Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(8)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38673835

RESUMEN

Virotherapy is one of the perspective technologies in the treatment of malignant neoplasms. Previously, we have developed oncolytic vaccinia virus VV-GMCSF-Lact and its high cytotoxic activity and antitumor efficacy against glioma was shown. In this work, using immortalized and patient-derived cells with different sensitivity to VV-GMCSF-Lact, we evaluated the cytotoxic effect of chemotherapy agents. Additionally, we studied the combination of VV-GMCSF-Lact with temozolomide which is the most preferred drug for glioma treatment. Experimental results indicate that first adding temozolomide and then the virus to the cells is inherently more efficient than dosing it in the reverse order. Testing these regimens in the U87 MG xenograft glioblastoma model confirmed this effect, as assessed by tumor growth inhibition index and histological analysis. Moreover, VV-GMCSF-Lact as monotherapy is more effective against U87 MG glioblastoma xenografts comparing temozolomide.


Asunto(s)
Glioma , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Viroterapia Oncolítica , Virus Oncolíticos , Temozolomida , Virus Vaccinia , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Animales , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Temozolomida/farmacología , Temozolomida/uso terapéutico , Línea Celular Tumoral , Ratones , Glioma/terapia , Glioma/tratamiento farmacológico , Glioma/patología , Virus Vaccinia/genética , Virus Vaccinia/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Ratones Desnudos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Glioblastoma/terapia , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Terapia Combinada
2.
Cells ; 12(22)2023 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-37998351

RESUMEN

Oncolytic virotherapy is a rapidly evolving approach that aims to selectively kill cancer cells. We designed a promising recombinant vaccinia virus, VV-GMCSF-Lact, for the treatment of solid tumors, including glioma. We assessed how VV-GMCSF-Lact affects human cells using immortalized and patient-derived glioma cultures and a non-malignant brain cell culture. Studying transcriptome changes in cells 12 h or 24 h after VV-GMCSF-Lact infection, we detected the common activation of histone genes. Additionally, genes associated with the interferon-gamma response, NF-kappa B signaling pathway, and inflammation mediated by chemokine and cytokine signaling pathways showed increased expression. By contrast, genes involved in cell cycle progression, including spindle organization, sister chromatid segregation, and the G2/M checkpoint, were downregulated following virus infection. The upregulation of genes responsible for Golgi vesicles, protein transport, and secretion correlated with reduced sensitivity to the cytotoxic effect of VV-GMCSF-Lact. Higher expression of genes encoding proteins, which participate in the maturation of pol II nuclear transcripts and mRNA splicing, was associated with an increased sensitivity to viral cytotoxicity. Genes whose expression correlates with the sensitivity of cells to the virus are important for increasing the effectiveness of cancer virotherapy. Overall, the results highlight molecular markers, biological pathways, and gene networks influencing the response of glioma cells to VV-GMCSF-Lact.


Asunto(s)
Glioma , Virus Oncolíticos , Humanos , Virus Oncolíticos/genética , Transcriptoma/genética , Replicación Viral/genética , Glioma/genética , Glioma/terapia , Glioma/patología , Virus Vaccinia/genética
3.
Curr Issues Mol Biol ; 44(11): 5294-5311, 2022 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-36354672

RESUMEN

Glioblastoma multiforme (GBM) is one of the most highly metastatic cancers. The study of the pathogenesis of GBM, as well as the development of targeted oncolytic drugs, require the use of actual cell models, in particular, the use of 3D cultures or neurospheres (NS). During the formation of NS, the adaptive molecular landscape of the transcriptome, which includes various regulatory RNAs, changes. The aim of this study was to reveal changes in the expression of microRNAs (miRNAs) and their target mRNAs in GBM cells under conditions of NS formation. Neurospheres were obtained from both immortalized U87 MG and patient-derived BR3 GBM cell cultures. Next generation sequencing analysis of small and long RNAs of adherent and NS cultures of GBM cells was carried out. It was found that the formation of NS proceeds with an increase in the level of seven and a decrease in the level of 11 miRNAs common to U87 MG and BR3, as well as an increase in the level of 38 and a decrease in the level of 12 mRNA/lncRNA. Upregulation of miRNAs hsa-miR: -139-5p; -148a-3p; -192-5p; -218-5p; -34a-5p; and -381-3p are accompanied by decreased levels of their target mRNAs: RTN4, FLNA, SH3BP4, DNPEP, ETS2, MICALL1, and GREM1. Downregulation of hsa-miR: -130b-5p, -25-5p, -335-3p and -339-5p occurs with increased levels of mRNA-targets BDKRB2, SPRY4, ERRFI1 and TGM2. The involvement of SPRY4, ERRFI1, and MICALL1 mRNAs in the regulation of EGFR/FGFR signaling highlights the role of hsa-miR: -130b-5p, -25-5p, -335-3p, and -34a-5p not only in the formation of NS, but also in the regulation of malignant growth and invasion of GBM. Our data provide the basis for the development of new approaches to the diagnosis and treatment of GBM.

4.
Cells ; 11(19)2022 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-36231068

RESUMEN

Glioma is the most common and heterogeneous primary brain tumor. The development of a new relevant preclinical models is necessary. As research moves from cultures of adherent gliomas to a more relevant model, neurospheres, it is necessary to understand the changes that cells undergo at the transcriptome level. In the present work, we used three patient-derived gliomas and two immortalized glioblastomas, while their cultivation was carried out under adherent culture and neurosphere (NS) conditions. When comparing the transcriptomes of monolayer (ML) and NS cell cultures, we used Enrichr genes sets enrichment analysis to describe transcription factors (TFs) and the pathways involved in the formation of glioma NS. It was observed that NS formation is accompanied by the activation of five common gliomas of TFs, SOX2, UBTF, NFE2L2, TCF3 and STAT3. The sets of transcripts controlled by TFs MYC and MAX were suppressed in NS. Upregulated genes are involved in the processes of the epithelial-mesenchymal transition, cancer stemness, invasion and migration of glioma cells. However, MYC/MAX-dependent downregulated genes are involved in translation, focal adhesion and apical junction. Furthermore, we found three EGFR and FGFR signaling feedback regulators common to all analyzed gliomas-SPRY4, ERRFI1, and RAB31-which can be used for creating new therapeutic strategies of suppressing the invasion and progression of gliomas.


Asunto(s)
Glioma , Transcriptoma , Línea Celular Tumoral , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Humanos , Transcriptoma/genética
5.
Life (Basel) ; 11(10)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34685455

RESUMEN

Glioblastoma is one of the most aggressive brain tumors. Given the poor prognosis of this disease, novel methods for glioblastoma treatment are needed. Virotherapy is one of the most actively developed approaches for cancer therapy today. VV-GMCSF-Lact is a recombinant vaccinia virus with deletions of the viral thymidine kinase and growth factor genes and insertions of the granulocyte-macrophage colony-stimulating factor and oncotoxic protein lactaptin genes. The virus has high cytotoxic activity against human cancer cells of various histogenesis and antitumor efficacy against breast cancer. In this work, we show VV-GMCSF-Lact to be a promising therapeutic agent for glioblastoma treatment. VV-GMCSF-Lact effectively decreases the viability of glioblastoma cells of both immortalized and patient-derived cultures in vitro, crosses the blood-brain barrier, selectively replicates into orthotopically transplanted human glioblastoma when intravenously injected, and inhibits glioblastoma xenograft and metastasis growth when injected intratumorally.

6.
Cancer Biol Med ; 2021 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-34259424

RESUMEN

OBJECTIVE: Glioma is a highly invasive tumor, frequently disposed in essential areas of the brain, which makes its surgical excision extremely difficult; meanwhile adjuvant therapy remains quite ineffective. METHODS: In the current report, a new therapeutic approach in curing malignant neoplasms has been performed on the U87 human glioblastoma model. This approach, termed "Karanahan", is aimed at the eradication of cancer stem cells (CSCs), which were recently shown to be capable of internalizing fragments of extracellular double-stranded DNA. After being internalized, these fragments interfere in the process of repairing interstrand cross-links caused by exposure to appropriate cytostatics, and such an interference results either in elimination of CSCs or in the loss of their tumorigenic potency. Implementation of the approach requires a scheduled administration of cytostatic and complex composite double-stranded DNA preparation. RESULTS: U87 cells treated in vitro in accordance with the Karanahan approach completely lost their tumorigenicity and produced no grafts upon intracerebral transplantation into immunodeficient mice. In SCID mice with developed subcutaneous grafts, the treatment resulted in reliable slowing down of tumor growth rate (P < 0.05). In the experiment with intracerebral transplantation of U87 cells followed by surgical excision of the developed graft and subsequent therapeutic treatment, the Karanahan approach was shown to reliably slow down the tumor growth rate and increase the median survival of the mice twofold relative to the control. CONCLUSIONS: The effectiveness of the Karanahan approach has been demonstrated both in vitro and in vivo in treating developed subcutaneous grafts as well as orthotopic grafts after surgical excision of the tumor.

7.
Surg Neurol Int ; 12: 72, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33767876

RESUMEN

BACKGROUND: Concepts of Chiari malformation Type 1 (CM1) surgery in the present time significantly different. The most common complications are pseudomeningocele (12%) and postoperative CSF leak (5%). The development of pseudomeningocele may be associated with inappropriate restoration of bone and muscles relations. METHODS: The pilot study involved 11 patients aged 24-64 years with a diagnosis of CM1 who had indications for surgical treatment. Special titanium implant enabling fixation of the occipital and cervical muscles at the projections of their normal attachments was developed, it was placed to occipital bone on the final stages of surgical intervention. Surgical technique promoted tightened wound closure neutralizing formation of "dead space" at the place of occipital craniectomy and between muscle layers. The implant was produced by direct metal laser sintering method for each patient individually. RESULTS: There were no complications during the hospitalization and follow-up period. Postoperative MRI demonstrated adequate formation of the cisterna magna and the absence of pseudomeningocele. During follow-up period there were no signs of pseudomeningocele, CSF leak, surgical scar complications, implant-associated infections, and other complications. CONCLUSION: In the study group, no pseudomeningocele cases as long as any other complications associated with surgery had been revealed. The efficacy of the proposed surgical technique using the developed implant should be evaluated in clinical trials with larger patient samples. To simplify preoperative planning and manufacturing of the implant for each patient individually, a set of implants with different specified sizes was developed.

8.
Int J Mol Sci ; 21(8)2020 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-32326230

RESUMEN

Besides an antigen-presenting function and ability to induce antitumor immune responses, dendritic cells (DCs) possess a direct tumoricidal activity. We previously reported that monocyte-derived IFNα-induced DCs (IFN-DCs) of glioblastoma multiforme patients express low levels of membrane TNFα molecule (mTNFα) and have impaired TNFα/TNF-R1-mediated cytotoxicity against immortalized tumor cell line HEp-2. However, whether the observed defect could affect killer activity of glioma patient DCs against autologous tumor cells remained unclear. Here, we show that donor IFN-DCs possess cytotoxic activity against glioblastoma cell lines derived from a primary tumor culture. Granule-mediated and TNFα/TNF-R1-dependent pathways were established as the main mechanisms underlying cytotoxic activity of IFN-DCs. Glioblastoma patient IFN-DCs showed lower cytotoxicity against autologous glioblastoma cells sensitive to TNFα/TNFR1-mediated lysis, which was associated with low TNFα mRNA expression and high TACE/ADAM-17 enzyme activity. Recombinant IL-2 (rIL-2) and human double-stranded DNA (dsDNA) increased 1.5-fold cytotoxic activity of patient IFN-DCs against autologous glioblastoma cells. dsDNA, but not rIL-2, enhanced the expression of TNFα mRNA and decreased expression and activity of TACE/ADAM-17 enzyme. In addition, dsDNA and rIL-2 stimulated the expression of perforin and granzyme B (in the presence of dsDNA), suggesting the possibility of enhancing DC cytotoxicity against autologous glioblastoma cells via various mechanisms.


Asunto(s)
Membrana Celular/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Glioblastoma/etiología , Glioblastoma/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Biomarcadores , Línea Celular Tumoral , Susceptibilidad a Enfermedades , Expresión Génica , Glioblastoma/patología , Humanos , Interleucina-2/metabolismo , Interleucina-2/farmacología , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética
9.
Steroids ; 151: 108460, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31344410

RESUMEN

In an endeavour to develop potent anti-tumor agents from diosgenin, a series of C-6 derived 1,2,3-triazolyl derivatives were designed and synthesized by employing Cu(I) catalyzed Huisgen 1,3-dipolar cycloaddition reaction of novel azides - (22R,25R)-6ß-azidospirostan-3ß,5α-diol and 6ß-azido-7α-hydroxyspirosta-1,4-dien-3-one with aryl(hetaryl)alkynes. All the derivatives were evaluated for cytotoxic activity by MTT assay against eight different human cancer cell lines: T-cellular leucosis (CEM-13), human monocytes (U-937), breast (MDA-MB-231, BT-474), prostate (DU-145) and glioblastoma (U-87MG, SNB-19, T98G). The results of this study suggested that 6-(4'-aryl-1',2',3'-triazolyl)spirostan-3,5-diols 2, 3, 4, 5 and 6 possessed a promising cytotoxic potential. The corresponding 6-substituted 7-hydroxy-1,4-spirostadien-3-ones shown less cytotoxity on the human cancer cells. Compounds 2, 3, 4, and 5 which demonstrated high grown inhibition against glioma cancer cells U-87 and T98G, and also on the human-derived N118669 primary glioblastoma cell line (with GI50 values in the range of 5-9 µM), were not affected the growth of SNB-19 cells. The data revealed that phenyl, 4-methoxyphenyl, 4-fluorophenyl, 3,4,5-trimethoxyphenyl or 2-pyridinyl substituent in the triazole moiety at the C-6 position significantly improved the anti-tumor activity. The mentioned position at the spirostan core may be favourable for the synthesis of potent anticancer leads from diosgenin.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Compuestos de Espiro/química , Triazoles/síntesis química , Triazoles/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Técnicas de Química Sintética , Humanos , Relación Estructura-Actividad , Triazoles/química
10.
J Interferon Cytokine Res ; 38(7): 298-310, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29932796

RESUMEN

Besides initiation of tumor-specific T cell immunity, dendritic cells (DCs) are endowed with tumoricidal activity. Previously, we showed that monocyte-derived DCs of high-grade glioma patients generated in the presence of interferon alpha (IFNα) (IFN-DCs) have impaired cytotoxic activity against tumor necrosis factor alpha (TNFα)-sensitive HEp-2 tumor cells. Herein, we demonstrate that decreased transmembrane TNFα (tmTNFα) expression, but not soluble TNFα (sTNFα) production by high-grade glioma patient IFN-DCs, determines the defective tumoricidal activity against TNFα-sensitive HEp-2 cells. Blocking TNFα-converting enzyme or stimulation of patient IFN-DCs with rIL-2 or dsDNA enhances tmTNFα expression on IFN-DCs and significantly increases their cytotoxicity. Decreased tmTNFα expression on patient IFN-DCs is not caused by downregulation of pNFκB. Neither rIL-2 nor dsDNA upregulates tmTNFα expression on patient IFN-DCs via an increase of pNFκB. The current study shows an important role of tmTNFα as mediator of IFN-DC tumoricidal activity and as molecular target for the restoration of defective DC killer activity in high-grade glioma patients.


Asunto(s)
ADN/inmunología , Células Dendríticas/inmunología , Glioma/inmunología , Glioma/patología , Interleucina-2/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proliferación Celular/efectos de los fármacos , Niño , Células Dendríticas/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/inmunología , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/biosíntesis , Adulto Joven
11.
Technol Cancer Res Treat ; 17: 1533034617753812, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29375020

RESUMEN

Poorly differentiated cell populations including tumor-initiating stem cells have been demonstrated to display a unique ability to natively internalize fragmented double-stranded DNA. Using this feature as a marker, we show that 0.1% to 6% of human glioblastoma cells from the bioptates can effectively internalize a fluorescently labeled DNA probe. Of these, using samples from 3 patients, 66% to 100% cells are also positive for CD133, a well-established surface marker of tumor-initiating glioma stem cells. Using the samples from primary malignant brain lesions (33 patients), we demonstrate that tumor grading significantly correlates ( R = .71) with the percentage of DNA-internalizing cells. No such correlation is observed for relapse samples (18 patients).


Asunto(s)
Biomarcadores de Tumor , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioma/metabolismo , Glioma/patología , Células Madre Neoplásicas/metabolismo , Antígeno AC133 , Neoplasias Encefálicas/cirugía , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Técnica del Anticuerpo Fluorescente , Glioma/genética , Glioma/cirugía , Humanos , Clasificación del Tumor , Células Madre Neoplásicas/patología , Células Tumorales Cultivadas
12.
Cancer Biol Ther ; 15(10): 1378-94, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25117082

RESUMEN

It has been established previously that up to 40% of mouse CD34(+) hematopoietic stem cells are capable of internalizing exogenous dsDNA fragments both in vivo and ex vivo. Importantly, when mice are treated with a combination of cyclophosphamide and dsDNA, the repair of interstrand crosslinks in hematopoietic progenitors is attenuated, and their pluripotency is altered. Here we show for the first time that among various actively proliferating mammalian cell populations there are subpopulations capable of internalizing dsDNA fragments. In the context of cancer, such dsDNA-internalizing cell subpopulations display cancer stem cell-like phenotype. Furthermore, using Krebs-2 ascites cells as a model, we found that upon combined treatment with cyclophosphamide and dsDNA, engrafted material loses its tumor-initiating properties which we attribute to the elimination of tumor-initiating stem cell subpopulation or loss of its tumorigenic potential.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Madre Neoplásicas/patología , Animales , Antineoplásicos/farmacología , Ascitis/metabolismo , Ascitis/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Carcinoma Krebs 2/metabolismo , Carcinoma Krebs 2/patología , Proliferación Celular/efectos de los fármacos , Ciclofosfamida/farmacología , ADN/metabolismo , ADN/farmacología , Endocitosis , Glioblastoma/metabolismo , Glioblastoma/patología , Xenoinjertos , Ratones Endogámicos CBA , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/efectos de los fármacos , Reparación del ADN por Recombinación/genética , Células Tumorales Cultivadas
13.
Cell Immunol ; 284(1-2): 146-53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23973877

RESUMEN

Recent studies have revealed that besides the important role in triggering the adoptive antitumor immunity, dendritic cells (DCs) possess direct cytotoxic antitumor activity. Here, we investigated brain glioma patient monocyte-derived DCs generated in the presence of IFNα and GM-CSF (IFN-DCs). These DCs were characterized by reduced cytotoxic activity against TRAIL-resistant HEp-2 cells. The impairment of DC cytotoxic function was observed mainly in high-grade glioma patients and associated with poor survival. The dysfunction of patient DC cytotoxicity was partially restored under in vitro pretreatment of DCs with double-stranded human DNA as well as rIL-2. In contrast to healthy donors, IFN-DCs in a part of high-grade glioma patients also failed to lyse primary autologous or allogeneic glioma cells. Our findings point to possible contribution of DC impairment in tumor pathogenesis in brain glioma and justify the necessity to evaluate and correct DC cytotoxic function when exploring DCs as cancer vaccines in glioma.


Asunto(s)
Neoplasias Encefálicas/terapia , Células Dendríticas/inmunología , Glioma/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Inmunoterapia Adoptiva/métodos , Interferón-alfa/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Adolescente , Adulto , Anciano , Neoplasias Encefálicas/inmunología , Estudios de Casos y Controles , Línea Celular Tumoral , Supervivencia Celular/inmunología , Niño , Pruebas Inmunológicas de Citotoxicidad , Femenino , Glioma/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Estimación de Kaplan-Meier , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA