Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Med ; 23(6): 2487-2502, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36764998

RESUMEN

Rhabdomyosarcoma (RMS) is an aggressive rare neoplasm that derives from mesenchymal cells, which frequently develops resistance to the current therapies and the formation of metastases. Thus, new therapies are needed. The alteration of iron metabolism in cancer cells was effective in reducing the progression of many tumors but not yet investigated in RMS. Here we investigated the effect of iron modulation in RMS both in vitro and in vivo. We first characterized the most used RMS cell lines representing the most common subtypes, embryonal (ERMS, RD cells) and alveolar (ARMS, RH30 cells), for their iron metabolism, in basal condition and in response to its modulation. Then we investigated the effects of both iron overload and chelation strategies in vitro and in vivo. RMS cell lines expressed iron-related proteins, even if at lower levels compared to hepatic cell lines and they are correctly modulated in response to iron increase and deprivation. Interestingly, the treatment with different doses of ferric ammonium citrate (FAC, as iron source) and with deferiprone (DFP, as iron chelator), significantly affected the cell viability of RD and RH30. Moreover, iron supplementation (in the form of iron dextran) or iron chelation (in the form of DFP) were also effective in vivo in inhibiting the tumor mass growth both derived from RD and RH30 with iron chelation treatment the most effective one. All the data suggest that the iron modulation could be a promising approach to overcome the RMS tumor growth. The mechanism of action seems to involve the apoptotic cell death for both iron supplementation and chelation with the concomitant induction of ferroptosis in the case of iron supplementation.


Asunto(s)
Rabdomiosarcoma , Humanos , Línea Celular Tumoral , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Apoptosis , Hierro , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico
2.
Pediatr Blood Cancer ; 69(11): e29864, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35731577

RESUMEN

BACKGROUND: Cytoreductive surgery (CRS) in combination with hyperthermic intraperitoneal chemotherapy (HIPEC) is an option in advanced peritoneal sarcomatosis. Nevertheless, CRS and HIPEC are not successful in all patients. An enhancement of HIPEC using photodynamic therapy (PDT) might be beneficial. Therefore, a combination of the photosensitizer hypericin (HYP) with HIPEC was evaluated in an animal model. PROCEDURE: An established HIPEC animal model for rhabdomyosarcoma (NOD/LtSz-scid IL2Rγnullmice, n = 80) was used. All groups received HYP (100 µg/200 µl) intraperitoneally with and without cisplatin-based (30 or 60 mg/m2 ) HIPEC (37°C or 42°C, for 60 minutes) (five groups, each n = 16). Peritoneal cancer index (PCI) was documented visually and by HYP-based photodynamic diagnosis (PDD). HYP-based PDT of the tumor was performed. Tissue samples were evaluated regarding proliferation (Ki-67) and apoptosis (TUNEL). RESULTS: HYP uptake was detected even in smallest tumor nodes (<1 mm) with improved tumor detection during PDD (PCI with PDD vs. PCI without PDD: 8.5 vs. 7, p < .001***). Apoptotic effects after PDT without HIPEC were limited to the tumor surface, whereas PDT after HIPEC (60 mg/m2 , 42°C) showed additional reduction of tumor proliferation in the top nine to 11 cell layers (50 µm). CONCLUSION: HYP as fluorescent photosensitizer offers an intraoperative diagnostic advantage detecting intraperitoneal tumor dissemination. The combination of HYP and cisplatin-based HIPEC was feasible in vivo, showing enhanced effects on tumor proliferation and apoptosis induction across the tumor surface. Further studies combining HYP and HIPEC will follow to establish a clinical application.


Asunto(s)
Hipertermia Inducida , Neoplasias Peritoneales , Rabdomiosarcoma Embrionario , Rabdomiosarcoma , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cisplatino/uso terapéutico , Terapia Combinada , Humanos , Quimioterapia Intraperitoneal Hipertérmica , Antígeno Ki-67 , Modelos Animales , Neoplasias Peritoneales/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma Embrionario/tratamiento farmacológico , Tasa de Supervivencia
3.
Asian Pac J Cancer Prev ; 23(2): 501-510, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35225462

RESUMEN

OBJECTIVE: Pomegranate ,a polyphenol-rich fruit, has been considered as one of the ancient fruits with anticancer effect. Cell cycle arrest is considered as an ordinary factor in human cancer, and apoptosis is the frequent drug target. This study aimed to evaluate the effectiveness of the Nimali variety of Sri Lankan Punica granatum L. fruit extracts on rhabdomyosarcoma (RD) cells concerning the apoptotic signaling pathway. METHODS: Antiproliferative activity of aqueous extracts of pomegranate peel, pericarp, was assessed using multiple extraction methods (sonication, microwaving, sonication followed by microwaving, keeping in a waterbath, and boiling at 100ºC). Total protein content, nitric oxide production, LDH, and caspase-8 and caspase-3 activities were analyzed in peel extracts prepared by sonicated or microwave methods. RT-qPCR was performed with intact RNA to explore the apoptotic pathway and gene expression. RESULTS: Peel extracts expressed minimum cell viability in a dose-dependent manner, induced cell death on RD cells. However, sonicated peel extract (SPL) indicated the lowest IC50 of 14.8±2.2 µg/mL comparative to healthy VERO cells (>1,000 µg/mL). A decrease of nitrite content in the supernatant was visualized in the graph plotted against concentration. Furthermore, SPL upregulated caspase-8 and caspase-3 signaling pathways and expression of p21 and p53 genes. CONCLUSION: The findings highlighted the promising therapeutic potential of SPL to inhibit RD growth and progression and to modulate the caspase-8 and caspase-3, p53, and p21 dependent pathway.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Frutas/química , Extractos Vegetales/farmacología , Granada (Fruta)/química , Rabdomiosarcoma/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Humanos , Transducción de Señal/efectos de los fármacos , Células Vero
4.
Photodiagnosis Photodyn Ther ; 36: 102565, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34614426

RESUMEN

BACKGROUND: Chemotherapy for rhabdomyosarcoma (RD) is effective, but it has critical side effects and unavoidable challenges. Photodynamic therapy (PDT) is an approach to treating cancer with relatively moderate side effects. Plant products are a rich source of polyphenols, which have potent antioxidant and anticancer activities. Therefore, their research has become an emerging field in recent decades. PURPOSE: This work aimed to evaluate the potential of hydrophobic extract of Ficus Carica (FC) to determine whether FC in the presence of low dose chemo and Aluminium Phthalocyanine (Photosense®) mediated photodynamic therapy synergistically enhances the treatment efficacy of RD cells. METHOD: FC with and without combination with individual therapeutic modalities like photosense mediated photodynamic therapy, chemotherapy, and their combinations were studied for cell viability and morphological changes in invitro RD cells. A semiconductor diode laser (630 nm) was used as a light source in PDT. The cytotoxic effect of FC on cell viability and cellular morphological changes were investigated by MTT reagent and a camera attached to an inverted visible light microscope. The effect of FC, followed by di-combination with low dose chemo (doxorubicin-HCl, and dacarbazine), Photosense® mediated PDT and chemo-Photosense® mediated PDT (tri-combination) at 630 nm diode laser and 10 J/cm2 fluency were also investigated by MTT reagent. The combination index method is used to identify the synergistic effect of combination therapy by using CompuSyn software based on the Chou-Talalay method. RESULTS: The dose-dependent effect of FC on cell viability and cellular morphological changes were observed in the RD cell line. It was found that the pre incubation of FC potentiated the anticancer effect as a neoadjuvant agent for doxorubicin-HCl and decarbazine based chemotherapy, Photosense® mediated PDT and chemo-PDT (tri-combination) with synergistic effect (CI<1). CONCLUSION: These results suggest a possible thread that the low dose combination of the aforementioned therapeutic modalities in the presence of FC remarkably enhances the treatment efficacy of RD in comparison with a single-agent treatment modality. The proposed sequence of FC with chemo and PDT might present better therapeutic outcomes in RD therapies and may provide result for RD metastasis. FC may also be used in the application of phyto-PDT to cancer in the future.


Asunto(s)
Ficus , Fotoquimioterapia , Rabdomiosarcoma , Línea Celular Tumoral , Humanos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Extractos Vegetales/farmacología , Hojas de la Planta , Rabdomiosarcoma/tratamiento farmacológico
5.
Phytother Res ; 35(3): 1378-1388, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33280183

RESUMEN

Rhabdomyosarcoma (RMS) is a rare type of soft tissue sarcoma most commonly found in pediatric patients. Despite progress, new and improved drug regimens are needed to increase survival rates. Citral, a natural product plant oil can induce cell death in cancer cells. Another compound, metformin, isolated originally from French lilac and used by diabetics, has been shown to reduce the incidence of cancer in these patients. Application of citral to RMS cells showed increase in cell death, and RD and RH30 cells showed half maximal inhibitory concentration (IC50 ) values as low as 36.28 µM and 62.37 µM, respectively. It was also shown that the citral initiated cell apoptosis through an increase in reactive oxygen species (ROS) and free calcium. In comparison, metformin only showed moderate cell death in RMS cell lines at a very high concentration (1,000 µM). Combinatorial experiments, however, indicated that citral and metformin worked antagonistically when used together. In particular, the ability of metformin to quench the ROS induced by citral could lead to the suppression of activity. These results clearly indicate that while clinical use of citral is a promising anti-tumor therapy, caution should be exercised in patients using metformin for diabetes.


Asunto(s)
Monoterpenos Acíclicos/uso terapéutico , Hipoglucemiantes/uso terapéutico , Medicina Tradicional China/métodos , Metformina/uso terapéutico , Rabdomiosarcoma/tratamiento farmacológico , Monoterpenos Acíclicos/farmacología , Niño , Humanos , Hipoglucemiantes/farmacología , Metformina/farmacología , Rabdomiosarcoma/patología
6.
Biomolecules ; 10(11)2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182753

RESUMEN

Magnoflorine (MGN) is a quaternary aporphine alkaloid that exhibits numerous therapeutic properties, including neuropsychopharmacological, anti-anxiety, immunomodulatory, anti-inflammatory, antioxidant, or antifungal activities. The aim of the present study was an investigation of the influence of MGN on viability, proliferation, induction of apoptosis, and cell cycle arrest in NCI-H1299 lung, MDA-MB-468 breast, T98G glioma, and TE671 rhabdomyosarcoma cancer cells. MGN was isolated from the roots of Berberis cretica L. by counter-current partition chromatography (CPC). Cell viability and proliferation assessments were performed by means of MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and 5-bromo-2'-deoxyuridine (BrDU) assays, respectively. The induction of apoptosis and cell cycle progression was measured using fluorescence-activated cell sorting analysis. MGN in high doses inhibits proliferation, induces apoptosis, and inhibits cell cycle in S/G2 phases in a dose-dependent manner. MGN seems to be a promising anti-cancer compound in therapy of some types of lung, breast, glioma, and rhabdomyosarcoma cancers, for which current standard therapies are limited or have severe strong side effects.


Asunto(s)
Antineoplásicos/farmacología , Aporfinas/farmacología , Berberis/química , Neoplasias de la Mama/tratamiento farmacológico , Glioma/tratamiento farmacológico , Extractos Vegetales/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Antineoplásicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Aporfinas/aislamiento & purificación , Neoplasias de la Mama/fisiopatología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Glioma/fisiopatología , Humanos , Extractos Vegetales/aislamiento & purificación , Rabdomiosarcoma/fisiopatología
7.
Molecules ; 25(9)2020 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-32353989

RESUMEN

Naturally occurring coumarins are bioactive compounds widely used in Asian traditional medicine. They have been shown to inhibit proliferation, induce apoptosis, and/or enhance the cytotoxicity of currently used drugs against a variety of cancer cell types. The aim of our study was to examine the antiproliferative activity of different linear furanocoumarins on human rhabdomyosarcoma, lung, and larynx cancer cell lines, and dissolve their cellular mechanism of action. The coumarins were isolated from fruits of Angelica archangelica L. or Pastinaca sativa L., and separated using high-performance counter-current chromatography (HPCCC). The identity and purity of isolated compounds were confirmed by HPLC-DAD and NMR analyses. Cell viability and toxicity assessments were performed by means of methylthiazolyldiphenyl-tetrazolium bromide (MTT) and lactate dehydrogenase (LDH) assays, respectively. Induction of apoptosis and cell cycle progression were measured using flow cytometry analysis. qPCR method was applied to detect changes in gene expression. Linear furanocoumarins in a dose-dependent manner inhibited proliferation of cancer cells with diverse activity regarding compounds and cancer cell type specificity. Imperatorin (IMP) exhibited the most potent growth inhibitory effects against human rhabdomyosarcoma and larynx cancer cell lines owing to inhibition of the cell cycle progression connected with specific changes in gene expression, including CDKN1A. As there are no specific chemotherapy treatments dedicated to laryngeal squamous cell carcinoma and rhabdomyosarcoma, and IMP seems to be non-toxic for normal cells, our results could open a new direction in the search for effective anti-cancer agents.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Furocumarinas/farmacología , Neoplasias Laríngeas/patología , Rabdomiosarcoma/patología , Angelica archangelica/química , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Cromatografía , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Fibroblastos/efectos de los fármacos , Citometría de Flujo , Frutas/química , Humanos , L-Lactato Deshidrogenasa/metabolismo , Neoplasias Laríngeas/tratamiento farmacológico , Pastinaca/química , Rabdomiosarcoma/tratamiento farmacológico
8.
Cancer Res ; 80(14): 3046-3056, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32354737

RESUMEN

Rhabdomyosarcoma is the most common childhood soft-tissue sarcoma, yet patients with metastatic or recurrent disease continue to do poorly, indicating a need for new treatments. The SRC family tyrosine kinase YES1 is upregulated in rhabdomyosarcoma and is necessary for growth, but clinical trials using single agent dasatinib, a SRC family kinase inhibitor, have failed in sarcomas. YAP1 (YES-associated protein) is highly expressed in rhabdomyosarcoma, driving growth and survival when the upstream Hippo tumor suppressor pathway is silenced, but efforts to pharmacologically inhibit YAP1 have been unsuccessful. Here we demonstrate that treatment of rhabdomyosarcoma with DNA methyltransferase inhibitor (DNMTi) upregulates Hippo activators RASSF1 and RASSF5 by promoter demethylation, activating canonical Hippo signaling and increasing inactivation of YAP1 by phosphorylation. Treatment with DNMTi decreased rhabdomyosarcoma cell growth and increased apoptosis and differentiation, an effect partially rescued by expression of constitutively active YAP (S127A), suggesting the effects of DNMTi treatment are, in part, due to Hippo-dependent inhibition of YAP1. In addition, YES1 and YAP1 interacted in the nucleus of rhabdomyosarcoma cells, and genetic or pharmacologic suppression of YES1 resulted in cytoplasmic retention of YAP1 and decreased YAP1 target gene expression, suggesting YES1 regulates YAP1 in a Hippo-independent manner. Combined treatment with DNMTi and dasatinib targeted both Hippo-dependent and Hippo-independent regulation of YAP1, ablating rhabdomyosarcoma cell growth in vitro and trending toward decreased tumor growth in vivo. These results show that the mechanisms regulating YAP1 in rhabdomyosarcoma can be inhibited by combinatorial therapy of DNMTi and dasatinib, laying the groundwork for future clinical investigations. SIGNIFICANCE: This study elucidates the signaling pathways that regulate the oncogenic protein YAP1 and identifies a combination therapy to target these pathways in the childhood tumor rhabdomyosarcoma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Azacitidina/análogos & derivados , Terapia Molecular Dirigida , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Rabdomiosarcoma/tratamiento farmacológico , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Apoptosis , Azacitidina/farmacología , Proliferación Celular , Niño , Femenino , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Humanos , Ratones , Ratones SCID , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP
9.
Int J Hyperthermia ; 37(1): 55-65, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31918587

RESUMEN

Introduction: An abscopal effect is a clinical observation whereby a local treatment is associated with regression of metastatic cancer at a site distant from the primary location of treatment. Here, we describe the clinical systemic effect induced by regional hyperthermia combined with low-dose chemotherapy and provide immunologic correlates.Case presentation: A 15-year-old patient had been diagnosed with alveolar rhabdomyosarcoma (ARMS). All previous treatment options failed in the patient including haploidentical stem cell transplantation and donor lymphocyte infusion. The patient presented with local and metastatic disease, and upon admission, underwent regional hyperthermia combined with low-dose chemotherapy. Immediately following therapy severe skin reactions were observed. Skin biopsies revealed an intraepithelial lymphocytic infiltration dominated by CD3+/CD8+ T cells with a regular network of dendritic cells. Clinical images compared before and during sequential treatment cycles showed complete metabolic response of the local tumor for more than 10 months of therapy. In addition, metastases completely regressed although they were not direct targets of regional hyperthermia. The systemic effect was associated with enhanced frequency of NK cells and T cells expressing the lectin-like natural-killer group 2 D activating receptor (NKG2D), an increase of the CD56bright subset of NK cells, as well as an increase of effector/memory and effector CD8+ and CD4+ T cells in the blood while the percentage of CD25+FOXP3+ regulatory T cells declined.Conclusions: Regional hyperthermia combined with low-dose chemotherapy had the potential to create a systemic effect which was associated with activation of NK cells and T cells.


Asunto(s)
Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/radioterapia , Adolescente , Femenino , Humanos , Hipertermia Inducida/métodos
10.
Semin Pediatr Surg ; 28(6): 150862, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31931970

RESUMEN

Soft tissue sarcomas of childhood are a heterogenous group of tumors with a wide spectrum of presentations and outcomes. Most patients require multimodal therapy with chemotherapy, surgery and/or radiation. Improved outcomes in recent decades have been achieved through improvements in the comprehensive care of these children through large cooperative group studies, even as little progress has been made in the standard chemotherapy backbone. A thorough understanding of the nuances of surgical therapy for these children is required to minimize both the risk of local failure and the possibility of loss of vital form or function.


Asunto(s)
Rabdomiosarcoma/cirugía , Sarcoma/cirugía , Neoplasias de los Tejidos Blandos/cirugía , Niño , Humanos , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/radioterapia , Sarcoma/tratamiento farmacológico , Sarcoma/radioterapia , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/radioterapia
11.
J Cancer Res Clin Oncol ; 144(9): 1717-1730, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29971532

RESUMEN

PURPOSE: Ferroptosis is a programmed form of iron-dependent cell death caused by lipid hydroperoxide accumulation, which can be prevented by glutathione peroxidase 4 (GPx4) activity. Here we investigated the effects of ferroptosis inducers called erastin and RSL3, which act by glutathione depletion and GPx4 inactivation, respectively, on muscle-derived cell lines of embryonal and alveolar rhabdomyosarcoma (RMS), and mouse normal skeletal C2C12 myoblasts. METHODS: Myogenic lines were exposed to stepwise increasing concentrations of ferroptosis inducers either alone or in combination with iron supplementation, iron chelating agents (bathophenanthrolinedisulfonic acid, BPS), antioxidant molecules (glutathione, N-acetylcysteine), lipid peroxidation inhibitors (ferrostatin-1), and chemotherapeutic agents (doxorubicin and actinomycin D). Drug susceptibility was quantified by measuring cell viability, proliferation and differentiation via neutral red assay, crystal violet assay and Giemsa staining, respectively. The detection of lipid hydroperoxide and protein levels was performed by immunofluorescence and Western blot analysis, respectively. RESULTS: Erastin and RSL3 increased lipid hydroperoxide levels preferentially in the embryonal U57810 and myoblast C2C12 lines, leading to ferroptosis that was accentuated by iron supplementation or prevented by co-treatment with BPS, glutathione, N-acetylcysteine and ferrostatin-1. The inhibition of extracellular regulated kinases (ERK) pathway prevented ferroptosis in U57810 and C2C12 cells, whereas its increased activation in the embryonal RD cells mediated by caveolin-1 (Cav-1) overexpression led to augmented ferroptosis susceptibility. Finally, we observed the combination of erastin or RSL3 with chemotherapeutic doxorubicin and actinomycin D agents to be effective in increasing cell death in all RMS lines. CONCLUSIONS: Erastin and RSL3 trigger ferroptosis in highly proliferating myogenic lines through a ERK pathway-dependent fashion.


Asunto(s)
Muerte Celular/fisiología , Proliferación Celular/fisiología , Mioblastos/patología , Rabdomiosarcoma/patología , Animales , Carbolinas/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclohexilaminas/farmacología , Dactinomicina/farmacología , Doxorrubicina/farmacología , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/fisiología , Ratones , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Fenilendiaminas/farmacología , Piperazinas/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo
12.
BMC Complement Altern Med ; 16(1): 484, 2016 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-27887609

RESUMEN

BACKGROUND: Mushrooms inspired the cuisines of many cultures and conventional medicaments for cancer. However, a substantial number of mushroom species are yet unexplored, possessing an unknown chemical, biological and pharmacological profiles. Fulviformes fastuosus is a terrestrial mushroom, which is commonly found in Sri Lankan woodlands. The current study was aimed at isolation and characterization of a potent cytotoxic compound from F. fastuosus and investigating the apoptotic effect induced by the active principle against cancer and normal cell lines. METHODS: Bioactivity guided isolation of active principles from the methanol extract of F. fastuosus was performed by a rapid extraction and isolation method using different chromatographic techniques. Potential cytotoxic compound was identified using one and two dimensional nuclear magnetic resonance spectroscopy and mass spectrometry. Isolated compound was screened for in vitro cytotoxicity against Hepatocellular carcinoma (HepG-2), Muscle rhabdomyosarcoma (RD) and Rat Wistar liver normal (CC-1) cell lines using 3 4, 5-(dimethylthiazol-2-yl) 2-5-diphenyl tetrazolium bromide (MTT) cell viability assay. Apoptotic features of cells were observed via microscopic examination and ethidium bromide/acridine orange fluorescent staining. RESULTS: The interpretation of spectral data resulted in the identification of the chemical structure as ergosta-4,6,8 (14),22-tetraen-3-one (ergone). Ergone exhibited promising cytotoxic properties against RD cells with less cytotoxicity effect on CC-1 cells. In addition, ergone also possesses a strong cytotoxic effect against HepG-2 cells showing low toxic level for CC-1 cells. Apoptotic features of treated cells were detected via morphological characterization and ethidium bromide/acridine orange staining. CONCLUSION: The present study elaborates the isolation of a potent cytotoxic compound; ergone, from F. fastuosus via a rapid and efficient isolation method. Importantly, ergone has exhibited greater cytotoxic activity against RD cells with high selectivity index compared to cytotoxicity against HepG-2 cells. Ergone can be used in the development of therapeutic strategies for curbing rhabdomyosarcoma.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Basidiomycota/química , Ergosterol/análogos & derivados , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular , Línea Celular Tumoral , Colestenonas , Ensayos de Selección de Medicamentos Antitumorales , Ergosterol/química , Ergosterol/aislamiento & purificación , Ergosterol/uso terapéutico , Células Hep G2 , Humanos , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Estructura Molecular , Neoplasias de los Músculos/tratamiento farmacológico , Ratas , Rabdomiosarcoma/tratamiento farmacológico , Sri Lanka , Coloración y Etiquetado
13.
Oncotarget ; 7(40): 65540-65552, 2016 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-27608846

RESUMEN

Long-term survival in patients with metastatic, relapsed, or recurrent Ewing sarcoma and rhabdomyosarcoma is dismal. Irinotecan, a topoisomerase 1 inhibitor, has activity in these sarcomas, but due to poor bioavailability of its active metabolite (SN-38) has had limited clinical efficacy. In this study we have evaluated the efficacy and toxicity of STA-8666, a novel drug conjugate which uses an HSP90 inhibitor to facilitate intracellular, tumor-targeted delivery of the topoisomerase 1 inhibitor SN-38, thus preferentially delivering and concentrating SN-38 within tumor tissue. We present in vivo evidence from mouse xenograft models that STA-8666 results in more persistent inhibition of topoisomerase 1 and prolonged DNA damage compared to irinotecan. This translates into superior antitumor efficacy and survival in multiple aggressive models of both diseases in mouse xenografts, as well as in an irinotecan-resistant model of pediatric osteosarcoma, demonstrated by dramatic tumor shrinkage, durable remission and prolonged complete regressions following short-term treatment, compared to conventional irinotecan. Gene expression analysis performed on xenograft tumors treated with either irinotecan or STA-8666 showed that STA-8666 affected expression of DNA damage and repair genes more robustly than irinotecan. These results suggest that STA-8666 may be a promising new agent for patients with pediatric-type sarcoma.


Asunto(s)
Antineoplásicos/uso terapéutico , Camptotecina/análogos & derivados , Resorcinoles/uso terapéutico , Rabdomiosarcoma/tratamiento farmacológico , Sarcoma de Ewing/tratamiento farmacológico , Inhibidores de Topoisomerasa I/uso terapéutico , Animales , Antineoplásicos/química , Camptotecina/química , Camptotecina/uso terapéutico , Línea Celular Tumoral , Niño , Daño del ADN , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Irinotecán , Ratones , Ratones Noqueados , Ratones SCID , Resorcinoles/química , Inhibidores de Topoisomerasa I/química , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Tumour Biol ; 37(1): 1025-33, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26269112

RESUMEN

Rhabdomyosarcoma (RMS) is a highly malignant and metastatic pediatric cancer that arises from the skeletal muscle. Recent studies have identified an important role of AKT signaling in RMS progression. In the current study, we investigated the activity of perifosine, an oral alkylphospholipid AKT inhibitor, against human RMS cells (RD and Rh-30 lines) both in vivo and in vitro, and studied the underlying mechanisms. We showed that perifosine significantly inhibited RMS cell growth in concentration- and time-dependent manners. Meanwhile, perifosine induced dramatic apoptosis in RMS cells. At the signaling level, perifosine blocked AKT activation, while inducing reactive oxygen species (ROS) production as well as JNK and P38 phosphorylations in RMS cells. Restoring AKT activation by introducing a constitutively active-AKT (CA-AKT) only alleviated (not abolished) perifosine-induced cytotoxicity in RD cells. Yet, the ROS scavenger N-acetyl cysteine (NAC) as well as pharmacological inhibitors against JNK (SP-600125) or P38 (SB-203580) suppressed perifosine-induced cytotoxicity in RMS cells. Thus, perifosine induces growth inhibition and apoptosis in RMS cells through mechanisms more than just blocking AKT. In vivo, oral administration of perifosine significantly inhibited growth of Rh-30 xenografts in severe combined immunodeficient (SCID) mice. Our data indicate that perifosine might be further investigated as a promising anti-RMS agent.


Asunto(s)
Antineoplásicos/farmacología , Fosforilcolina/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones SCID , Fosforilcolina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Pediatr Blood Cancer ; 62(9): 1562-6, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26207356

RESUMEN

BACKGROUND: Sorafenib is an oral small molecule inhibitor of multiple kinases controlling tumor growth and angiogenesis. The purpose of the phase 2 study was to determine the response rate of sorafenib and gain further information on the associated toxicities, pharmacokinetics, and pharmacodynamics of sorafenib in children and young adults with relapsed or refractory tumors including rhabdomyosarcoma, Wilms tumor, hepatocellular carcinoma (HCC), and papillary thyroid carcinoma (PTC). PROCEDURE: Sorafenib, 200 mg/m(2) /dose, was administered every 12 hr continuously for 28 day cycles using a two-stage design in two primary strata (rhabdomyosarcoma and Wilms tumor) and two secondary strata (HCC and PTC). Correlative studies in consenting patients included determination of sorafenib steady state trough concentrations and assessments of VEGF and sVEGFR2. RESULTS: Twenty patients (median age of 11 years; range, 5-21) enrolled. No objective responses (RECIST) were observed in the 10 evaluable patients enrolled in each of the two primary disease strata of rhabdomyosarcoma and Wilms tumor. No patients with HCC or PTC were enrolled. Sorafenib was not associated with an excessive rate of dose-limiting toxicity (DLT). The mean ± SD steady state concentration during cycle 1 day 15 was 6.5 ± 3.9 µg/ml (n = 10). CONCLUSIONS: Sorafenib was well tolerated in children at 200 mg/m(2) /dose twice daily on a continuous regimen with toxicity profile and steady state drug concentrations similar to those previously reported. Single agent sorafenib was inactive in children with recurrent or refractory rhabdomyosarcoma or Wilms tumor.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Rabdomiosarcoma/tratamiento farmacológico , Terapia Recuperativa , Tumor de Wilms/tratamiento farmacológico , Adolescente , Inhibidores de la Angiogénesis/efectos adversos , Inhibidores de la Angiogénesis/farmacocinética , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Niño , Preescolar , Femenino , Humanos , Neoplasias Renales/sangre , Neoplasias Renales/enzimología , Masculino , Proteínas de Neoplasias/sangre , Niacinamida/efectos adversos , Niacinamida/farmacocinética , Niacinamida/uso terapéutico , Compuestos de Fenilurea/efectos adversos , Compuestos de Fenilurea/farmacocinética , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Rabdomiosarcoma/sangre , Rabdomiosarcoma/enzimología , Sorafenib , Insuficiencia del Tratamiento , Factor A de Crecimiento Endotelial Vascular/sangre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre , Tumor de Wilms/sangre , Tumor de Wilms/enzimología , Adulto Joven
16.
Genetika ; 51(2): 147-55, 2015 Feb.
Artículo en Ruso | MEDLINE | ID: mdl-25966580

RESUMEN

This paper studies the effect of plant peptides of thionine Ns-W2 extracted from seeds of fennel flower (Nigella sativa) and ß-purothionine from wheat germs (Triticum kiharae), as well as a synthetic antimutagen (crown-compound), on the expression of several genes involved in the.control of cellular homeostasis, processes of carcinogenesis, and radiation response in human rhabdomyosarcoma cells (RD cells), T-lymphoblastoid cell line Jurkat, and blood cells. All of these agents acted as antimutagens-anticarcinogens, reducing the expression of genes involved in carcinogenesis (genes of families MMP, TIMP, and IAP and G-protein genes) in a tumor cell. A pronounced reduction in the mRNA level of these genes was caused by thionine Ns-W2, and the least effect was demonstrated by ß-purothionine. Antimutagens had very little effect on the mRNA levels of the several studied genes in normal blood cells.


Asunto(s)
Antimutagênicos/administración & dosificación , Péptidos/administración & dosificación , Fenotiazinas/administración & dosificación , Extractos Vegetales/administración & dosificación , Rabdomiosarcoma/genética , Antimutagênicos/química , Carcinogénesis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Nigella sativa/química , Péptidos/química , Fenotiazinas/química , Extractos Vegetales/química , Radiación Ionizante , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Triticum/química , Proteína p53 Supresora de Tumor/biosíntesis
17.
Pediatr Blood Cancer ; 62(1): 91-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25263539

RESUMEN

BACKGROUND: BMN 673 is a potent inhibitor of poly-ADP ribose polymerase (PARP) that is in clinical testing with a primary focus on BRCA-mutated cancers. BMN 673 is active both through inhibiting PARP catalytic activity and by tightly trapping PARP to DNA at sites of single strand breaks. PROCEDURE: BMN 673 was tested in vitro at concentrations ranging from 0.1 nM to 1 µM and in vivo at a daily dose of 0.33 mg/kg administered orally twice daily (Mon-Fri) and once daily on weekends (solid tumors) for 28 days. RESULTS: The median relative IC50 (rIC50 ) concentration against the PPTP cell lines was 25.8 nM. The median rIC50 for the Ewing cell lines was lower than for the remaining cell lines (6.4 vs. 31.1 nM, respectively). In vivo BMN 673 induced statistically significant differences in EFS distribution in 17/43 (39.5%) xenograft models. Three objective regressions were observed: a complete response (CR) in a medulloblastoma line (BT-45), a maintained CR in a Wilms tumor line (KT-10), and a maintained CR in an ependymoma line (BT-41). BMN 673 maintained its high level of activity against KT-10 with a threefold reduction in dose. KT-10 possesses a truncating mutation in PALB2 analogous to PALB2 mutations associated with hereditary breast and ovarian cancer that abrogate homologous recombination (HR) repair. CONCLUSIONS: The PPTP results suggest that single agent BMN 673 may have limited clinical activity against pediatric cancers. Single agent activity is more likely for patients whose tumors have defects in HR repair.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Mutación/genética , Proteínas Nucleares/genética , Ftalazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Proteínas Supresoras de Tumor/genética , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Evaluación Preclínica de Medicamentos , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , Neuroblastoma/patología , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/genética , Sarcoma de Ewing/patología
18.
Pediatr Blood Cancer ; 61(5): 922-4, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24166988

RESUMEN

Pixantrone, a novel aza-anthracenedione with cytotoxic activity, was tested against the PPTP in vitro panel (3.0 nM to 30.0 µM) and against a limited panel of PPTP Wilms tumors and sarcomas (7.5 mg/kg) administered intravenously using an every 4 day × 3 schedule. In vitro pixantrone showed a median relative IC50 value of 54 nM (range <3 nM to 1.03 µM). In vivo pixantrone induced significant differences in EFS distribution compared to controls in two of eight solid tumor xenografts at dose levels relevant to human drug exposure. A complete response was observed for one Wilms tumor xenograft.


Asunto(s)
ADN-Topoisomerasas de Tipo II/química , Isoquinolinas/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Sarcoma de Ewing/tratamiento farmacológico , Inhibidores de Topoisomerasa II/farmacología , Tumor de Wilms/tratamiento farmacológico , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Isoquinolinas/farmacocinética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Ratones , Ratones SCID , Rabdomiosarcoma/patología , Sarcoma de Ewing/patología , Distribución Tisular , Inhibidores de Topoisomerasa II/farmacocinética , Células Tumorales Cultivadas , Tumor de Wilms/patología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Clin Cancer Res ; 19(21): 5972-83, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23989978

RESUMEN

PURPOSE: To investigate the antitumor effect of nab-paclitaxel, an albumin-stabilized nanoparticle formulation of paclitaxel, on pediatric solid tumor models. EXPERIMENTAL DESIGN: A panel of three rhabdomyosarcoma, one osteosarcoma and seven neuroblastoma cell lines were exposed to increasing concentrations of nab-paclitaxel in vitro. Cell viability was evaluated using the Alamar Blue Assay. Antitumor effect was further assessed in vivo in NOD/SCID xenograft and metastatic neuroblastoma mouse models. Tumor sections were analyzed by immunohistochemistry for cleaved caspase-3 and phospho-histone H3. Plasma and intratumoral paclitaxel concentrations were measured by liquid chromatography-mass spectrometry. Ratio of intratumoral and plasma concentration was compared between nab-paclitaxel and paclitaxel treatment groups. RESULTS: Nab-paclitaxel displayed significant cytotoxicity against most pediatric solid tumor cell lines in vitro in a dose-dependent manner. In vivo, nab-paclitaxel showed antitumor activity in both rhabdomyosarcoma (RH4 and RD) and neuroblastoma [SK-N-BE(2) and CHLA-20] xenograft models. In the SK-N-BE(2) metastatic model, nab-paclitaxel treatment significantly extended animal survival compared with control (P < 0.01). Nab-paclitaxel treatment induced tumor cell-cycle arrest and apoptosis in vivo. In the RH4 model, increased local relapse-free intervals were observed with nab-paclitaxel treatment (37.7 ± 3.2 days) comparing with paclitaxel (13.6 ± 2.07 days). Local relapsed tumors following paclitaxel treatment proved to be paclitaxel-resistant and remained responsive to nab-paclitaxel. Mechanistically, a higher tumor/plasma paclitaxel drug ratio in favor of nab-paclitaxel was observed. CONCLUSIONS: Nab-paclitaxel showed significant antitumor activity against all pediatric solid tumors associated with an enhanced drug intratumor delivery. Furthermore, testing of nab-paclitaxel in pediatric solid-tumor patient population is under development.


Asunto(s)
Albúminas/farmacología , Antineoplásicos Fitogénicos/farmacología , Neoplasias/patología , Paclitaxel/farmacología , Albúminas/administración & dosificación , Albúminas/toxicidad , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/toxicidad , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/mortalidad , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Paclitaxel/administración & dosificación , Paclitaxel/toxicidad , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Rev. chil. urol ; 77(2): 98-104, 2012. ilus, tab
Artículo en Español | LILACS | ID: lil-783393

RESUMEN

El rabdomiosarcoma (RMS) representa el 3° tumor sólido extracraneal pediátrico. El uso de braquiterapia nos ha dado una nueva herramienta en el tratamiento de esta patología. En este trabajo queremos reportar la experiencia del uso de cirugía más conservadora asociada a braquiterapia en pacientes con RMS urológicos. Método: Revisión de todos los casos del año 2004-2011 de RMS urológicos manejados con braquiterapia postoperatoria y/o intraoperatoria, asociado a quimioterapia (QMT) preoperatoria. Resultados: En los 8 años de estudio hubo 6 RMS; se incluyen 5pacientes dado que uno fue RMS testicular que no requirió radioterapia. Caso 1: Masculino de 2 años RMS embrionario Grupo IV Estadio 4 de próstata, con QMT según protocolo y braquiterapia en la semana 24 de QMT. Lleva 76 meses libre de enfermedad. Caso 2: Masculino de 5 años, RMS embrionario Grupo lll Estadio 2 en vejiga, con QMT preoperatoria (12 semanas), cirugía con resección de tumor en cara anterior de vejiga y braquiterapia; completó esquema QMT. Lleva 30 meses libre de enfermedad, sin alteraciones miccionales. Caso 3: Masculino de 7 años, RMS embrionario Grupo lll Estadio 3 de próstata, con QMT según protocolo y braquiterapia en la semana 22 de QMT. Lleva 17meses libre de enfermedad, función vesical e intestinal normal. Caso 4: Femenino de 4 años, RMSbotroide Grupo lll Estadio 1 de vagina, con quimioterapia preoperatoria (12 semanas), cirugia y braquiterapia postquirúrgica, completó esquema de QMT. Lleva 4 meses libre de enfermedad, sin alteración miccional ni intestinal. Caso 5: Femenino de 2 años, RMS embrionario Grupo lll Estadio 3de psoas con compromiso de vejiga, con cirugía, QMT según protocolo y radioterapia externa; pre-sentó recidiva local, por lo que inicia QMT, cirugía resectiva del tumor en cara posterior de vejiga y uréter derecho + radioterapia intraoperatoria con cono. Lleva 2 meses libre de enfermedad. Sin alteración miccional ni intestinal...


The rhabdomyosarcoma (RMS) represents the 3m’ extracraneal solid tumor in children. Brachytherapy use has given a new tool in this disease treatment. In this investigation we want to report the experience of conservative surgery associated to brachytherapy in patients with urological RMS. Method: Retrospective review of all the urological RMS cases between the years 2004-2011managed with postoperative and or intraoperative brachytherapy, associated to preoperative chemotherapy ( CM T). Results: ln a 8 year period there were 6 RMS; 5 are included because one case was a testicular RMS that did not required radiotherapy. Case 1: Two years old male children, prostatic group l V stage 4 embryonal RMS, with CMT according to protocol and brachytherapy in the 24”’ week of CMT Has been 76 months free of disease. Case 2: Five years old boy, bladder group lll stage 2embryonal RMS, with preoperative CMT (12 weeks), surgery including resection of tumor in the bladder anterior wall and brachytherapy,‘ Completed CMT protocol. Has been 30 months free of disease, without voiding disorders. Case 3: Seven years old boy, prostatic group lll stage 3 embryonal RMS, with CMT according to protocol and brachytherapy in 22'“ CMT week. Has been 17 months free of disease, normal bladder and intestinal function. Case 4: Four years old girl, vagina group lll stage 1botyroid RMS, with preoperative CMT (12 weeks), resective surgery and post-operative brachytherapy, completed CMT according to protocol. Has been 4 months free of disease, without voiding or intestinal disorders. Case 5: Two years old girl, psoas compromising bladder group llI stage 3 embryonal RMS, with surgery, CMT according to protocol and external radiotherapy. Presented local recurrence reason why initiates new CMT protocol, resective posterior wall bladder and right ureter surgery intraoperative radiotherapy cone. Has been 2 months free of disease. Without voiding or intestinal...


Asunto(s)
Humanos , Masculino , Femenino , Lactante , Preescolar , Niño , Braquiterapia/métodos , Neoplasias Urológicas/cirugía , Neoplasias Urológicas/radioterapia , Rabdomiosarcoma/cirugía , Rabdomiosarcoma/radioterapia , Estadificación de Neoplasias , Estudios Retrospectivos , Neoplasias Urológicas/tratamiento farmacológico , Rabdomiosarcoma/tratamiento farmacológico , Terapia Combinada
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA