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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 141(13): 1610-1625, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36508699

RESUMEN

Myeloid neoplasms with erythroid or megakaryocytic differentiation include pure erythroid leukemia, myelodysplastic syndrome with erythroid features, and acute megakaryoblastic leukemia (FAB M7) and are characterized by poor prognosis and limited treatment options. Here, we investigate the drug sensitivity landscape of these rare malignancies. We show that acute myeloid leukemia (AML) cells with erythroid or megakaryocytic differentiation depend on the antiapoptotic protein B-cell lymphoma (BCL)-XL, rather than BCL-2, using combined ex vivo drug sensitivity testing, genetic perturbation, and transcriptomic profiling. High-throughput screening of >500 compounds identified the BCL-XL-selective inhibitor A-1331852 and navitoclax as highly effective against erythroid/megakaryoblastic leukemia cell lines. In contrast, these AML subtypes were resistant to the BCL-2 inhibitor venetoclax, which is used clinically in the treatment of AML. Consistently, genome-scale CRISPR-Cas9 and RNAi screening data demonstrated the striking essentiality of BCL-XL-encoding BCL2L1 but not BCL2 or MCL1, for the survival of erythroid/megakaryoblastic leukemia cell lines. Single-cell and bulk transcriptomics of patient samples with erythroid and megakaryoblastic leukemias identified high BCL2L1 expression compared with other subtypes of AML and other hematological malignancies, where BCL2 and MCL1 were more prominent. BCL-XL inhibition effectively killed blasts in samples from patients with AML with erythroid or megakaryocytic differentiation ex vivo and reduced tumor burden in a mouse erythroleukemia xenograft model. Combining the BCL-XL inhibitor with the JAK inhibitor ruxolitinib showed synergistic and durable responses in cell lines. Our results suggest targeting BCL-XL as a potential therapy option in erythroid/megakaryoblastic leukemias and highlight an AML subgroup with potentially reduced sensitivity to venetoclax-based treatments.


Asunto(s)
Leucemia Megacarioblástica Aguda , Leucemia Mieloide Aguda , Linfoma de Células B , Animales , Ratones , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Línea Celular Tumoral , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Proteína bcl-X/genética , Leucemia Megacarioblástica Aguda/tratamiento farmacológico , Leucemia Megacarioblástica Aguda/genética , Diferenciación Celular , Apoptosis
2.
Brief Bioinform ; 23(1)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34472587

RESUMEN

Chemosensitivity assays are commonly used for preclinical drug discovery and clinical trial optimization. However, data from independent assays are often discordant, largely attributed to uncharacterized variation in the experimental materials and protocols. We report here the launching of Minimal Information for Chemosensitivity Assays (MICHA), accessed via https://micha-protocol.org. Distinguished from existing efforts that are often lacking support from data integration tools, MICHA can automatically extract publicly available information to facilitate the assay annotation including: 1) compounds, 2) samples, 3) reagents and 4) data processing methods. For example, MICHA provides an integrative web server and database to obtain compound annotation including chemical structures, targets and disease indications. In addition, the annotation of cell line samples, assay protocols and literature references can be greatly eased by retrieving manually curated catalogues. Once the annotation is complete, MICHA can export a report that conforms to the FAIR principle (Findable, Accessible, Interoperable and Reusable) of drug screening studies. To consolidate the utility of MICHA, we provide FAIRified protocols from five major cancer drug screening studies as well as six recently conducted COVID-19 studies. With the MICHA web server and database, we envisage a wider adoption of a community-driven effort to improve the open access of drug sensitivity assays.

3.
Brief Bioinform ; 22(2): 1656-1678, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-32055842

RESUMEN

Drug development involves a deep understanding of the mechanisms of action and possible side effects of each drug, and sometimes results in the identification of new and unexpected uses for drugs, termed as drug repurposing. Both in case of serendipitous observations and systematic mechanistic explorations, confirmation of new indications for a drug requires hypothesis building around relevant drug-related data, such as molecular targets involved, and patient and cellular responses. These datasets are available in public repositories, but apart from sifting through the sheer amount of data imposing computational bottleneck, a major challenge is the difficulty in selecting which databases to use from an increasingly large number of available databases. The database selection is made harder by the lack of an overview of the types of data offered in each database. In order to alleviate these problems and to guide the end user through the drug repurposing efforts, we provide here a survey of 102 of the most promising and drug-relevant databases reported to date. We summarize the target coverage and types of data available in each database and provide several examples of how multi-database exploration can facilitate drug repurposing.


Asunto(s)
Bases de Datos Factuales , Reposicionamiento de Medicamentos , Biología Computacional/métodos , Sistemas de Liberación de Medicamentos , Encuestas y Cuestionarios
4.
BMC Bioinformatics ; 23(1): 245, 2022 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-35729494

RESUMEN

BACKGROUND: Drug-target interactions (DTIs) are critical for drug repurposing and elucidation of drug mechanisms, and are manually curated by large databases, such as ChEMBL, BindingDB, DrugBank and DrugTargetCommons. However, the number of curated articles likely constitutes only a fraction of all the articles that contain experimentally determined DTIs. Finding such articles and extracting the experimental information is a challenging task, and there is a pressing need for systematic approaches to assist the curation of DTIs. To this end, we applied Bidirectional Encoder Representations from Transformers (BERT) to identify such articles. Because DTI data intimately depends on the type of assays used to generate it, we also aimed to incorporate functions to predict the assay format. RESULTS: Our novel method identified 0.6 million articles (along with drug and protein information) which are not previously included in public DTI databases. Using 10-fold cross-validation, we obtained ~ 99% accuracy for identifying articles containing quantitative drug-target profiles. The F1 micro for the prediction of assay format is 88%, which leaves room for improvement in future studies. CONCLUSION: The BERT model in this study is robust and the proposed pipeline can be used to identify previously overlooked articles containing quantitative DTIs. Overall, our method provides a significant advancement in machine-assisted DTI extraction and curation. We expect it to be a useful addition to drug mechanism discovery and repurposing.


Asunto(s)
Reposicionamiento de Medicamentos , Proteínas , Bases de Datos Factuales , Interacciones Farmacológicas , Proteínas/metabolismo , PubMed
5.
Brief Bioinform ; 21(1): 211-220, 2020 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-30566623

RESUMEN

Knowledge of the full target space of drugs (or drug-like compounds) provides important insights into the potential therapeutic use of the agents to modulate or avoid their various on- and off-targets in drug discovery and precision medicine. However, there is a lack of consolidated databases and associated data exploration tools that allow for systematic profiling of drug target-binding potencies of both approved and investigational agents using a network-centric approach. We recently initiated a community-driven platform, Drug Target Commons (DTC), which is an open-data crowdsourcing platform designed to improve the management, reproducibility and extended use of compound-target bioactivity data for drug discovery and repurposing, as well as target identification applications. In this work, we demonstrate an integrated use of the rich bioactivity data from DTC and related drug databases using Drug Target Profiler (DTP), an open-source software and web tool for interactive exploration of drug-target interaction networks. DTP was designed for network-centric modeling of mode-of-action of multi-targeting anticancer compounds, especially for precision oncology applications. DTP enables users to construct an interaction network based on integrated bioactivity data across selected chemical compounds and their protein targets, further customizable using various visualization and filtering options, as well as cross-links to several drug and protein databases to provide comprehensive information of the network nodes and interactions. We demonstrate here the operation of the DTP tool and its unique features by several use cases related to both drug discovery and drug repurposing applications, using examples of anticancer drugs with shared target profiles. DTP is freely accessible at http://drugtargetprofiler.fimm.fi/.

6.
Pancreatology ; 21(2): 397-405, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33461933

RESUMEN

BACKGROUND/OBJECTIVES: Inflammation is related to the development and progression of pancreatic cancer (PC). Locally, anti-inflammatory macrophages (M2), and systemically, high levels of certain inflammation-modulating cytokines associate with poor prognosis in PC. The detailed effects of systemic inflammation on circulating monocytes and macrophage polarisation remain unknown. We aimed to find out how intracellular signalling of peripheral blood monocytes is affected by the systemic inflammatory state in PC patients and how it affects their differentiation into macrophages. METHODS: Monocytes were isolated from 50 consenting PC patients and 20 healthy controls (HC). The phosphorylation status of the signalling molecules was assessed by flow cytometry both from unstimulated and appropriately stimulated monocytes. Monocytes derived from HC and PC patients were co-cultured with cancer cells (MIA PaCa-2 and HPAF-II) in media supplemented with autologous serum, and the CD marker expression of the obtained macrophages was assessed by flow cytometry. RESULTS: Phosphorylation levels of unstimulated STAT2, STAT3 and STAT6 were higher (p < 0.05) and those of stimulated NF-kB (p = 0.004) and STAT5 (p = 0.006) were lower in patients than in controls. The expression of CD86, a proinflammatory (M1) marker, was higher in control- than patient-derived co-cultured macrophages (p = 0.029). CONCLUSIONS: Circulating monocytes from PC patients showed constitutive phosphorylation and weaker response to stimuli, indicating aberrant activation and immune suppression. When co-culturing the patient-derived monocytes with cancer cells, they differentiated into macrophages with reduced levels of M1 macrophage marker CD86, suggesting compromised anti-tumour features. The results highlight the need for global management of tumour-associated immune aberrations in PC treatment.


Asunto(s)
Diferenciación Celular/fisiología , Macrófagos/fisiología , Monocitos/fisiología , Neoplasias Pancreáticas/patología , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Línea Celular Tumoral , Movimiento Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Transducción de Señal
7.
Mol Ther ; 24(8): 1435-43, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27357626

RESUMEN

Adoptive T-cell transfer is a promising treatment approach for metastatic cancer, but efficacy in solid tumors has only been achieved with toxic pre- and postconditioning regimens. Thus, adoptive T-cell therapies would benefit from complementary modalities that enable their full potential without excessive toxicity. We aimed to improve the efficacy and safety of adoptive T-cell transfer by using adenoviral vectors for direct delivery of immunomodulatory murine cytokines into B16.OVA melanoma tumors with concomitant T-cell receptor transgenic OT-I T-cell transfer. Armed adenoviruses expressed high local and low systemic levels of cytokine when injected into B16.OVA tumors, suggesting safety of virus-mediated cytokine delivery. Antitumor efficacy was significantly enhanced with adenoviruses coding for murine interleukin-2 (mIL-2) and tumor necrosis factor-α (mTNFα) when compared with T-cell transfer alone or viruses alone. Further improvement in efficacy was achieved with a triple combination of mIL-2, mTNFα, and OT-I T-cells. Mechanistic studies suggest that mIL-2 has an important role in activating T-cells at the tumor, while mTNFα induces chemokine expression. Furthermore, adenovirus treatments enhanced tumor-infiltration of OT-I T-cells as demonstrated by SPECT/CT imaging of (111)In-labeled cells. Our results suggest the utility of cytokine-coding adenoviruses for improving the efficacy of adoptive T-cell therapies.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/genética , Inmunoterapia Adoptiva , Interleucina-2/genética , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Factor de Necrosis Tumoral alfa/genética , Animales , Antígeno B7-H1/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Animales de Enfermedad , Expresión Génica , Terapia Genética , Vectores Genéticos/administración & dosificación , Huésped Inmunocomprometido , Inyecciones Intralesiones , Interleucina-2/metabolismo , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/diagnóstico , Melanoma Experimental/terapia , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único , Factor de Necrosis Tumoral alfa/metabolismo
8.
Mol Ther ; 24(1): 175-83, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26310629

RESUMEN

Despite many clinical trials conducted with oncolytic viruses, the exact tumor-level mechanisms affecting therapeutic efficacy have not been established. Currently there are no biomarkers available that would predict the clinical outcome to any oncolytic virus. To assess the baseline immunological phenotype and find potential prognostic biomarkers, we monitored mRNA expression levels in 31 tumor biopsy or fluid samples from 27 patients treated with oncolytic adenovirus. Additionally, protein expression was studied from 19 biopsies using immunohistochemical staining. We found highly significant changes in several signaling pathways and genes associated with immune responses, such as B-cell receptor signaling (P < 0.001), granulocyte macrophage colony-stimulating factor (GM-CSF) signaling (P < 0.001), and leukocyte extravasation signaling (P < 0.001), in patients surviving a shorter time than their controls. In immunohistochemical analysis, markers CD4 and CD163 were significantly elevated (P = 0.020 and P = 0.016 respectively), in patients with shorter than expected survival. Interestingly, T-cell exhaustion marker TIM-3 was also found to be significantly upregulated (P = 0.006) in patients with poor prognosis. Collectively, these data suggest that activation of several functions of the innate immunity before treatment is associated with inferior survival in patients treated with oncolytic adenovirus. Conversely, lack of chronic innate inflammation at baseline may predict improved treatment outcome, as suggested by good overall prognosis.


Asunto(s)
Adenoviridae/fisiología , Perfilación de la Expresión Génica/métodos , Inmunidad Innata , Neoplasias/genética , Neoplasias/terapia , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos CD4/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Pronóstico , Receptores de Superficie Celular/metabolismo , Resultado del Tratamiento
9.
J Virol ; 89(20): 10637-47, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26269187

RESUMEN

UNLABELLED: Glioblastoma is a terminal disease with no effective treatment currently available. Among the new therapy candidates are oncolytic viruses capable of selectively replicating in cancer cells, causing tumor lysis and inducing adaptive immune responses against the tumor. However, tumor antiviral responses, primarily mediated by type I interferon (IFN-I), remain a key problem that severely restricts viral replication and oncolysis. We show here that the Semliki Forest virus (SFV) strain SFV4, which causes lethal encephalitis in mice, is able to infect and replicate independent of the IFN-I defense in mouse glioblastoma cells and cell lines originating from primary human glioblastoma patient samples. The ability to tolerate IFN-I was retained in SFV4-miRT124 cells, a derivative cell line of strain SFV4 with a restricted capacity to replicate in neurons due to insertion of target sites for neuronal microRNA 124. The IFN-I tolerance was associated with the viral nsp3-nsp4 gene region and distinct from the genetic loci responsible for SFV neurovirulence. In contrast to the naturally attenuated strain SFV A7(74) and its derivatives, SFV4-miRT124 displayed increased oncolytic potency in CT-2A murine astrocytoma cells and in the human glioblastoma cell lines pretreated with IFN-I. Following a single intraperitoneal injection of SFV4-miRT124 into C57BL/6 mice bearing CT-2A orthotopic gliomas, the virus homed to the brain and was amplified in the tumor, resulting in significant tumor growth inhibition and improved survival. IMPORTANCE: Although progress has been made in development of replicative oncolytic viruses, information regarding their overall therapeutic potency in a clinical setting is still lacking. This could be at least partially dependent on the IFN-I sensitivity of the viruses used. Here, we show that the conditionally replicating SFV4-miRT124 virus shares the IFN-I tolerance of the pathogenic wild-type SFV, thereby allowing efficient targeting of a glioma that is refractory to naturally attenuated therapy vector strains sensitive to IFN-I. This is the first evidence of orthotopic syngeneic mouse glioma eradication following peripheral alphavirus administration. Our findings indicate a clear benefit in harnessing the wild-type virus replicative potency in development of next-generation oncolytic alphaviruses.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Interferón Tipo I/inmunología , MicroARNs/inmunología , Virus Oncolíticos/fisiología , Virus de los Bosques Semliki/fisiología , Anciano , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/virología , Línea Celular Tumoral , Células Clonales , Resistencia a Antineoplásicos , Femenino , Regulación de la Expresión Génica , Glioblastoma/inmunología , Glioblastoma/mortalidad , Glioblastoma/virología , Humanos , Interferón Tipo I/genética , Masculino , Ratones , MicroARNs/genética , Neuronas/inmunología , Neuronas/patología , Neuronas/virología , Viroterapia Oncolítica/métodos , Transducción de Señal , Análisis de Supervivencia , Carga Tumoral , Replicación Viral
10.
Mol Ther ; 23(5): 964-973, 2015 05.
Artículo en Inglés | MEDLINE | ID: mdl-25655312

RESUMEN

The quality of the antitumor immune response is decisive when developing new immunotherapies for cancer. Oncolytic adenoviruses cause a potent immunogenic stimulus and arming them with costimulatory molecules reshapes the immune response further. We evaluated peripheral blood T-cell subsets of 50 patients with refractory solid tumors undergoing treatment with oncolytic adenovirus. These data were compared to changes in antiviral and antitumor T cells, treatment efficacy, overall survival, and T-cell subsets in pre- and post-treatment tumor biopsies. Treatment caused a significant (P < 0.0001) shift in T-cell subsets in blood, characterized by a proportional increase of CD8(+) cells, and decrease of CD4(+) cells. Concomitant treatment with cyclophosphamide and temozolomide resulted in less CD4(+) decrease (P = 0.041) than cyclophosphamide only. Interestingly, we saw a correlation between T-cell changes in peripheral blood and the tumor site. This correlation was positive for CD8(+) and inverse for CD4(+) cells. These findings give insight to the interconnections between peripheral blood and tumor-infiltrating lymphocyte (TIL) populations regarding oncolytic virotherapy. In particular, our data suggest that induction of T-cell response is not sufficient for clinical response in the context of immunosuppressive tumors, and that peripheral blood T cells have a complicated and potentially misleading relationship with TILs.


Asunto(s)
Adenoviridae , Terapia Genética , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Subgrupos de Linfocitos T/inmunología , Adenoviridae/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Niño , Femenino , Humanos , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias/diagnóstico , Neoplasias/genética , Virus Oncolíticos/genética , Subgrupos de Linfocitos T/metabolismo , Transducción Genética , Transgenes , Adulto Joven
11.
Int J Cancer ; 136(5): 1065-72, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25042001

RESUMEN

Oncolytic Western Reserve strain vaccinia virus selective for epidermal growth factor receptor pathway mutations and tumor-associated hypermetabolism was armed with human granulocyte-macrophage colony-stimulating factor (GMCSF) and a tdTomato fluorophore. As the assessment of immunological responses to human transgenes is challenging in the most commonly used animal models, we used immunocompetent Syrian golden hamsters, known to be sensitive to human GMCSF and semipermissive to vaccinia virus. Efficacy was initially tested in vitro on various human and hamster cell lines and oncolytic potency of transgene-carrying viruses was similar to unarmed virus. The hGMCSF-encoding virus was able to completely eradicate subcutaneous pancreatic tumors in hamsters, and to fully protect the animals from subsequent rechallenge with the same tumor. Induction of specific antitumor immunity was also shown by ex vivo co-culture experiments with hamster splenocytes. In addition, histological examination revealed increased infiltration of neutrophils and macrophages in GMCSF-virus-treated tumors. These findings help clarify the mechanism of action of GMCSF-armed vaccinia viruses undergoing clinical trials.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Viroterapia Oncolítica , Neoplasias Pancreáticas/inmunología , Virus Vaccinia/genética , Replicación Viral/inmunología , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Chlorocebus aethiops , Técnicas de Cocultivo , Cricetinae , ADN Viral/genética , Humanos , Técnicas para Inmunoenzimas , Macrófagos , Mesocricetus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T , Virus Vaccinia/inmunología , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Int J Cancer ; 136(4): 945-54, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24975392

RESUMEN

Despite originating from several different tissues, soft-tissue sarcomas (STS) are often grouped together as they share mesenchymal origin and treatment guidelines. Also, with some exceptions, a common denominator is that when the tumor cannot be cured with surgery, the efficacy of current therapies is poor and new treatment modalities are thus needed. We have studied the combination of a capsid-modified oncolytic adenovirus CGTG-102 (Ad5/3-D24-GMCSF) with doxorubicin, with or without ifosfamide, the preferred first-line chemotherapeutic options for most types of STS. We show that CGTG-102 and doxorubicin plus ifosfamide together are able to increase cell killing of Syrian hamster STS cells over single agents, as well as upregulate immunogenic cell death markers. When tested in vivo against established STS tumors in fully immunocompetent Syrian hamsters, the combination was highly effective. CGTG-102 and doxorubicin (without ifosfamide) resulted in synergistic antitumor efficacy against human STS xenografts in comparison with single agent treatments. Doxorubicin increased adenoviral replication in human and hamster STS cells, potentially contributing to the observed therapeutic synergy. In conclusion, the preclinical data generated here support clinical translation of the combination of CGTG-102 and doxorubicin, or doxorubicin plus ifosfamide, for the treatment of STS, and provide clues on the mechanisms of synergy.


Asunto(s)
Adenoviridae/inmunología , Antibióticos Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Leiomiosarcoma/terapia , Melanoma Experimental/terapia , Virus Oncolíticos/inmunología , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular , Terapia Combinada , Cricetinae , Doxorrubicina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Ifosfamida/farmacología , Ifosfamida/uso terapéutico , Leiomiosarcoma/inmunología , Masculino , Melanoma Experimental/inmunología , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Viroterapia Oncolítica , Sarcoma , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Int J Cancer ; 137(7): 1775-83, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25821063

RESUMEN

Metastatic melanoma is refractory to irradiation and chemotherapy, but amenable to immunological approaches such as immune-checkpoint-inhibiting antibodies or adoptive cell therapies. Oncolytic virus replication is an immunogenic phenomenon, and viruses can be armed with immunostimulatory molecules. Therefore, oncolytic immuno-virotherapy of malignant melanoma is an appealing approach, which was recently validated by a positive phase 3 trial. We investigated the potency of oncolytic adenovirus Ad5/3-D24-GMCSF on a panel of melanoma cell lines and animal models, and summarized the melanoma-specific human data from the Advanced Therapy Access Program (ATAP). The virus effectively eradicated human melanoma cells in vitro and subcutaneous SK-MEL-28 melanoma xenografts in nude mice when combined with low-dose cyclophosphamide. Furthermore, virally-expressed granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulated the differentiation of human monocytes into macrophages. In contrast to human cells, RPMI 1846 hamster melanoma cells exhibited no response to oncolytic viruses and the chimeric 5/3 fiber failed to increase the efficacy of transduction, suggesting limited utility of the hamster model in the context of viruses with this capsid. In ATAP, treatments appeared safe and well-tolerated. Four out of nine melanoma patients treated were evaluable for possible therapy benefit with modified RECIST criteria: one patient had minor response, two had stable disease, and one had progressive disease. Two patients were alive at 559 and 2,149 days after treatment. Ad5/3-D24-GMCSF showed promising efficacy in preclinical studies and possible antitumor activity in melanoma patients refractory to other forms of therapy. This data supports continuing the clinical development of oncolytic adenoviruses for treatment of malignant melanoma.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Melanoma/terapia , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Cricetinae , Ciclofosfamida/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Macrófagos/patología , Macrófagos/virología , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/virología , Ratones , Ratones Desnudos , Monocitos/patología , Monocitos/virología , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
14.
BMC Vet Res ; 11: 170, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26215394

RESUMEN

BACKGROUND: Dogs suffer from spontaneous tumors which may be amenable to therapies developed for human cancer patients, and dogs may serve as large-animal cancer models. A non-pathogenic Semliki Forest virus vector VA7-EGFP previously showed promise in targeting human tumor xenografts in mice, but the oncolytic capacity of the virus in canine cancer cells and the safety of the virus in higher mammals such as dogs, are not known. We therefore assessed the oncolytic potency of VA7-EGFP against canine cancer cells by infectivity and viability assays in two dog solid tumor cell lines. Furthermore we performed a 3-week safety study in two adult Beagles which received a single intravenous injection of ~2 × 10(5) plaque forming units of parental A7(74) strain. RESULTS: VA7-EGFP was able to replicate in and kill both canine cancer cell lines tested. No adverse events were observed in either of the two virus-injected adult Beagles and no infective virus could be recovered from any of the biological samples collected over the course of the study. Neutralizing antibodies to Semliki Forest virus became detectable in the dogs at 5 days post infection and remained elevated until study termination. CONCLUSIONS: Based on these results, testing of the oncolytic potential of attenuated Semliki Forest virus in canine cancer patients appears feasible.


Asunto(s)
Vacunas contra el Cáncer/efectos adversos , Enfermedades de los Perros/inducido químicamente , Animales , Anticuerpos Neutralizantes/sangre , Línea Celular Tumoral , Perros , Femenino , Virus de los Bosques Semliki , Replicación Viral/fisiología
15.
J Virol ; 87(4): 2363-6, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221568

RESUMEN

Attenuated Semliki Forest virus (SFV) may be suitable for targeting malignant glioma due to its natural neurotropism, but its replication in brain tumor cells may be restricted by innate antiviral defenses. We attempted to facilitate SFV replication in glioma cells by combining it with vaccinia virus, which is capable of antagonizing such defenses. Surprisingly, we found parenchymal mouse brain tumors to be refractory to both viruses. Also, vaccinia virus appears to be sensitive to SFV-induced antiviral interference.


Asunto(s)
Glioma/terapia , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus de los Bosques Semliki/inmunología , Virus Vaccinia/crecimiento & desarrollo , Virus Vaccinia/inmunología , Animales , Modelos Animales de Enfermedad , Ratones
16.
Blood Adv ; 8(7): 1621-1633, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38197948

RESUMEN

ABSTRACT: Monosomy 7 and del(7q) (-7/-7q) are frequent chromosomal abnormalities detected in up to 10% of patients with acute myeloid leukemia (AML). Despite unfavorable treatment outcomes, no approved targeted therapies exist for patients with -7/-7q. Therefore, we aimed to identify novel vulnerabilities. Through an analysis of data from ex vivo drug screens of 114 primary AML samples, we discovered that -7/-7q AML cells are highly sensitive to the inhibition of nicotinamide phosphoribosyltransferase (NAMPT). NAMPT is the rate-limiting enzyme in the nicotinamide adenine dinucleotide salvage pathway. Mechanistically, the NAMPT gene is located at 7q22.3, and deletion of 1 copy due to -7/-7q results in NAMPT haploinsufficiency, leading to reduced expression and a therapeutically targetable vulnerability to the inhibition of NAMPT. Our results show that in -7/-7q AML, differentiated CD34+CD38+ myeloblasts are more sensitive to the inhibition of NAMPT than less differentiated CD34+CD38- myeloblasts. Furthermore, the combination of the BCL2 inhibitor venetoclax and the NAMPT inhibitor KPT-9274 resulted in the death of significantly more leukemic blasts in AML samples with -7/-7q than the NAMPT inhibitor alone. In conclusion, our findings demonstrate that AML with -7/-7q is highly sensitive to NAMPT inhibition, suggesting that NAMPT inhibitors have the potential to be an effective targeted therapy for patients with monosomy 7 or del(7q).


Asunto(s)
Antineoplásicos , Leucemia Mieloide Aguda , Humanos , Nicotinamida Fosforribosiltransferasa , Leucemia Mieloide Aguda/genética , Deleción Cromosómica , Cromosomas Humanos Par 7
17.
Mol Ther ; 20(8): 1529-39, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22434140

RESUMEN

In our recent study, replicative alphaviral vector VA7 was found to be effective against orthotopic human U87-glioma xenografts in an athymic mouse model eradicating the tumors with single intravenous (i.v.) injection. Here, we tested the efficacy of VA7 in immunocompetent orthotopic GL261 and CT-2A glioma models of C57BL/6 mouse in vivo. The cell lines were susceptible to VA7 infection in vitro, but GL261 infection was highly restricted in confluent cell cultures, and mouse interferon-ß (IFNß) pretreatment prevented the replication of VA7 in both cell lines. When mice bearing orthotopic GL261 or CT-2A tumors were administered neurotropic VA7, either i.v. or intracranially (i.c.), the vector was unable to infect the tumor and no survival benefit was achieved. Pretreatments with immunosuppressive cyclophosphamide (CPA) and rapamycin markedly lowered serum-neutralizing antibodies (NAbs) but had no effect on tumor infection or survival. Intracranial GL261 tumors were refractory also in athymic C57BL/6 mice, which have serious defects in their adaptive immunity. Implanted VA7-infected GL261 cells formed tumors with only slightly delayed kinetics and without improving survival thus excluding the participation of physical barriers and indicating robust host IFN action. Mouse and human IFNß do not seem be species cross-reactive, which might limit the translational relevance of xenograft models in oncolytic virotherapy.


Asunto(s)
Alphavirus/genética , Glioma/tratamiento farmacológico , Glioma/terapia , Interferón beta/uso terapéutico , Viroterapia Oncolítica/métodos , Animales , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Inmunohistoquímica , Interferón beta/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Sci Rep ; 12(1): 21116, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36477604

RESUMEN

The drug development process consumes 9-12 years and approximately one billion US dollars in costs. Due to the high finances and time costs required by the traditional drug discovery paradigm, repurposing old drugs to treat cancer and rare diseases is becoming popular. Computational approaches are mainly data-driven and involve a systematic analysis of different data types leading to the formulation of repurposing hypotheses. This study presents a novel scoring algorithm based on chemical and genomic data to repurpose drugs for 669 diseases from 22 groups, including various cancers, musculoskeletal, infections, cardiovascular, and skin diseases. The data types used to design the scoring algorithm are chemical structures, drug-target interactions (DTI), pathways, and disease-gene associations. The repurposed scoring algorithm is strengthened by integrating the most comprehensive manually curated datasets for each data type. At DrugRepo score ≥ 0.4, we repurposed 516 approved drugs across 545 diseases. Moreover, hundreds of novel predicted compounds can be matched with ongoing studies at clinical trials. Our analysis is supported by a web tool available at: http://drugrepo.org/ .


Asunto(s)
Genómica
20.
Cancers (Basel) ; 14(3)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35158794

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a silent killer, often diagnosed late. However, it is also dishearteningly resistant to nearly all forms of treatment. New therapies are urgently needed, and with the advent of organoid culture for pancreatic cancer, an increasing number of innovative approaches are being tested. Organoids can be derived within a short enough time window to allow testing of several anticancer agents, which opens up the possibility for functional precision medicine for pancreatic cancer. At the same time, organoid model systems are being refined to better mimic the cancer, for example, by incorporation of components of the tumor microenvironment. We review some of the latest developments in pancreatic cancer organoid research and in novel treatment design. We also summarize our own current experiences with pancreatic cancer organoid drug sensitivity and resistance testing (DSRT) in 14 organoids from 11 PDAC patients. Our data show that it may be necessary to include a cell death read-out in ex vivo DSRT assays, as metabolic viability quantitation does not capture actual organoid killing. We also successfully adapted the organoid platform for drug combination synergy discovery. Lastly, live organoid culture 3D confocal microscopy can help identify individual surviving tumor cells escaping cell death even during harsh combination treatments. Taken together, the organoid technology allows the development of novel precision medicine approaches for PDAC, which paves the way for clinical trials and much needed new treatment options for pancreatic cancer patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA