium benzoate in 100 cc distilled water). Unfortunately, these striking observations did not gain interest in the medical community; despite experimental models that showed that methenamine synergized with hyperthermia, radiation, and chemotherapy to block cancer growth. As the hypoxic core of tumours have an acidic pH, it would be plausible to expect that methenamine would selectively target dormant, non-proliferative, and treatment-resistant cancer clones in large tumours. Moreover, previous data suggests that methenamine can be safely used intravenously and for treatment of infections of the central nervous system. It may therefore be an effective adjuvant in treatment of systemic cancers and glioblastoma.


Assuntos
Anti-Infecciosos Urinários/farmacologia , Reposicionamento de Medicamentos , Glioblastoma/tratamento farmacológico , Metenamina/farmacologia , Radiossensibilizantes/farmacologia , Hipóxia Tumoral/efeitos dos fármacos , Animais , Anti-Infecciosos Urinários/uso terapêutico , Glioblastoma/patologia , Glioblastoma/radioterapia , Humanos , Metenamina/uso terapêutico , Radiossensibilizantes/uso terapêutico , Hipóxia Tumoral/efeitos da radiação
8.
Angew Chem Int Ed Engl ; 58(42): 15069-15075, 2019 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-31429172

RESUMO

Hypoxia, as a characteristic feature of solid tumor, can significantly adversely affect the outcomes of cancer radiotherapy (RT), photodynamic therapy, or chemotherapy. In this study, a strategy is developed to overcome tumor hypoxia-induced radiotherapy tolerance. Specifically, a novel two-dimensional Pd@Au bimetallic core-shell nanostructure (TPAN) was employed for the sustainable and robust production of O2 in long-term via the catalysis of endogenous H2 O2 . Notably, the catalytic activity of TPAN could be enhanced via surface plasmon resonance (SPR) effect triggered by NIR-II laser irradiation, to enhance the O2 production and thereby relieve tumor hypoxia. Thus, TPAN could enhance radiotherapy outcomes by three aspects: 1) NIR-II laser triggered SPR enhanced the catalysis of TPAN to produce O2 for relieving tumor hypoxia; 2) high-Z element effect arising from Au and Pd to capture X-ray energy within the tumor; and 3) TPAN affording X-ray, photoacoustic, and NIR-II laser derived photothermal imaging, for precisely guiding cancer therapy, so as to reduce the side effects from irradiation.


Assuntos
Ouro/química , Nanopartículas/química , Neoplasias , Oxigênio/metabolismo , Paládio/química , Tolerância a Radiação , Ressonância de Plasmônio de Superfície/métodos , Hipóxia Tumoral/efeitos dos fármacos , Animais , Catálise , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Feminino , Ouro/administração & dosagem , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Neoplasias/metabolismo , Neoplasias/radioterapia , Paládio/administração & dosagem , Tamanho da Partícula , Propriedades de Superfície , Hipóxia Tumoral/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Pharmacol Exp Ther ; 365(3): 536-543, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29581154

RESUMO

Previously, we demonstrated that nitric oxide (NO) synthase (NOS) is uncoupled in a wide range of solid tumors and that restoring NOS coupling with the tetrahydrobiopterin precursor sepiapterin (SP) inhibits tumor progression. Endothelial dysfunction characterizes the poorly functional vasculature of solid tumors, and since NO is critical for regulation of endothelial function we asked whether SP, by recoupling NOS, improves tumor vasculature structure and function-enhancing chemotherapeutic delivery and response to radiotherapy. MMTV-neu mice with spontaneous breast tumors were treated with SP by oral gavage and evaluated by multispectral optoacoustic tomographic analysis of tumor HbO2 and by tissue staining for markers of hypoxia, blood perfusion, and markers of endothelial and smooth muscle proteins. Recoupling tumor NOS activity results in vascular normalization observed as reduced tumor hypoxia, improved tumor percentage of HbO2 and perfusion, as well as increased pericyte coverage of tumor blood vessels. The normalized vasculature and improved tumor oxygenation led to a greater than 2-fold increase in radiation-induced apoptosis compared with radiation or SP alone. High-performance liquid chromatography analysis of tumor doxorubicin levels showed a greater than 50% increase in doxorubicin uptake and a synergistic effect on tumor cell apoptosis. This study highlights for the first time the importance of NOS uncoupling and endothelial dysfunction in the development of tumor vasculature and presents a new approach for improving the tumoricidal efficacies of chemotherapy and radiotherapy.


Assuntos
Antineoplásicos/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Pterinas/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Animais , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/efeitos da radiação , Circulação Sanguínea/efeitos dos fármacos , Circulação Sanguínea/efeitos da radiação , Vasos Sanguíneos/fisiopatologia , Vasos Sanguíneos/efeitos da radiação , Linhagem Celular Tumoral , Doxorrubicina/metabolismo , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Humanos , Camundongos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/metabolismo , Oxigênio/metabolismo , Hipóxia Tumoral/efeitos dos fármacos , Hipóxia Tumoral/efeitos da radiação
10.
Acta Oncol ; 57(9): 1216-1224, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29630428

RESUMO

BACKGROUND: Earlier in vitro studies show that irradiation with an ultra-low dose-rate of 15 mGy/h delivered with [3H]-valine leads to loss of clonogenicity in hypoxic T-47D cells. Here, the aim was to determine if [3H]-valine could be used to deliver low dose-rate irradiation in a colorectal cancer model. METHODS: Clonogenicity was measured in cultured cancer cell line HT29 irradiated with 15 mGy/h combined with intermittent hypoxia. Mice with HT29 xenografts were irradiated by repeated injections of [3H]-valine intravenously. The activity in the tumor tissue was measured by scintillation counting and tumor growth, hypoxic fraction and tritium distribution within tumors were assessed by pimonidazole staining and autoradiography. RESULTS: Ultra-low dose-rate irradiation decreased clonogenicity in hypoxic colorectal cancer cells. In vivo, the tumor growth, hypoxic fraction and weight of the mice were similar between the treated and untreated group. Autoradiography showed no [3H]-valine uptake in hypoxic tumor regions in contrast to aerobic tissue. CONCLUSION: Continuous low-dose-rate irradiation was well tolerated by aerobic tissue. This indicates a potential use of low dose-rate irradiation to target hypoxic tumor cells in combination with high dose-rate irradiation to eradicate the well oxygenated tumor regions. However, [3H]-valine is not the appropriate method to deliver ultra-low dose-rate irradiation in vivo.


Assuntos
Neoplasias Colorretais/patologia , Neoplasias Colorretais/radioterapia , Trítio/uso terapêutico , Hipóxia Tumoral/efeitos da radiação , Valina/uso terapêutico , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Dosagem Radioterapêutica , Valina/química , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Bull Math Biol ; 80(5): 1207-1235, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29488054

RESUMO

Current protocols for delivering radiotherapy are based primarily on tumour stage and nodal and metastases status, even though it is well known that tumours and their microenvironments are highly heterogeneous. It is well established that the local oxygen tension plays an important role in radiation-induced cell death, with hypoxic tumour regions responding poorly to irradiation. Therefore, to improve radiation response, it is important to understand more fully the spatiotemporal distribution of oxygen within a growing tumour before and during fractionated radiation. To this end, we have extended a spatially resolved mathematical model of tumour growth, first proposed by Greenspan (Stud Appl Math 51:317-340, 1972), to investigate the effects of oxygen heterogeneity on radiation-induced cell death. In more detail, cell death due to radiation at each location in the tumour, as determined by the well-known linear-quadratic model, is assumed also to depend on the local oxygen concentration. The oxygen concentration is governed by a reaction-diffusion equation that is coupled to an integro-differential equation that determines the size of the assumed spherically symmetric tumour. We combine numerical and analytical techniques to investigate radiation response of tumours with different intratumoral oxygen distribution profiles. Model simulations reveal a rapid transient increase in hypoxia upon regrowth of the tumour spheroid post-irradiation. We investigate the response to different radiation fractionation schedules and identify a tumour-specific relationship between inter-fraction time and dose per fraction to achieve cure. The rich dynamics exhibited by the model suggest that spatial heterogeneity may be important for predicting tumour response to radiotherapy for clinical applications.


Assuntos
Modelos Biológicos , Neoplasias/radioterapia , Morte Celular/efeitos da radiação , Simulação por Computador , Fracionamento da Dose de Radiação , Humanos , Modelos Lineares , Conceitos Matemáticos , Neoplasias/metabolismo , Neoplasias/patologia , Oxigênio/metabolismo , Tolerância a Radiação , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Esferoides Celulares/efeitos da radiação , Hipóxia Tumoral/efeitos da radiação
12.
Mar Drugs ; 16(12)2018 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-30558324

RESUMO

Tumor hypoxia is a major mechanism of resistance to radiation therapy (RT), which is associated with poor prognosis in affected cancer patients. Various approaches to treat hypoxic and radioresistant cancers, including pancreatic cancer, have shown limited success. Fucoidan, a polysaccharide from brown seaweed, has antitumor and antiangiogenesis activities. Here, we discuss the development of fucoidan-coated manganese dioxide nanoparticles (Fuco-MnO2-NPs) and testing of the therapeutic potential with RT using pancreatic cancer models. In vitro data showed that Fuco-MnO2-NPs generated oxygen efficiently in the presence of H2O2 and substantially suppressed HIF-1 expression under a hypoxic condition in human pancreatic cancer cells. Fuco-MnO2-NPs reversed hypoxia-induced radioresistance by decreasing clonogenic survival and increasing DNA damage and apoptotic cell death in response to RT. In a BxPC3 xenograft mouse model, the combination treatment with Fuco-MnO2-NPs and RT resulted in a greater tumor growth delay than RT alone. Fucoidan-coated NPs, but not naked ones, further suppressed tumor angiogenesis, as judged by immunohistochemistry data with diminished expression of phosphorylated vascular endothelial growth factor receptor 2 (VEGFR2) and CD31. These data suggest that Fuco-MnO2-NPs may potentiate the effects of RT via dual targeting of tumor hypoxia and angiogenesis, and they are of great clinical potential in the treatment of hypoxic, radioresistant pancreatic cancer.


Assuntos
Antineoplásicos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Neoplasias Pancreáticas/terapia , Polissacarídeos/farmacologia , Hipóxia Tumoral/efeitos dos fármacos , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Quimiorradioterapia/métodos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Compostos de Manganês/química , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Óxidos/química , Neoplasias Pancreáticas/patologia , Polissacarídeos/uso terapêutico , Tolerância a Radiação/efeitos dos fármacos , Alga Marinha/química , Resultado do Tratamento , Hipóxia Tumoral/efeitos da radiação , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Adv Exp Med Biol ; 977: 287-296, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28685458

RESUMO

Modern standards for radiation treatment do not take into account tumor oxygenation for radiation treatment planning. Strong correlation between tumor oxygenation and radiation treatment success suggests that oxygen-guided radiation therapy (OGRT) may be a promising enhancement of cancer radiation treatment. We have developed an OGRT protocol for rodents. Electron paramagnetic resonance (EPR) imaging is used for recording oxygen maps with high spatial resolution and excellent accuracy better than 1 torr. Radiation is delivered with an animal intensity modulated radiation therapy (IMRT) XRAD225Cx micro-CT/ therapy system. The radiation plan is delivered in two steps. First, a uniform 15% tumor control dose (TCD15) is delivered to the whole tumor. In the second step, an additional booster dose amounting to the difference between TCD98 and TCD15 is delivered to radio-resistant, hypoxic tumor regions. Delivery of the booster dose is performed using a multiport conformal beam protocol. For radiation beam shaping we used individual radiation blocks 3D-printed from tungsten infused ABS polymer. Calculation of beam geometry and the production of blocks is performed next to the EPR imager, immediately after oxygen imaging. Preliminary results demonstrate the sub-millimeter precision of the radiation delivery and high dose accuracy. The efficacy of the radiation treatment is currently being tested on syngeneic FSa fibrosarcoma tumors grown in the legs of C3H mice.


Assuntos
Fibrossarcoma/radioterapia , Neoplasias Musculares/radioterapia , Oxigênio/análise , Radioterapia Guiada por Imagem/métodos , Radioterapia de Intensidade Modulada/métodos , Hipóxia Tumoral/efeitos da radiação , Animais , Calibragem , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Espectroscopia de Ressonância de Spin Eletrônica/normas , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C3H , Neoplasias Musculares/metabolismo , Neoplasias Musculares/patologia , Oxigênio/metabolismo , Pressão Parcial , Dosagem Radioterapêutica , Radioterapia Guiada por Imagem/normas , Microtomografia por Raio-X
14.
Strahlenther Onkol ; 192(5): 279-87, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26961686

RESUMO

BACKGROUND: By studying the bioenergetic status we could show that the development of tumor hypoxia is accompanied, apart from myriad other biologically relevant effects, by a substantial accumulation of adenosine (ADO). ADO has been shown to act as a strong immunosuppressive agent in tumors by modulating the innate and adaptive immune system. In contrast to ADO, standard radiotherapy (RT) can either stimulate or abrogate antitumor immune responses. Herein, we present ADO-mediated mechanisms that may thwart antitumor immune responses elicited by RT. MATERIALS AND METHODS: An overview of the generation, accumulation, and ADO-related multifaceted inhibition of immune functions, contrasted with the antitumor immune effects of RT, is provided. RESULTS: Upon hypoxic stress, cancer cells release ATP into the extracellular space where nucleotides are converted into ADO by hypoxia-sensitive, membrane-bound ectoenzymes (CD39/CD73). ADO actions are mediated upon binding to surface receptors, mainly A2A receptors on tumor and immune cells. Receptor activation leads to a broad spectrum of strong immunosuppressive properties facilitating tumor escape from immune control. Mechanisms include (1) impaired activity of CD4 (+) T and CD8 (+) T, NK cells and dendritic cells (DC), decreased production of immuno-stimulatory lymphokines, and (2) activation of Treg cells, expansion of MDSCs, promotion of M2 macrophages, and increased activity of major immunosuppressive cytokines. In addition, ADO can directly stimulate tumor proliferation and angiogenesis. CONCLUSION: ADO mechanisms described can thwart antitumor immune responses elicited by RT. Therapeutic strategies alleviating tumor-promoting activities of ADO include respiratory hyperoxia or mild hyperthermia, inhibition of CD39/CD73 ectoenzymes or blockade of A2A receptors, and inhibition of ATP-release channels or ADO transporters.


Assuntos
Adenosina/imunologia , Imunidade Inata/imunologia , Fatores Imunológicos/imunologia , Neoplasias/imunologia , Neoplasias/radioterapia , Radioterapia/métodos , Sobrevivência Celular/imunologia , Sobrevivência Celular/efeitos da radiação , Medicina Baseada em Evidências , Humanos , Imunidade Inata/efeitos da radiação , Modelos Imunológicos , Resultado do Tratamento , Hipóxia Tumoral/imunologia , Hipóxia Tumoral/efeitos da radiação
15.
Eur J Nucl Med Mol Imaging ; 43(12): 2139-2146, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27251643

RESUMO

BACKGROUND AND PURPOSE: Increased tumour hypoxia is associated with a worse overall survival in patients with head and neck squamous cell carcinoma (HNSCC). The aims of this study were to evaluate treatment-associated changes in [18F]HX4-PET, hypoxia-related blood biomarkers, and their interdependence. MATERIAL AND METHODS: [18F]HX4-PET/CT scans of 20 patients with HNSCC were acquired at baseline and after ±20Gy of radiotherapy. Within the gross-tumour-volumes (GTV; primary and lymph nodes), mean and maximum standardized uptake values, the hypoxic fraction (HF) and volume (HV) were calculated. Also, the changes in spatial uptake pattern were evaluated using [18F]HX4-PET/CT imaging. For all patients, the plasma concentration of CAIX, osteopontin and VEGF was assessed. RESULTS: At baseline, tumour hypoxia was detected in 69 % (22/32) of the GTVs. During therapy, we observed a significant decrease in all image parameters. The HF decreased from 21.7 ± 19.8 % (baseline) to 3.6 ± 10.0 % (during treatment; P < 0.001). Only two patients had a HV > 1 cm3 during treatment, which was located for >98 % within the baseline HV. During treatment, no significant changes in plasma CAIX or VEGF were observed, while osteopontin was increased. CONCLUSIONS: [18F]HX4-PET/CT imaging allows monitoring changes in hypoxia during (chemo)radiotherapy whereas the blood biomarkers were not able to detect a treatment-associated decrease in hypoxia.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias de Cabeça e Pescoço/radioterapia , Imidazóis , Tomografia por Emissão de Pósitrons/métodos , Triazóis , Hipóxia Tumoral/efeitos da radiação , Idoso , Feminino , Neoplasias de Cabeça e Pescoço/sangue , Neoplasias de Cabeça e Pescoço/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Compostos Radiofarmacêuticos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Resultado do Tratamento
16.
Eur J Nucl Med Mol Imaging ; 43(12): 2147-2154, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27251644

RESUMO

PURPOSE: The purpose of this study was to prospectively investigate reoxygenation in the early phase of fractionated radiotherapy and serial changes of tumoricidal effects associated with intensity-modulated radiation therapy (IMRT) in patients with head and neck cancer (HNC) using F-18 fluoromisonidazole (FMISO) PET and F-18 fluorodeoxyglucose (FDG) PET. METHODS: Patients with untreated HNC underwent FMISO-PET and FDG-PET studies prospectively. A PET evaluation was conducted before each IMRT (Pre-IMRT), during IMRT (at 30 Gy/15 fr) (Inter-IMRT), and after completion of IMRT (70 Gy/35 fr) (Post-IMRT). FMISO-PET images were scanned by a PET/CT scanner at 4 h after the FMISO injection. We quantitatively analyzed the FMISO-PET images of the primary lesion using the maximum standardized uptake (SUVmax) and tumor-to-muscle ratio (TMR). The hypoxic volume (HV) was calculated as an index of tumor hypoxia, and was defined as the volume when the TMR was ≥ 1.25. Each FDG-PET scan was started 1 h after injection. The SUVmax and metabolic tumor volume (MTV) values obtained by FDG-PET were analyzed. RESULTS: Twenty patients finished the complete PET study protocol. At Pre-IMRT, 19 patients had tumor hypoxia in the primary tumor. In ten patients, the tumor hypoxia disappeared at Inter-IMRT. Another seven patients showed the disappearance of tumor hypoxia at Post-IMRT. Two patients showed tumor hypoxia at Post-IMRT. The FMISO-PET results showed that the reduction rates of both SUVmax and TMR from Pre-IMRT to Inter-IMRT were significantly higher than the corresponding reductions from Inter-IMRT to Post-IMRT (SUVmax: 27 % vs. 10 %, p = 0.025; TMR: 26 % vs. 12 %, p = 0.048). The reduction rate of SUVmax in FDG-PET from Pre-IMRT to Inter-IMRT was similar to that from Inter-IMRT to Post-IMRT (47 % vs. 48 %, p = 0.778). The reduction rate of the HV in FMISO-PET from Pre-IMRT to Inter-IMRT tended to be larger than that from Inter-IMRT to Post-IMRT (63 % vs. 40 %, p = 0.490). Conversely, the reduction rate of the MTV in FDG-PET from Pre-IMRT to Inter-IMRT was lower than that from Inter-IMRT to Post-IMRT (47 % vs. 74 %, p = 0.003). CONCLUSIONS: Both the intensity and the volume of tumor hypoxia rapidly decreased in the early phase of radiotherapy, indicating reoxygenation of the tumor hypoxia. In contrast, the FDG uptake declined gradually with the course of radiotherapy, indicating that the tumoricidal effect continues over the entire course of radiation treatment.


Assuntos
Fluordesoxiglucose F18/farmacocinética , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/radioterapia , Oxigênio/metabolismo , Radioterapia Conformacional/métodos , Hipóxia Tumoral/efeitos da radiação , Adulto , Idoso , Fracionamento da Dose de Radiação , Regulação para Baixo/efeitos da radiação , Feminino , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Humanos , Masculino , Pessoa de Meia-Idade , Misonidazol/análogos & derivados , Misonidazol/farmacocinética , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Resultado do Tratamento
18.
Semin Radiat Oncol ; 34(3): 323-336, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38880541

RESUMO

A large proportion of cancer patients present with unresectable bulky disease at baseline or following treatment failure. The data available in the literature suggest that the vast majority of these patients do not benefit from available standard therapies. Therefore the clinical outcomes are poor; patients are desperate and usually relegated to palliative or best supportive care as the only options. Large tumor masses are usually hypoxic, resistant to radiation and systemic therapy, with extensive regional infiltration of the surrounding critical organs, the presence of which makes it impossible to deliver a radical dose of radiation. Promising data in terms of improved therapeutic ratio where such complex tumors are concerned can be seen with the use of new emerging unconventional radiotherapy techniques known as spatially fractionated radiotherapies (SFRT). One of them is PATHY, or PArtial Tumor irradiation targeting HYpoxic segment, which is characterized by a very short treatment course offering a large spectrum of therapeutic benefits in terms of the symptom relief, quality of life, local tumor control, neoadjuvant and immunomodulatory effects.


Assuntos
Fracionamento da Dose de Radiação , Neoplasias , Humanos , Neoplasias/radioterapia , Qualidade de Vida , Hipóxia Tumoral/efeitos da radiação
19.
Cancer Res Commun ; 4(7): 1690-1701, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38904265

RESUMO

Tumor hypoxia has been shown to predict poor patient outcomes in several cancer types, partially because it reduces radiation's ability to kill cells. We hypothesized that some of the clinical effects of hypoxia could also be due to its impact on the tumor microbiome. Therefore, we examined the RNA sequencing data from the Oncology Research Information Exchange Network database of patients with colorectal cancer treated with radiotherapy. We identified microbial RNAs for each tumor and related them to the hypoxic gene expression scores calculated from host mRNA. Our analysis showed that the hypoxia expression score predicted poor patient outcomes and identified tumors enriched with certain microbes such as Fusobacterium nucleatum. The presence of other microbes, such as Fusobacterium canifelinum, predicted poor patient outcomes, suggesting a potential interaction between hypoxia, the microbiome, and radiation response. To experimentally investigate this concept, we implanted CT26 colorectal cancer cells into immune-competent BALB/c and immune-deficient athymic nude mice. After growth, in which tumors passively acquired microbes from the gastrointestinal tract, we harvested tumors, extracted nucleic acids, and sequenced host and microbial RNAs. We stratified tumors based on their hypoxia score and performed a metatranscriptomic analysis of microbial gene expression. In addition to hypoxia-tropic and -phobic microbial populations, analysis of microbial gene expression at the strain level showed expression differences based on the hypoxia score. Thus, hypoxia gene expression scores seem to associate with different microbial populations and elicit an adaptive transcriptional response in intratumoral microbes, potentially influencing clinical outcomes. SIGNIFICANCE: Tumor hypoxia reduces radiotherapy efficacy. In this study, we explored whether some of the clinical effects of hypoxia could be due to interaction with the tumor microbiome. Hypoxic gene expression scores associated with certain microbes and elicited an adaptive transcriptional response in others that could contribute to poor clinical outcomes.


Assuntos
Neoplasias Colorretais , Camundongos Endogâmicos BALB C , Camundongos Nus , Hipóxia Tumoral , Neoplasias Colorretais/radioterapia , Neoplasias Colorretais/microbiologia , Animais , Camundongos , Humanos , Hipóxia Tumoral/efeitos da radiação , Microbiota/efeitos da radiação , Linhagem Celular Tumoral , Feminino
20.
Int J Nanomedicine ; 19: 6499-6513, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38946887

RESUMO

Purpose: To address the problem of suboptimal reactive oxygen species (ROS) production in Radiation therapy (RT) which was resulted from exacerbated tumor hypoxia and the heterogeneous distribution of radiation sensitizers. Materials and Methods: In this work, a novel nanomedicine, designated as PLGA@IR780-Bi-DTPA (PIBD), was engineered by loading the radiation sensitizer Bi-DTPA and the photothermal agent IR780 onto poly(lactic-co-glycolic acid) (PLGA). This design leverages the tumor-targeting ability of IR780 to ensure selective accumulation of the nanoparticles in tumor cells, particularly within the mitochondria. The effect of the photothermal therapy-enhanced radiation therapy was also examined to assess the alleviation of hypoxia and the enhancement of radiation sensitivity. Results: The PIBD nanoparticles exhibited strong capacity in mitochondrial targeting and selective tumor accumulation. Upon activation by 808 nm laser irradiation, the nanoparticles effectively alleviated local hypoxia by photothermal effect enhanced blood supplying to improve oxygen content, thereby enhancing the ROS production for effective RT. Comparative studies revealed that PIBD-induced RT significantly outperformed conventional RT in treating hypoxic tumors. Conclusion: This design of tumor-targeting photothermal therapy-enhanced radiation therapy nanomedicine would advance the development of targeted drug delivery system for effective RT regardless of hypoxic microenvironment.


Assuntos
Nanopartículas , Terapia Fototérmica , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Espécies Reativas de Oxigênio , Animais , Terapia Fototérmica/métodos , Espécies Reativas de Oxigênio/metabolismo , Nanopartículas/química , Linhagem Celular Tumoral , Humanos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Camundongos , Indóis/farmacologia , Indóis/química , Hipóxia Tumoral/efeitos dos fármacos , Hipóxia Tumoral/efeitos da radiação , Radiossensibilizantes/farmacologia , Radiossensibilizantes/química , Camundongos Endogâmicos BALB C , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neoplasias/radioterapia , Neoplasias/terapia , Neoplasias/metabolismo , Nanomedicina
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa