Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Mais filtros

Medicinas Complementares
Tipo de documento
Intervalo de ano de publicação
1.
Stem Cells Transl Med ; 13(6): 546-558, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38457239

RESUMO

Human neural progenitor cells (hNPCs) hold promise for treating spinal cord injury. Studies to date have focused on improving their regenerative potential and therapeutic effect. Equally important is ensuring successful delivery and engraftment of hNPCs at the injury site. Unfortunately, no current imaging solution for cell tracking is compatible with long-term monitoring in vivo. The objective of this study was to apply a novel bright-ferritin magnetic resonance imaging (MRI) mechanism to track hNPC transplants longitudinally and on demand in the rat spinal cord. We genetically modified hNPCs to stably overexpress human ferritin. Ferritin-overexpressing (FT) hNPCs labeled with 0.2 mM manganese provided significant T1-induced bright contrast on in vitro MRI, with no adverse effect on cell viability, morphology, proliferation, and differentiation. In vivo, 2 M cells were injected into the cervical spinal cord of Rowett nude rats. MRI employed T1-weighted acquisitions and T1 mapping on a 3 T scanner. Conventional short-term cell tracking was performed using exogenous Mn labeling prior to cell transplantation, which displayed transient bright contrast on MRI 1 day after cell transplantation and disappeared after 1 week. In contrast, long-term cell tracking using bright-ferritin allowed on-demand signal recall upon Mn supplementation and precise visualization of the surviving hNPC graft. In fact, this new cell tracking technology identified 7 weeks post-transplantation as the timepoint by which substantial hNPC integration occurred. Spatial distribution of hNPCs on MRI matched that on histology. In summary, bright-ferritin provides the first demonstration of long-term, on-demand, high-resolution, and specific tracking of hNPCs in the rat spinal cord.


Assuntos
Rastreamento de Células , Ferritinas , Imageamento por Ressonância Magnética , Células-Tronco Neurais , Ratos Nus , Medula Espinal , Animais , Imageamento por Ressonância Magnética/métodos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/transplante , Células-Tronco Neurais/metabolismo , Rastreamento de Células/métodos , Humanos , Ratos , Ferritinas/metabolismo , Medula Espinal/metabolismo , Medula Espinal/diagnóstico por imagem , Transplante de Células-Tronco/métodos , Diferenciação Celular , Traumatismos da Medula Espinal/terapia
2.
Comp Med ; 71(4): 309-317, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34187631

RESUMO

Melatonin, the circadian nighttime neurohormone, and eicosapentaenoic acid (EPA) and docosahexaenoic acids (DHA), which are omega-3 fatty acids (FA) found in high concentrations in fish oil (FO) and plants, abrogate the oncogenic effects of linoleic acid (LA), an omega-6 FA, on the growth of rodent tumors and human breast, prostate, and head and neck squamous cell carcinoma (HNSCC) xenografts in vivo. Here we determined and compared the long-term effects of these inhibitory agents on tumor regression and LA uptake and metabolism to the mitogenic agent 13-[S]-hydroxyoctadecadienoic acid (13-[S]-HODE) in human prostate cancer 3 (PC3) and FaDu HNSCC xenografts in tumor-bearing male nude rats. Rats in this study were split into 3 groups and fed one of 2 diets: one diet containing 5% corn oil (CO, high LA), 5% CO oil and melatonin (2 µg/mL) or an alternative diet 5% FO (low LA). Rats whose diet contained melatonin had a faster rate of regression of PC3 prostate cancer xenografts than those receiving the FO diet, while both in the melatonin and FO groups induced the same rate of regression of HNSCC xenografts. The results also demonstrated that dietary intake of melatonin or FO significantly inhibited tumor LA uptake, cAMP content, 13-[S]-HODE formation, [³H]-thymidine incorporation into tumor DNA, and tumor DNA content. Therefore, long-term ingestion of either melatonin or FO can induce regression of PC3 prostate and HNSCC xenografts via a mechanism involving the suppression of LA uptake and metabolism by the tumor cells.


Assuntos
Melatonina , Neoplasias , Animais , Dieta , Xenoenxertos , Humanos , Ácido Linoleico , Ácidos Linoleicos , Masculino , Ratos , Ratos Nus
3.
Biomed Res Int ; 2021: 8875503, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33628824

RESUMO

In order to explore the specific mechanism of YiqiChutan formula (YQCTF) in inhibiting the angiogenesis of lung cancer and its relationship with delta-like ligand 4- (DLL4-) Notch signaling, 30 healthy BALB/c-nu/nu rats were selected and divided into three groups: A549 group (implanted with lung adenocarcinoma cell line A549), NCI-H460 group (implanted with human lung large-cell carcinoma cell line NCI-H460), and NCI-H446 group (implanted with human lung small cell carcinoma cell line NCI-H446) for constructing lung cancer transplanted tumor models. After modeling, the group treated with normal saline was taken as control group, 200 mg/kg of YQCTF was adopted for intervention, and the tumor volume and growth inhibition rate were compared with the vascular targeted inhibitor Sorafenib. HE staining, CD31 fluorescent antibody staining, and microelectron microscopy were adopted to observe the neovascular endothelial cells of the transplanted tumor. The expression of VEGF, HIF-1α, DLL4, and Notch-1 in the transplanted tumors in each group was detected by Western blot and RT-PCR at the protein level or mRNA level. Compared with the control group, the YQCTF-treated group had obvious inhibitory effect on lung cancer transplanted tumor and lung cancer angiogenesis. In the YQCTF-treated group, the density of angiogenesis decreased significantly and the vascular lumen structure also decreased, and the expression levels of VEGF, HIF-1α, DLL4, and Notch-1 in the YQCTF-treated group were all lower than those in the control group. YQCTF could inhibit the growth of lung cancer transplanted tumor through antiangiogenesis, and it could also reduce the amount of angiogenesis in lung cancer transplanted tumor. In addition, the generation of lumen structure was also hindered, which was realized through the VEGF signaling pathway and DLL4-Notch signaling pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenocarcinoma de Pulmão , Proteínas de Ligação ao Cálcio/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Neoplasias Pulmonares , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células A549 , Adenocarcinoma de Pulmão/irrigação sanguínea , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/metabolismo , Adenocarcinoma de Pulmão/patologia , Animais , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ratos , Ratos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Transl Res ; 229: 100-114, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33164812

RESUMO

Abundant intraperitoneal (IP) accumulation of extracellular mucus in patients with appendiceal mucinous carcinoma peritonei (MCP) causes compressive organ dysfunction and prevents delivery of chemotherapeutic drugs to cancer cells. We hypothesized that reducing extracellular mucus would decrease tumor-related symptoms and improve chemotherapeutic effect in patient-derived models of MCP. Mucolysis was achieved using a combination of bromelain (BRO) and N-acetylcysteine (NAC). Ex vivo experiments of mucolysis and chemotherapeutic drug delivery/effect were conducted with MCP and non-MCP tissue explants. In vivo experiments were performed in mouse and rat patient-derived xenograft (PDX) models of early and late (advanced) MCP. MCP tumor explants were less chemosensitive than non-MCP explants. Chronic IP administration of BRO + NAC in a mouse PDX model of early MCP and a rat PDX model of late (advanced) MCP converted solid mucinous tumors into mucinous ascites (mucolysis) that could be drained via a percutaneous catheter (rat model only), significantly reduced solid mucinous tumor growth and improved the efficacy of chemotherapeutic drugs. Combination of BRO + NAC efficiently lyses extracellular mucus in clinically relevant models of MCP. Conversion of solid mucinous tumors into mucinous ascites decreases tumor bulk and allows for minimally invasive drainage of liquified tumors. Lysis of extracellular mucus removes the protective mucinous coating surrounding cancer cells and improves chemotherapeutic drug delivery/efficacy in cancer cells. Our data provide a preclinical rationale for the clinical evaluation of BRO + NAC as a therapeutic strategy for MCP.


Assuntos
Adenocarcinoma Mucinoso/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias do Apêndice/tratamento farmacológico , Muco/efeitos dos fármacos , Neoplasias Peritoneais/tratamento farmacológico , Acetilcisteína/administração & dosagem , Acetilcisteína/farmacologia , Adenocarcinoma Mucinoso/patologia , Animais , Neoplasias do Apêndice/patologia , Bromelaínas/administração & dosagem , Bromelaínas/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Camundongos Nus , Neoplasias Peritoneais/patologia , Ratos Nus , Técnicas de Cultura de Tecidos/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Int J Hyperthermia ; 37(1): 101-109, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31969028

RESUMO

Objective: To establish the technique of intratumoral combination therapy of radiofrequency hyperthermia (RFH) with herpes simplex virus-thymidine kinase/ganciclovir (HSV-TK/GCV) gene therapy for rat ovarian cancers.Material and methods: This study consisted of three parts: (1) in vitro experiments to establish the 'proof of principal' that combination of RFH and HSV-TK gene therapy has the synergistic effect on human ovarian cancer cells; (2) creation of bioluminescence imaging-detectable rat ovarian cancer model; and (3) in vivo experiments using this rat model to validate the technical feasibility of the combination therapy. Cells and nude rats were divided into four groups: (i) combination therapy (HSV-TK/GCV + RFH); (ii) RFH; (iii) HSV-TK/GCV; and (iv) phosphate-buffered saline (PBS). Data were analyzed using Dunnett t-test or Kruskal-Wallis test.Results: Cell proliferation assay demonstrated significantly greater reduction in viable cells with the combination therapy [0.52 (0.43, 0.61)] compared to other treatments [RFH 0.90 (0.84, 0.96), HSV-TK/GCV 0.71 (0.53, 0.88), PBS 1 (1, 1); p < .05]. For 24 rat models with bioluminescence imaging-detectable orthotopic ovarian cancer (n = 6 per group), optical imaging demonstrated significantly decreased relative bioluminescence signal with the combination therapy [0.81 (0.52, 1.08)] compared to other treatments [RFH 3.60 (2.34, 4.86), HSV-TK/GCV 2.21 (1.71, 2.71), PBS 3.74 (3.19, 4.29); p < .001]. Ultrasound imaging demonstrated the smallest relative tumor volume with the combination therapy [0.78 (0.45, 1.11) versus 3.50 (2.67, 4.33), 2.10 (0.83, 3.37), 3.70 (1.79, 5.61); p < .05].Conclusion: The feasibility of intratumoral RFH-enhanced HSV-TK/GCV gene therapy was established on a unique rat model with molecular imaging-detectable orthotopic ovarian cancer.


Assuntos
Terapia Genética/métodos , Hipertermia Induzida/métodos , Imagem Molecular/métodos , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/radioterapia , Simplexvirus/efeitos dos fármacos , Timidina Quinase/uso terapêutico , Animais , Feminino , Humanos , Ratos , Ratos Nus , Timidina Quinase/farmacologia
6.
Clin Exp Metastasis ; 36(6): 511-518, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31541325

RESUMO

Pseudomyxoma peritonei (PMP) is a rare cancer commonly originating from appendiceal neoplasms that presents with mucinous tumor spread in the peritoneal cavity. Patients with PMP are treated with curative intent by cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). The value of adding HIPEC to CRS has not been proven in randomized trials, and the objective of this study was to investigate the efficacy of intraperitoneal mitomycin C (MMC) and regional hyperthermia as components of this complex treatment. Xenograft tissue established from a patient with histologically high-grade PMP with signet ring cell differentiation was implanted intraperitoneally in 65 athymic nude male rats and the animals were stratified into three treatment groups; the cytoreductive surgery group (CRSG, CRS only), the normothermic group (NG, CRS and intraperitoneal chemotherapy perfusion (IPEC) with MMC at 35 ºC), and the hyperthermic group (HG, CRS and IPEC at 41 ºC). The main endpoints were survival and tumor weight at autopsy. Adequate imitation of the clinical setting and treatment approach was achieved. The median survival was 31 days in the CRSG, 60 days in NG and 67 days in HG. The median tumor weights at autopsy were 34 g in CRSG, 23 g NG and 20 g in HG. In conclusion, the addition of IPEC with MMC after CRS doubled the survival time and reduced tumor growth compared to CRS alone. Adding regional hyperthermia resulted in a modest improvement of treatment outcome.


Assuntos
Cistadenocarcinoma Mucinoso/mortalidade , Procedimentos Cirúrgicos de Citorredução/mortalidade , Hipertermia Induzida/mortalidade , Mitomicina/administração & dosagem , Neoplasias Peritoneais/mortalidade , Pseudomixoma Peritoneal/mortalidade , Animais , Antibióticos Antineoplásicos/administração & dosagem , Terapia Combinada , Cistadenocarcinoma Mucinoso/patologia , Cistadenocarcinoma Mucinoso/terapia , Feminino , Humanos , Injeções Intraperitoneais , Masculino , Camundongos Endogâmicos BALB C , Modelos Teóricos , Neoplasias Peritoneais/secundário , Neoplasias Peritoneais/terapia , Pseudomixoma Peritoneal/patologia , Pseudomixoma Peritoneal/terapia , Ratos Nus , Taxa de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1126-1127: 121734, 2019 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-31401450

RESUMO

Non-small cell lung cancer (NSCLC), as the most common histological type of lung cancer with high mortality rates, has been widely treated by Compound Kushen Injection (CKI) in China. Among various active components with many pharmacologic properties, matrine, oxymatrine, sophoridine, oxysophocarpine and N-methylcytisine, are the main bioactive alkaloids of CKI. Therefore, a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed to simultaneously quantify the five alkaloids in nude rat plasma in this study. With aminopyrine as internal standard (IS), plasma samples were extracted by alkalified chloroform and then separated on an Agela Venusil MP C18 column (2.1 mm × 100 mm, 3 µm) using gradient elution. Water containing 10 mmol/L ammonium acetate (adjusted to pH 8.0 with ammonia water, A) and methanol (B) constituted the mobile phase system. Notably, the analysis was rapid and accurate due to a short running time of 6 min and a stereoisomer separation between matrine and sophoridine. Detection was implemented in MRM mode with an electrospray positive ionization source. Validation parameters were all in accordance with current criterion. The established method has been effectively employed to compare the pharmacokinetic behaviors of five alkaloids between normal and NSCLC nude rats. Results indicated that the pharmacokinetic behaviors of oxymatrine, sophoridine and N-methylcytisine from CKI could be changed when it was intravenously administrated to the NSCLC model rats, and possible reasons have been proposed. This study could provide meaningful reference for further clinical medication of CKI when combined with time-effect curve and clinical symptoms.


Assuntos
Alcaloides/sangue , Alcaloides/farmacocinética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Medicamentos de Ervas Chinesas/farmacocinética , Neoplasias Pulmonares/metabolismo , Alcaloides/química , Animais , Cromatografia Líquida/métodos , Medicamentos de Ervas Chinesas/administração & dosagem , Limite de Detecção , Modelos Lineares , Masculino , Ratos , Ratos Nus , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem/métodos
8.
Nat Biotechnol ; 37(8): 895-906, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31375810

RESUMO

The epicardium and its derivatives provide trophic and structural support for the developing and adult heart. Here we tested the ability of human embryonic stem cell (hESC)-derived epicardium to augment the structure and function of engineered heart tissue in vitro and to improve efficacy of hESC-cardiomyocyte grafts in infarcted athymic rat hearts. Epicardial cells markedly enhanced the contractility, myofibril structure and calcium handling of human engineered heart tissues, while reducing passive stiffness compared with mesenchymal stromal cells. Transplanted epicardial cells formed persistent fibroblast grafts in infarcted hearts. Cotransplantation of hESC-derived epicardial cells and cardiomyocytes doubled graft cardiomyocyte proliferation rates in vivo, resulting in 2.6-fold greater cardiac graft size and simultaneously augmenting graft and host vascularization. Notably, cotransplantation improved systolic function compared with hearts receiving either cardiomyocytes alone, epicardial cells alone or vehicle. The ability of epicardial cells to enhance cardiac graft size and function makes them a promising adjuvant therapeutic for cardiac repair.


Assuntos
Coração/fisiologia , Células-Tronco Embrionárias Humanas , Infarto do Miocárdio/terapia , Miócitos Cardíacos , Regeneração , Animais , Embrião de Galinha , Regulação da Expressão Gênica , Humanos , Masculino , Ratos , Ratos Nus , Ratos Sprague-Dawley , Engenharia Tecidual
9.
J Pineal Res ; 67(2): e12586, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31077613

RESUMO

Disruption of circadian time structure and suppression of circadian nocturnal melatonin (MLT) production by exposure to dim light at night (dLAN), as occurs with night shift work and/or disturbed sleep-wake cycles, is associated with a significantly increased risk of breast cancer and resistance to tamoxifen and doxorubicin. Melatonin inhibition of human breast cancer chemoresistance involves mechanisms including suppression of tumor metabolism and inhibition of kinases and transcription factors which are often activated in drug-resistant breast cancer. Signal transducer and activator of transcription 3 (STAT3), frequently overexpressed and activated in paclitaxel (PTX)-resistant breast cancer, promotes the expression of DNA methyltransferase one (DNMT1) to epigenetically suppress the transcription of tumor suppressor Aplasia Ras homolog one (ARHI) which can sequester STAT3 in the cytoplasm to block PTX resistance. We demonstrate that breast tumor xenografts in rats exposed to dLAN and circadian MLT disrupted express elevated levels of phosphorylated and acetylated STAT3, increased DNMT1, but reduced sirtuin 1 (SIRT1) and ARHI. Furthermore, MLT and/or SIRT1 administration blocked/reversed interleukin 6 (IL-6)-induced acetylation of STAT3 and its methylation of ARH1 to increase ARH1 mRNA expression in MCF-7 breast cancer cells. Finally, analyses of the I-SPY 1 trial demonstrate that elevated MT1 receptor expression is significantly correlated with pathologic complete response following neo-adjuvant therapy in breast cancer patients. This is the first study to demonstrate circadian disruption of MLT by dLAN driving intrinsic resistance to PTX via epigenetic mechanisms increasing STAT3 expression and that MLT administration can reestablish sensitivity of breast tumors to PTX and drive tumor regression.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Melatonina/farmacologia , Paclitaxel/farmacologia , Fator de Transcrição STAT3/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Proteínas rho de Ligação ao GTP/biossíntese , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ritmo Circadiano/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Ratos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Int J Mol Sci ; 19(12)2018 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-30513935

RESUMO

Despite the similar phenotypes, including weight loss, reduction of food intake, and lower adiposity, associated with caloric restriction (CR) and cancer cachexia (CC), CC is a progressive wasting syndrome, while mild CR improves whole body metabolism. In the present study, we compared adipose metabolic changes in a novel rat model of CC, mild CR (70% of the food intake of control rats, which is similar to the food consumption of CC rats), and severe CR (30% of the food intake of controls). We show that CC and severe CR are associated with much smaller adipocytes with significantly lower mitochondrial DNA content; but, that mild CR is not. CC and both mild and severe CR similarly upregulated proteins involved in lipolysis. CC also downregulated proteins involved in fatty acid biosynthesis, but mild CR upregulated these. These findings suggest that CC might impair de novo fatty acid biosynthesis and reduce mitochondrial biogenesis, similar to severe CR. We also found that rikkunshito, a traditional Japanese herbal medicine, does not ameliorate the enhanced lipolysis and mitochondrial impairment, but rather, rescues de novo fatty acid biosynthesis, suggesting that rikkunshito administration might have partially similar effects to mild CR.


Assuntos
Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Caquexia/complicações , Caquexia/tratamento farmacológico , Restrição Calórica , Medicamentos de Ervas Chinesas/uso terapêutico , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Adipócitos/efeitos dos fármacos , Adipócitos/patologia , Tecido Adiposo/efeitos dos fármacos , Animais , Atrofia , Caquexia/genética , Caquexia/patologia , Tamanho Celular/efeitos dos fármacos , DNA Mitocondrial/genética , Medicamentos de Ervas Chinesas/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neoplasias/genética , Neoplasias/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Nus , Ratos Wistar
11.
Sci Rep ; 8(1): 11417, 2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-30061692

RESUMO

Most applications of nanotechnology in cancer have focused on systemic delivery of cytotoxic drugs. Systemic delivery relies on accumulation of nanoparticles in a target tissue through enhanced permeability of leaky vasculature and retention effect of poor lymphatic drainage to increase the therapeutic index. Systemic delivery is limited, however, by toxicity and difficulty crossing natural obstructions, like the blood spine barrier. Magnetic drug targeting (MDT) is a new technique to reach tumors of the central nervous system. Here, we describe a novel therapeutic approach for high-grade intramedullary spinal cord tumors using magnetic nanoparticles (MNP). Using biocompatible compounds to form a superparamagnetic carrier and magnetism as a physical stimulus, MNP-conjugated with doxorubicin were successfully localized to a xenografted tumor in a rat model. This study demonstrates proof-of-concept that MDT may provide a novel technique for effective, concentrated delivery of chemotherapeutic agents to intramedullary spinal cord tumors without the toxicity of systemic administration.


Assuntos
Sistemas de Liberação de Medicamentos , Magnetismo , Neoplasias da Medula Espinal/terapia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Humanos , Nanopartículas de Magnetita/química , Ratos Nus , Neoplasias da Medula Espinal/tratamento farmacológico , Neoplasias da Medula Espinal/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Radiology ; 288(2): 612-620, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29893649

RESUMO

Purpose To validate the feasibility and efficacy of intratumoral radiofrequency hyperthermia (RFH)-enhanced herpes simplex virus (HSV) thymidine kinase (TK) and ganciclovir (GCV) (hereafter, HSV-TK/GCV) gene therapy for non-small-cell lung cancer (NSCLC). Materials and Methods This study was performed from November 11, 2015, to April 14, 2017, and included (a) in vitro experiments with human NSCLC cells to establish the proof of principle, (b) in vivo experiments using mice with subcutaneous NSCLC to further demonstrate the principle, and (c) in vivo experiments using rats with orthotopic NSCLC to validate the technical feasibility. Cells, nude mice, and nude rats were randomly divided into four groups (six animals per group): (a) combination therapy (HSV-TK/GCV combined with RFH), (b) RFH, (c) HSV-TK/GCV, and (d) phosphate-buffered saline. Data were analyzed by using the Dunnett t test or Kruskal-Wallis test. Results For in vitro experiments, the cell proliferation assay showed significantly diminished viable cells with combination therapy (mean, 0.56; 95% confidence interval [CI]: 0.44, 0.68) versus RFH (mean, 0.89; 95% CI: 0.82, 0.97), HSV-TK/GCV (mean, 0.71; 95% CI: 0.56, 0.86), and phosphate-buffered saline (mean, 1; 95% CI: 1, 1) (P < .05 for all). For in vivo experiments, optical imaging showed significantly decreased relative bioluminescence signal with combination therapy (mean, 0.71 [95% CI: 0.03, 1.39] in mice; 1.29 [95% CI: 0.51, 2.06] in rats) compared with RFH (mean, 2.66 [95% CI: 1.73, 3.59] in mice; 2.26 [95% CI: 1.51, 3.01] in rats), HSV-TK/GCV (mean, 1.37 [95% CI: 0.65, 2.08] in mice; 1.76 [95% CI: 1.20, 2.31] in rats), and phosphate-buffered saline (mean, 3.07 [95% CI: 2.50, 3.65] in mice; 2.94 [95% CI: 2.29, 3.58] in rats) (P < .001 for all). US showed that the smallest relative tumor volumes occurred with combination therapy (mean, 0.60; 95% CI: 0.15, 1.05) versus RFH (mean, 2.43; 95% CI: 1.80, 3.06), HSV-TK/GCV (mean, 1.32; 95% CI: 0.75, 1.89), and phosphate-buffered saline (mean, 2.56; 95% CI: 1.75, 3.38) (P < .05 for all) in the mouse subcutaneous model. Conclusion Intratumoral radiofrequency hyperthermia-enhanced herpes simplex virus thymidine kinase and ganciclovir gene therapy for non-small-cell lung cancer is feasible and can be guided by molecular imaging. © RSNA, 2018.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/terapia , Ganciclovir/uso terapêutico , Terapia Genética/métodos , Hipertermia Induzida/métodos , Neoplasias Pulmonares/terapia , Timidina Quinase/genética , Animais , Antivirais/uso terapêutico , Estudos de Viabilidade , Feminino , Humanos , Técnicas In Vitro , Camundongos , Camundongos Nus , Imagem Molecular , Ratos , Ratos Nus , Reprodutibilidade dos Testes , Simplexvirus/enzimologia , Resultado do Tratamento , Células Tumorais Cultivadas
13.
Mol Pharm ; 15(9): 3634-3641, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-29450993

RESUMO

Near infrared photoimmunotherapy (NIR-PIT) is a new target-cell-specific cancer treatment that induces highly selective necrotic/immunogenic cell death after systemic administration of a photoabsorber antibody conjugate and subsequent NIR light exposure. However, the depth of NIR light penetration in tissue (approximately 2 cm) with external light sources limits the therapeutic effects of NIR-PIT. Interstitial light exposure using cylindrical diffusing optical fibers can overcome this limitation. The purpose in this study was to compare three NIR light delivery methods for treating tumors with NIR-PIT using a NIR laser system at an identical light energy; external exposure alone, interstitial exposure alone, and the combination. Panitumumab conjugated with the photoabsorber IRDye-700DX (pan-IR700) was intravenously administered to mice with A431-luc xenografts which are epithelial growth factor receptor (EGFR) positive. One and 2 days later, NIR light was administered to the tumors using one of three methods. Interstitial exposure alone and in combination with external sources showed the greatest decrease in bioluminescence signal intensity. Additionally, the combination of external and interstitial NIR light exposure showed significantly greater tumor size reduction and prolonged survival after NIR-PIT compared to external exposure alone. This result suggested that the combination of external and interstitial NIR light exposure was more effective than externally applied light alone. Although external exposure is the least invasive means of delivering light, the combination of external and interstitial exposures produces superior therapeutic efficacy in tumors greater than 2 cm in depth from the tissue surface.


Assuntos
Imunoterapia/métodos , Luz , Fototerapia/métodos , Animais , Carcinoma de Células Escamosas/diagnóstico por imagem , Carcinoma de Células Escamosas/tratamento farmacológico , Receptores ErbB/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Imagem Óptica , Panitumumabe/uso terapêutico , Ratos Nus
14.
ChemMedChem ; 12(21): 1776-1793, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-28961375

RESUMO

Selective inhibition of exclusively transcription-regulating PTEFb/CDK9 is a promising new approach in cancer therapy. Starting from lead compound BAY-958, lead optimization efforts strictly focusing on kinase selectivity, physicochemical and DMPK properties finally led to the identification of the orally available clinical candidate atuveciclib (BAY 1143572). Structurally characterized by an unusual benzyl sulfoximine group, BAY 1143572 exhibited the best overall profile in vitro and in vivo, including high efficacy and good tolerability in xenograft models in mice and rats. BAY 1143572 is the first potent and highly selective PTEFb/CDK9 inhibitor to enter clinical trials for the treatment of cancer.


Assuntos
Quinase 9 Dependente de Ciclina/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Sulfonamidas/uso terapêutico , Triazinas/uso terapêutico , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Quinase 9 Dependente de Ciclina/metabolismo , Meia-Vida , Células HeLa , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Camundongos , Camundongos Nus , Conformação Molecular , Simulação de Acoplamento Molecular , Neoplasias/patologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/toxicidade , Estrutura Terciária de Proteína , Ratos , Ratos Nus , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/toxicidade , Transplante Heterólogo , Triazinas/química , Triazinas/toxicidade
15.
Theranostics ; 7(2): 436-451, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28255341

RESUMO

Despite recent progress in conventional therapeutic approaches, the vast majority of glioblastoma recur locally, indicating that a more aggressive local therapy is required. Interstitial photodynamic therapy (iPDT) appears as a very promising and complementary approach to conventional therapies. However, an optimal fractionation scheme for iPDT remains the indispensable requirement. To achieve that major goal, we suggested following iPDT tumor response by a non-invasive imaging monitoring. Nude rats bearing intracranial glioblastoma U87MG xenografts were treated by iPDT, just after intravenous injection of AGuIX® nanoparticles, encapsulating PDT and imaging agents. Magnetic Resonance Imaging (MRI) and Magnetic Resonance Spectroscopy (MRS) allowed us an original longitudinal follow-up of post-treatment effects to discriminate early predictive markers. We successfully used conventional MRI, T2 star (T2*), Diffusion Weighted Imaging (DWI) and MRS to extract relevant profiles on tissue cytoarchitectural alterations, local vascular disruption and metabolic information on brain tumor biology, achieving earlier assessment of tumor response. From one day post-iPDT, DWI and MRS allowed us to identify promising markers such as the Apparent Diffusion Coefficient (ADC) values, lipids, choline and myoInositol levels that led us to distinguish iPDT responders from non-responders. All these responses give us warning signs well before the tumor escapes and that the growth would be appreciated.


Assuntos
Monitoramento de Medicamentos/métodos , Glioblastoma/diagnóstico , Glioblastoma/terapia , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética/métodos , Fotoquimioterapia , Prótons , Animais , Meios de Contraste/administração & dosagem , Modelos Animais de Doenças , Xenoenxertos , Estudos Longitudinais , Nanopartículas/administração & dosagem , Fármacos Fotossensibilizantes/administração & dosagem , Ratos Nus , Resultado do Tratamento
16.
Radiology ; 282(1): 103-112, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27404050

RESUMO

Purpose To determine the feasibility of using intraesophageal radiofrequency (RF) hyperthermia to enhance local chemotherapy in a rat model with orthotopic esophageal squamous cancers. Materials and Methods The animal protocol was approved by the institutional animal care and use committee and the institutional review board. Human esophageal squamous cancer cells were transduced with luciferase lentiviral particles. Cancer cells, mice with subcutaneous cancer esophageal xenografts, and nude rats with orthotopic esophageal cancers in four study groups of six animals per group were treated with (a) combination therapy of magnetic resonance imaging heating guidewire-mediated RF hyperthermia (42°C) plus local chemotherapy (cisplatin and 5-fluorouracil), (b) chemotherapy alone, (c) RF hyperthermia alone, and (d) phosphate-buffered saline. Bioluminescent optical imaging and transcutaneous ultrasonographic imaging were used to observe bioluminescence signal and changes in tumor size among the groups over 2 weeks, which were correlated with subsequent histologic results. The nonparametric Mann-Whitney U test was used for comparisons of variables. Results Compared with chemotherapy alone, RF hyperthermia alone, and phosphate-buffered saline, combination therapy with RF hyperthermia and chemotherapy induced the lowest cell proliferation (relative absorbance of formazan: 23.4% ± 7, 44.6% ± 7.5, 95.8% ± 2, 100%, respectively; P < .0001), rendered the smallest relative tumor volume (0.65 mm3 ± 0.15, P < .0001) and relative bioluminescence optical imaging photon signal (0.57 × 107 photons per second per square millimeter ± 0.15, P < .001) of mice with esophageal cancer xenografts, as well as the smallest relative tumor volume (0.68 mm3 ± 0.13, P < .05) and relative photon signal (0.56 × 107 photons per second per square millimeter ± 0.11. P < .001) of rat orthotopic esophageal cancers. Conclusion Intraesophageal RF hyperthermia can enhance the effect of chemotherapy on esophageal squamous cell cancers. © RSNA, 2016.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/terapia , Neoplasias Esofágicas/terapia , Hipertermia Induzida/métodos , Animais , Apoptose , Carcinoma de Células Escamosas/diagnóstico por imagem , Terapia Combinada , Modelos Animais de Doenças , Neoplasias Esofágicas/diagnóstico por imagem , Carcinoma de Células Escamosas do Esôfago , Xenoenxertos , Imageamento por Ressonância Magnética , Camundongos Nus , Microscopia Confocal , Ratos , Ratos Nus , Taxa de Sobrevida , Carga Tumoral , Células Tumorais Cultivadas , Ultrassonografia
17.
Asian J Androl ; 19(2): 203-207, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27270338

RESUMO

The objective is to develop an easier technique for regenerating corpora cavernosa tissue through transplantation of human bone marrow-derived CD133 + cells into a rat corpora cavernosa defect model. We excised 2 mm × 2 mm squares of the right corpora cavernosa of twenty-three 8-week-old male nude rats. Alginate gel sponge sheets supplemented with 1 × 10 4 CD133 + cells were then placed over the excised area of nine rats. Functional and histological evaluations were carried out 8 weeks later. The mean intracavernous pressure/mean arterial pressure ratio for the nine rats (0.34258 ± 0.0831) was significantly higher than that for eight rats with only the excision (0.0580 ± 0.0831, P = 0.0238) and similar to that for five rats for which the penis was exposed, and there was no excision (0.37228 ± 0.1051, P = 0.8266). Immunohistochemical analysis revealed that the nine fully treated rats had venous sinus-like structures and quantitative reverse transcription polymerase chain reaction analysis of extracts from their alginate gel sponge sheets revealed that the amounts of mRNA encoding the nerve growth factor (NGF), and vascular endothelial growth factor (VEGF) were significantly higher than those for rats treated with alginate gel sheets without cell supplementation (NGF: P = 0.0309; VEGF: P < 0.0001). These findings show that transplantation of CD133 + cells accelerates functional and histological recovery in the corpora cavernosa defect model.


Assuntos
Alginatos/uso terapêutico , Transplante de Medula Óssea/métodos , Géis/uso terapêutico , Ereção Peniana , Pênis/patologia , Regeneração , Antígeno AC133/metabolismo , Implantes Absorvíveis , Actinas/metabolismo , Animais , Células da Medula Óssea/metabolismo , Ácido Glucurônico/uso terapêutico , Ácidos Hexurônicos/uso terapêutico , Humanos , Masculino , Fator de Crescimento Neural/genética , Pênis/metabolismo , Ratos , Ratos Nus , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética
18.
Tissue Eng Part A ; 22(13-14): 985-93, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27369635

RESUMO

Here we present a new model for alveolar jaw bone regeneration, which uses human dental pulp cells (hDPCs) combined with tyrosine-derived polycarbonate polymer scaffolds [E1001(1k)] containing beta-tricalcium phosphate (ß-TCP) [E1001(1k)/ß-TCP]. E1001(1k)/ß-TCP scaffolds (5 mm diameter × 1 mm thickness) were fabricated to fit a 5 mm rat mandibular ramus critical bone defect. Five experimental groups were examined in this study: (1) E1001(1k)/ß-TCP scaffolds seeded with a high density of hDPCs, 5.0 × 10(5) hDPCs/scaffold (CH); (2) E1001(1k)/ß-TCP scaffolds seeded with a lower density of hDPCs, 2.5 × 10(5) hDPCs/scaffold (CL); (3) acellular E1001(1k)/ß-TCP scaffolds (SA); (4) acellular E1001(1k)/ß-TCP scaffolds supplemented with 4 µg recombinant human bone morphogenetic protein-2 (BMP); and (5) empty defects (EDs). Replicate hDPC-seeded and acellular E1001(1k)/ß-TCP scaffolds were cultured in vitro in osteogenic media for 1 week before implantation for 3 and 6 weeks. Live microcomputed tomography (µCT) imaging at 3 and 6 weeks postimplantation revealed robust bone regeneration in the BMP implant group. CH and CL groups exhibited similar uniformly distributed mineralized tissue coverage throughout the defects, but less than the BMP implants. In contrast, SA-treated defects exhibited sparse areas of mineralized tissue regeneration. The ED group exhibited slightly reduced defect size. Histological analyses revealed no indication of an immune response. In addition, robust expression of dentin and bone differentiation marker expression was observed in hDPC-seeded scaffolds, whereas, in contrast, BMP and SA implants exhibited only bone and not dentin differentiation marker expression. hDPCs were detected in 3-week but not in 6-week hDPC-seeded scaffold groups, indicating their survival for at least 3 weeks. Together, these results show that hDPC-seeded E1001(1k)/ß-TCP scaffolds support the rapid regeneration of osteo-dentin-like mineralized jaw tissue, suggesting a promising new therapy for alveolar jaw bone repair and regeneration.


Assuntos
Regeneração Óssea , Polpa Dentária/metabolismo , Mandíbula/metabolismo , Traumatismos Mandibulares/terapia , Cimento de Policarboxilato/química , Transplante de Células-Tronco , Células-Tronco/metabolismo , Alicerces Teciduais/química , Animais , Feminino , Humanos , Mandíbula/patologia , Traumatismos Mandibulares/metabolismo , Traumatismos Mandibulares/patologia , Osteogênese , Ratos , Ratos Nus , Tirosina/química
19.
Pancreatology ; 16(4): 632-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27289344

RESUMO

OBJECTIVE: The origin of pancreatic cancer has been identified as a population of malignant pancreatic stem cells CD133+ CXCR4+ immunophenotype. These cells have high capacity for early locoregional invasion, being responsible for early recurrence and high mortality rates of pancreatic cancer. We propose a study for decreasing tumor progression of pancreatic cancer by reducing the volume and neoplastic subpopulation of pancreatic cancer stem cells CD133+ CXCR4+. Therefore, we develop a new therapeutic model, characterized by the application of HIPEC (Hyperthermic Intraperitoneal Chemotherapy) with gemcitabine. DESIGN: Pancreatic tumor cell line: human cell line BxPC-3. The animal model involved 18 immunosuppressed rats 5 weeks weighing 150-200 gr. The implantation of 13 × 10(6) cells/mL was performed with homogeneous distribution in the 13 abdominopelvic quadrants according to the peritoneal carcinomatosis index (PCI) and were randomized into three treatment groups. Group I (4 rats) received intravenous saline. Group II (6 rats) received intravenous gemcitabine. Group III (8 rats) received HIPEC at 41 °C for 30 min with gemcitabine + gemcitabine IV. A histological study confirmed pancreatic cancer and immunohistochemical quantification of pancreatic cancer stem cells CD133+ CXCR4+ tumor cells. RESULTS: There was a population decline of pancreatic cancer stem cells CD133+ CXCR4+ in the HIPEC group with respect to the other two groups (p < 0.001). There was a decrease in PCI between treatment groups (p < 0.05). CONCLUSION: The initial results are encouraging since there is a declining population of cancer stem cells CD133+ CXCR4+ in the HIPEC group and decreased tumor volume compared to the other two treatment groups. All the conclusions are only valid for BxPC3 cell line, and the effects HIPEC on Kras-driven pancreatic tumors remain to be determined.


Assuntos
Antígeno AC133/imunologia , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma/tratamento farmacológico , Desoxicitidina/análogos & derivados , Hipertermia Induzida/métodos , Células-Tronco Neoplásicas/patologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Receptores CXCR4/imunologia , Animais , Linhagem Celular Tumoral , Terapia Combinada , Desoxicitidina/administração & dosagem , Desoxicitidina/uso terapêutico , Progressão da Doença , Humanos , Injeções Intraperitoneais , Masculino , Transplante de Neoplasias , Neoplasias Pancreáticas/patologia , Ratos , Ratos Nus , Gencitabina
20.
PLoS One ; 11(6): e0158005, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27332712

RESUMO

Clinical studies using definitive-intent stereotactic radiation therapy (SRT) for the local treatment of canine osteosarcoma (OSA) have shown canine patients achieving similar median survival times as the current standard of care (amputation and adjuvant chemotherapy). Despite this, there remains an unacceptable high risk of pathologic fracture following radiation treatment. Zoledronic acid (ZA) and parathyroid hormone (PTH) are therapeutic candidates for decreasing this fracture risk post-irradiation. Due to differing mechanisms, we hypothesized that the combined treatment with ZA and PTH would significantly improve bone healing more than ZA or PTH treatment alone. Using an orthotopic model of canine osteosarcoma in athymic rats, we evaluated bone healing following clinically-relevant doses of radiation therapy (12 Gy x 3 fractions, 36 Gy total). Groups included 36 Gy SRT only, 36 Gy SRT plus ZA, 36 Gy SRT plus ZA and PTH, 36 Gy SRT plus PTH, and 36 Gy SRT plus localized PTH treatment. Our study showed significant increases in bone volume and increased polar moments of inertia (in the distal femoral metaphysis) 8 weeks after radiation in the combined (ZA/PTH) treatment group as compared to radiation treatment alone. Histomorphometric analysis revealed evidence of active mineralization at the study endpoint as well as successful tumor-cell kill across all treatment groups. This work provides further evidence for the expanding potential indications for ZA and PTH therapy, including post-irradiated bone disease due to osteosarcoma.


Assuntos
Osso e Ossos/patologia , Difosfonatos/uso terapêutico , Imidazóis/uso terapêutico , Osteossarcoma/tratamento farmacológico , Osteossarcoma/radioterapia , Hormônio Paratireóideo/uso terapêutico , Técnicas Estereotáxicas , Animais , Osso e Ossos/diagnóstico por imagem , Calcificação Fisiológica , Terapia Combinada , Cães , Relação Dose-Resposta à Radiação , Quimioterapia Combinada , Feminino , Luminescência , Ratos Nus , Fosfatase Ácida Resistente a Tartarato/metabolismo , Fatores de Tempo , Microtomografia por Raio-X , Ácido Zoledrônico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA