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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 130(7): 1187-1195, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38316993

RESUMEN

BACKGROUND: Pancreatic cancer is an aggressive, immunologically "cold" tumor. Oncolytic virotherapy is a promising treatment to overcome this problem. We developed a telomerase-specific oncolytic adenovirus armed with p53 gene (OBP-702). METHODS: We investigated the efficacy of OBP-702 for pancreatic cancer, focusing on its long-term effects via long-lived memory CD8 + T cells including tissue-resident memory T cells (TRMs) and effector memory T cells (TEMs) differentiated from effector memory precursor cells (TEMps). RESULTS: First, in vitro, OBP-702 significantly induced adenosine triphosphate (ATP), which is important for memory T cell establishment. Next, in vivo, OBP-702 local treatment to murine pancreatic PAN02 tumors increased TEMps via ATP induction from tumors and IL-15Rα induction from macrophages, leading to TRM and TEM induction. Activation of these memory T cells by OBP-702 was also maintained in combination with gemcitabine+nab-paclitaxel (GN) in a PAN02 bilateral tumor model, and GN + OBP-702 showed significant anti-tumor effects and increased TRMs in OBP-702-uninjected tumors. Finally, in a neoadjuvant model, in which PAN02 cells were re-inoculated after resection of treated-PAN02 tumors, GN + OBP-702 provided long-term anti-tumor effects even after tumor resection. CONCLUSION: OBP-702 can be a long-term immunostimulant with sustained anti-tumor effects on immunologically cold pancreatic cancer.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias Pancreáticas , Telomerasa , Humanos , Animales , Ratones , Adenoviridae/genética , Adenoviridae/metabolismo , Proteína p53 Supresora de Tumor/genética , Telomerasa/genética , Telomerasa/metabolismo , Línea Celular Tumoral , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Adenosina Trifosfato , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo
2.
Mol Ther ; 31(7): 2077-2088, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37060176

RESUMEN

OBP-301 is an oncolytic adenovirus modified to replicate within cancer cells and lyse them. This open-label, non-comparative, phase I dose-escalation trial aimed to assess its safety and optimal dosage in 20 patients with advanced hepatocellular carcinoma. Good tolerance was shown with a maximum tolerated dose of 6 × 1012 viral particles. The most common treatment-emergent adverse events were influenza-like illness, pyrexia, fatigue, decreased platelet count, abdominal distension, and anemia. Cohorts 4 and 5 had approximately 50% higher levels of CD8+ T cells in the peripheral blood after injection. The best target response occurred in 14 patients, 4 of whom had progressive disease. Multiple intratumoral injections of OBP-301 were well tolerated in patients with advanced hepatocellular carcinoma. The stable disease rate for the injected tumors was greater than the overall response rate, even with no obvious tumor response. OBP-301 might have a greater impact on local response as histological examination revealed that the presence of OBP-301 was consistent with the necrotic area at the injection site. Increased infiltration of CD8+ T cells and <1% PD-L1 expression were observed in tumors after injection. Improved antitumor efficacy might be achieved in future studies via viral injection with volume adjustment and in combination with other immuno-therapeutics.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Viroterapia Oncolítica , Virus Oncolíticos , Telomerasa , Humanos , Adenoviridae/genética , Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Línea Celular Tumoral , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética
3.
Acta Med Okayama ; 78(2): 151-161, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38688833

RESUMEN

Soft-tissue sarcoma (STS) is a heterogeneous group of rare tumors originating predominantly from the embryonic mesoderm. Despite the development of combined modalities including radiotherapy, STSs are often refractory to antitumor modalities, and novel strategies that improve the prognosis of STS patients are needed. We previously demonstrated the therapeutic potential of two telomerase-specific replication-competent oncolytic adenoviruses, OBP-301 and tumor suppressor p53-armed OBP-702, in human STS cells. Here, we demonstrate in vitro and in vivo antitumor effects of OBP-702 in combination with ionizing radiation against human STS cells (HT1080, NMS-2, SYO-1). OBP-702 synergistically promoted the antitumor effect of ionizing radiation in the STS cells by suppressing the expression of B-cell lymphoma-X large (BCL-xL) and enhancing ionizing radiation-induced apoptosis. The in vivo experiments demonstrated that this combination therapy significantly suppressed STS tumors' growth. Our results suggest that OBP-702 is a promising antitumor reagent for promoting the radiosensitivity of STS tumors.


Asunto(s)
Viroterapia Oncolítica , Tolerancia a Radiación , Sarcoma , Proteína p53 Supresora de Tumor , Proteína bcl-X , Sarcoma/terapia , Sarcoma/radioterapia , Humanos , Viroterapia Oncolítica/métodos , Proteína bcl-X/genética , Proteína bcl-X/metabolismo , Línea Celular Tumoral , Animales , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ratones , Apoptosis , Adenoviridae/genética
4.
Cancer Immunol Immunother ; 72(5): 1285-1300, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36436021

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is often refractory to treatment with gemcitabine (GEM) and immune checkpoint inhibitors including anti-programmed cell death ligand 1 (PD-L1) antibody. However, the precise relationship between GEM-resistant PDAC and development of an immunosuppressive tumor microenvironment (TME) remains unclear. In this study, we investigated the immunosuppressive TME in parental and GEM-resistant PDAC tumors and assessed the therapeutic potential of combination therapy with the telomerase-specific replication-competent oncolytic adenovirus OBP-702, which induces tumor suppressor p53 protein and PD-L1 blockade against GEM-resistant PDAC tumors. Mouse PDAC cells (PAN02) and human PDAC cells (MIA PaCa-2, BxPC-3) were used to establish GEM-resistant PDAC lines. PD-L1 expression and the immunosuppressive TME were analyzed using parental and GEM-resistant PDAC cells. A cytokine array was used to investigate the underlying mechanism of immunosuppressive TME induction by GEM-resistant PAN02 cells. The GEM-resistant PAN02 tumor model was used to evaluate the antitumor effect of combination therapy with OBP-702 and PD-L1 blockade. GEM-resistant PDAC cells exhibited higher PD-L1 expression and produced higher granulocyte-macrophage colony-stimulating factor (GM-CSF) levels compared with parental cells, inducing an immunosuppressive TME and the accumulation of myeloid-derived suppressor cells (MDSCs). OBP-702 significantly inhibited GEM-resistant PAN02 tumor growth by suppressing GM-CSF-mediated MDSC accumulation. Moreover, combination treatment with OBP-702 significantly enhanced the antitumor efficacy of PD-L1 blockade against GEM-resistant PAN02 tumors. The present results suggest that combination therapy involving OBP-702 and PD-L1 blockade is a promising antitumor strategy for treating GEM-resistant PDAC with GM-CSF-induced immunosuppressive TME formation.


Asunto(s)
Carcinoma Ductal Pancreático , Células Supresoras de Origen Mieloide , Virus Oncolíticos , Neoplasias Pancreáticas , Ratones , Animales , Humanos , Gemcitabina , Células Supresoras de Origen Mieloide/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Inmunosupresores/uso terapéutico , Microambiente Tumoral , Línea Celular Tumoral , Neoplasias Pancreáticas
5.
Gan To Kagaku Ryoho ; 50(10): 1102-1103, 2023 Oct.
Artículo en Japonés | MEDLINE | ID: mdl-38035844

RESUMEN

Pancreatic ductal adenocarcinoma(PDAC)is lethal malignancy with abundant stroma. Cancer-associated fibroblasts (CAFs) exist in the PDAC stroma and contribute to progression of malignant transformation, treatment resistance, and recurrence. However, effective treatment to control PDAC stroma has not been established. We have developed tumor suppressor gene p53-armed oncolytic adenovirus(OBP-702), and have clarified therapeutic effects on PDAC cells. In this study, we investigate the therapeutic effect of OBP-702 on PDAC CAF.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Adenoviridae/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Páncreas/patología , Microambiente Tumoral , Neoplasias Pancreáticas
6.
Invest New Drugs ; 40(2): 300-307, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34613570

RESUMEN

Background Dysregulation of histone deacetylases (HDACs) is common in cancer and is critical to the development and progression of the majority of tumors. This first-in-human Phase Ia study assessed the safety, efficacy, and pharmacokinetics (PK) of OBP-801, a cyclic depsipeptide class I HDAC inhibitor. Methods Adult patients with advanced solid tumors were treated in 3 dose cohorts (1.0 mg/m2, 2.0 mg/m2 or 2.8 mg/m2) of OBP-801 that was administered via intravenous infusion weekly. Initially, an accelerated titration design was used that was followed by a 3 + 3 dose escalation strategy. Primary objective was assessment of safety. Secondary objectives included determination of PK and objective response rate. Results Seventeen patients were enrolled, of which 8 patients were evaluable for efficacy. Drug-related ≥ Grade 3 treatment-emergent adverse events included abdominal pain, anemia, fatigue, gamma glutamyl-transferase increase, hypertriglyceridemia and vomiting. No dose-limiting toxicity was observed in the 1.0 mg/m2 cohort. The PK data showed that OBP-801 and its active metabolite OBP-801-SH exposure increased proportionally and more than proportionally, respectively. No accumulation of either agent was noticed after repeat administration. Best response was stable disease (37.5%), with one patient each in the three cohorts. Conclusion Further investigations of the OBP-801 1.0 mg/m2 dose will be needed to better understand the efficacy of the agent, either alone or in combination. Trial registration: NCT02414516 (ClinicalTrials.gov) registered on April 10, 2015.


Asunto(s)
Depsipéptidos , Inhibidores de Histona Desacetilasas , Neoplasias , Adulto , Depsipéptidos/efectos adversos , Inhibidores de Histona Desacetilasas/efectos adversos , Humanos , Dosis Máxima Tolerada , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Péptidos Cíclicos/efectos adversos
7.
Mol Ther ; 29(10): 2920-2930, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34023506

RESUMEN

Extracellular vesicles (EVs) play important roles in various intercellular communication processes. The abscopal effect is an interesting phenomenon in cancer treatment, in which immune activation is generally considered a main factor. We previously developed a telomerase-specific oncolytic adenovirus, Telomelysin (OBP-301), and occasionally observed therapeutic effects on distal tumors after local treatment in immunodeficient mice. In this study, we hypothesized that EVs may be involved in the abscopal effect of OBP-301. EVs isolated from the supernatant of HCT116 human colon carcinoma cells treated with OBP-301 were confirmed to contain OBP-301, and they showed cytotoxic activity (apoptosis and autophagy) similar to OBP-301. In bilateral subcutaneous HCT116 and CT26 tumor models, intratumoral administration of OBP-301 produced potent antitumor effects on tumors that were not directly treated with OBP-301, involving direct mediation by tumor-derived EVs containing OBP-301. This indicates that immune activation is not the main factor in this abscopal effect. Moreover, tumor-derived EVs exhibited high tumor tropism in orthotopic HCT116 rectal tumors, in which adenovirus E1A and adenovirus type 5 proteins were observed in metastatic liver tumors after localized rectal tumor treatment. In conclusion, local treatment with OBP-301 has the potential to produce abscopal effects via tumor-derived EVs.


Asunto(s)
Adenoviridae/genética , Neoplasias del Colon/terapia , Vesículas Extracelulares/trasplante , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Animales , Línea Celular Tumoral , Supervivencia Celular , Vesículas Extracelulares/virología , Células HCT116 , Humanos , Ratones , Virus Oncolíticos/genética , Tropismo Viral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Acta Med Okayama ; 76(2): 203-215, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35503449

RESUMEN

The epithelial-mesenchymal transition (EMT), a normal biological process by which epithelial cells acquire a mesenchymal phenotype, is associated with migration, metastasis, and chemoresistance in cancer cells, and with poor prognosis in patients with esophageal cancer. However, therapeutic strategies to inhibit EMT in tumor environments remain elusive. Here, we show the therapeutic potential of telomerase-specific replication- competent oncolytic adenovirus OBP-301 in human esophageal cancer TE4 and TE6 cells with an EMT phenotype. Transforming growth factor-ß (TGF-ß) administration induced the EMT phenotype with spindleshaped morphology, upregulation of mesenchymal markers and EMT transcription factors, migration, and chemoresistance in TE4 and TE6 cells. OBP-301 significantly inhibited the EMT phenotype via E1 accumulation. EMT cancer cells were susceptible to OBP-301 via massive autophagy induction. OBP-301 suppressed tumor growth and lymph node metastasis of TE4 cells co-inoculated with TGF-ß-secreting fibroblasts. Our results suggest that OBP-301 inhibits the TGF-ß-induced EMT phenotype in human esophageal cancer cells. OBP-301-mediated E1A overexpression is a promising antitumor strategy to inhibit EMT-mediated esophageal cancer progression.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias Esofágicas , Adenoviridae/genética , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Neoplasias Esofágicas/patología , Humanos , Factor de Crecimiento Transformador beta/farmacología , Factores de Crecimiento Transformadores
9.
Gan To Kagaku Ryoho ; 49(10): 1127-1129, 2022 Oct.
Artículo en Japonés | MEDLINE | ID: mdl-36281608

RESUMEN

Pancreatic cancer has poor prognosis despite the various developed multimodal treatment strategies. Currently, neoadjuvant chemotherapy and immunotherapy have attracted substantial attention as effective treatment strategies. However, amplifying immune response with existing treatments is difficult. We developed telomerase-specific oncolytic adenoviruses (OAs), including OBP-301 that is currently tested in a clinical trial of combined anti-PD-1 antibody and p53-armed OBP- 301 variant(OBP-702). OAs have immune-modulation functions and induce CD8+ T cells into tumors by releasing immunogenic cell death markers, such as extracellular adenosine triphosphate. Here, we investigated the effectiveness of OBP- 702 in pancreatic cancer treatments, focusing on the influence on CD8+ memory T cells.


Asunto(s)
Viroterapia Oncolítica , Neoplasias Pancreáticas , Telomerasa , Humanos , Adenosina Trifosfato/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Linfocitos T CD8-positivos , Línea Celular Tumoral , Genes p53 , Células T de Memoria , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Telomerasa/genética , Telomerasa/metabolismo , Proteína p53 Supresora de Tumor/genética , Ensayos Clínicos como Asunto , Neoplasias Pancreáticas
10.
Cancer Immunol Immunother ; 70(5): 1405-1417, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33151368

RESUMEN

Immune checkpoint inhibitors including anti-programmed cell death 1 (PD-1) antibody have recently improved clinical outcome in certain cancer patients; however, osteosarcoma (OS) patients are refractory to PD-1 blockade. Oncolytic virotherapy has emerged as novel immunogenic therapy to augment antitumor immune response. We developed a telomerase-specific replication-competent oncolytic adenovirus OBP-502 that induces lytic cell death via binding to integrins. In this study, we assessed the combined effect of PD-1 blockade and OBP-502 in OS cells. The expression of coxsackie and adenovirus receptor (CAR), integrins αvß3 and αvß5, and programmed cell death ligand 1 (PD-L1) was analyzed in two murine OS cells (K7M2, NHOS). The cytopathic activity of OBP-502 in both cells was analyzed using the XTT assay. OBP-502-induced immunogenic cell death was assessed by analyzing the level of extracellular ATP and high-mobility group box protein B1 (HMGB1). Subcutaneous tumor models for K7M2 and NHOS cells were used to evaluate the antitumor effect and number of tumor-infiltrating CD8+ cells in combination therapy. K7M2 and NHOS cells showed high expression of integrins αvß3 and αvß5, but not CAR. OBP-502 significantly suppressed the viability of both cells, in which PD-L1 expression and the release of ATP and HMGB1 were significantly increased. Intratumoral injection of OBP-502 significantly augmented the efficacy of PD-1 blockade on subcutaneous K2M2 and NHOS tumor models via enhancement of tumor-infiltrating CD8+ T cells. Our results suggest that telomerase-specific oncolytic virotherapy is a promising antitumor strategy to promote the efficacy of PD-1 blockade in OS.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Viroterapia Oncolítica/métodos , Osteosarcoma/terapia , Neoplasias Cutáneas/terapia , Adenoviridae/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Telomerasa/genética
11.
Mol Ther ; 28(3): 794-804, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31991110

RESUMEN

The clinical benefit of monotherapy involving immune checkpoint inhibitors (ICIs) such as anti-programmed death-1 antibody (PD-1 Ab) is limited to small populations. We previously developed a telomerase-specific oncolytic adenovirus, Telomelysin (OBP-301), the safety of which was confirmed in a phase I clinical study. Here, we examined the potential of OBP-502, an OBP-301 variant, as an agent for inducing immunogenic cell death (ICD) and synergistically enhancing the efficacy of OBP-502 with PD-1 Ab using CT26 murine colon cancer and PAN02 murine pancreatic cancer cell lines. OBP-502 induced the release of ICD molecules such as adenosine triphosphate (ATP) and high-mobility group box protein 1 (HMGB1) from CT26 and PAN02 cells, leading to recruitment of CD8-positive lymphocytes and inhibition of Foxp3-positive lymphocyte infiltration into tumors. Combination therapy involving OBP-502 intratumoral administration and PD-1 Ab systemic administration significantly suppressed the growth of not only OBP-502-treated tumors but also tumors not treated with OBP-502 (so-called abscopal effect) in CT26 and PAN02 bilateral subcutaneous tumor models, in which active recruitment of CD8-positve lymphocytes was observed even in tumors not treated with OBP-502. This combined efficacy was similar to that observed in a CT26 rectal orthotopic tumor model involving liver metastases. In conclusion, telomerase-specific oncolytic adenoviruses are promising candidates for combined therapies with ICIs.


Asunto(s)
Adenoviridae/inmunología , Antineoplásicos Inmunológicos/farmacología , Terapia Genética , Inmunomodulación , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Telomerasa/inmunología , Adenoviridae/genética , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Terapia Combinada , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Terapia Genética/efectos adversos , Terapia Genética/métodos , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Sci ; 109(1): 231-240, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29151279

RESUMEN

Circulating tumor cells (CTC) are newly discovered biomarkers of cancers. Although many systems detect CTC, a gold standard has not yet been established. We analyzed CTC in uterine cervical cancer patients using an advanced version of conditionally replicative adenovirus targeting telomerase-positive cells, which was enabled to infect coxsackievirus-adenovirus receptor-negative cells and to reduce false-positive signals in myeloid cells. Blood samples from cervical cancer patients were hemolyzed and infected with the virus and then labeled with fluorescent anti-CD45 and anti-pan cytokeratin antibodies. GFP (+)/CD45 (-) cells were isolated and subjected to whole-genome amplification followed by polymerase chain reaction analysis of human papillomavirus (HPV) DNA. CTC were detected in 6 of 23 patients with cervical cancers (26.0%). Expression of CTC did not correlate with the stage of cancer or other clinicopathological factors. In 5 of the 6 CTC-positive cases, the same subtype of HPV DNA as that of the corresponding primary lesion was detected, indicating that the CTC originated from HPV-infected cancer cells. These CTC were all negative for cytokeratins. The CTC detected by our system were genetically confirmed. CTC derived from uterine cervical cancers had lost epithelial characteristics, indicating that epithelial marker-dependent systems do not have the capacity to detect these cells in cervical cancer patients.


Asunto(s)
Infecciones por Adenoviridae/genética , Adenoviridae/fisiología , Células Neoplásicas Circulantes/metabolismo , Telomerasa/genética , Neoplasias del Cuello Uterino/sangre , Adenoviridae/genética , Línea Celular Tumoral , Femenino , Células HeLa , Humanos , Queratinas/metabolismo , Replicación Viral
13.
Cancer Sci ; 109(10): 3263-3271, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30076658

RESUMEN

Gastric cancer patients positive for peritoneal cytology are at increased risk of tumor recurrence, but although a certain proportion of cytology-positive patients relapse rapidly with aggressive progression, others survive longer with conventional chemotherapies. This heterogeneity makes it difficult to stratify patients for more intensive therapy and poses a substantial challenge for the implementation of precision medicine. We developed a new approach to identify biologically malignant subpopulations in cytology-positive gastric cancer patients, using a green fluorescent protein (GFP)-expressing attenuated adenovirus in which the telomerase promoter regulates viral replication (TelomeScan, OBP-401). The fluorescence emitted from TelomeScan-positive cells was successfully quantified using a multi-mode microplate reader. We then analyzed clinical peritoneal washes obtained from 68 gastric cancer patients and found that patients positive for TelomeScan had a significantly worse prognosis. In 21 cytology-positive patients, the median survival time of those who were TelomeScan positive (235 days) was significantly shorter than that for those who were TelomeScan negative (671 days; P = 0.0062). This fluorescent virus-guided cytology detects biologically malignant cancer cells from the peritoneal washes of gastric cancer patients and may thus be useful for both therapy stratification and precision medicine approaches based on genetic profiling of disseminated cells.


Asunto(s)
Citodiagnóstico/métodos , Vectores Genéticos/química , Recurrencia Local de Neoplasia/patología , Neoplasias Peritoneales/patología , Neoplasias Gástricas/patología , Adenoviridae/genética , Anciano , Productos Biológicos/química , Línea Celular Tumoral , Separación Celular , Femenino , Citometría de Flujo/métodos , Técnica del Anticuerpo Fluorescente/métodos , Perfilación de la Expresión Génica , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Nanomedicina/métodos , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/cirugía , Lavado Peritoneal , Neoplasias Peritoneales/mortalidad , Neoplasias Peritoneales/secundario , Proyectos Piloto , Pronóstico , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ADN , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/cirugía , Análisis de Supervivencia , Telomerasa/genética
14.
Biol Pharm Bull ; 41(10): 1615-1619, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30270333

RESUMEN

In recent times, oncolytic viruses expressing an extraneous gene have attracted great interest; in fact, they have been engaged in multiple applications, such as medicine for cancer. Our group made an oncolytic adenovirus, namely, OBP-301, for use in treating solid cancers and press clinical trial to get approval for a pharmaceutical product. In this study, we applied a flow cytometry-based method to determine the titer of adenoviruses expressing an extraneous gene as well as assess their quality. We considered using the green fluorescent protein (GFP)50 titer as a measure of viral quality. The GFP50 titer (GFP50/mL) is the viral load required to render the HeLa S3 cell line 50% GFP-positive by analysing flow cytometry data. We measured the GFP50 titers for three types of recombinant adenoviruses (OBP-401, OBP-1101, and OBP-1106). We compared GFP50/mL and tissue culture infectious dose (TCID50/mL), a conventional titration index, and found that these titers showed a linear correlation, with a correlation coefficient of >0.9. Moreover, GFP50/mL showed high repetitive accuracy. We expect this flow cytometry-based method to be useful in case of clinically relevant viruses expressing an extraneous gene, in particular, to control viral quality.


Asunto(s)
Adenoviridae/genética , Proteínas Fluorescentes Verdes/genética , Virus Oncolíticos/genética , Infecciones por Adenoviridae/virología , Línea Celular Tumoral , Citometría de Flujo/métodos , Células HeLa , Humanos , Microorganismos Modificados Genéticamente , Carga Viral
15.
J Cell Biochem ; 118(11): 3635-3642, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-27171483

RESUMEN

We have established an orthotopic nude-mouse model of gastric cancer carcinomatosis peritonitis, a recalcitrant disease in human patients. Human MKN45 poorly-differentiated human gastric cancer cells developed carcinomatosis peritonitis upon orthotopic transplantation in nude mice. The MKN45 cells expressed the fluorescent ubiquitination-based cell cycle indicator (FUCCI) that color codes the phases of the cell cycle. The intra-peritoneal tumors and ascites contained mostly quiescent G1 /Go cancer cells visualized as red by FUCCI imaging. Cisplatinum (CDDP) treatment did not reduce bloody ascites, and larger tumors formed in the peritoneal cavity after CDDP treatment in an early-stage carcinomatosis peritonitis orthotopic mouse model. Paclitaxel-treated mice had reduced ascites, but also had large tumor masses in the peritonium after treatment with cancer cells mostly in G0 /G1 , visualized by FUCCI red. In contrast, OBP-301 telomerase-dependent adenovirus-treated mice had no ascites and only small tumor nodules consisting of cancer cells mostly in S/G2 phases in the early-stage carcinomatosis peritonitis model, visualized by FUCCI green. Furthermore, OBP-301 significantly reduced the size of tumors (P < 0.01) and ascites even in a late-stage carcinomatosis peritonitis model. These results suggest that quiescent peritoneally-disseminated gastric cancer cells are resistant to conventional chemotherapy, but OBP-301 significantly reduced the weight of the tumors and increased survival, suggesting clinical potential. J. Cell. Biochem. 118: 3635-3642, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Adenocarcinoma/enzimología , Adenoviridae , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/enzimología , Neoplasias Peritoneales/enzimología , Neoplasias Gástricas/enzimología , Telomerasa/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Fase G2/genética , Humanos , Ratones Desnudos , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/patología , Fase S/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Telomerasa/genética
16.
Cancer Sci ; 108(9): 1870-1880, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28685948

RESUMEN

Osteosarcoma is an aggressive malignant bone tumor that causes bone destruction. Although tumor-specific replicating oncolytic adenovirus OBP-301 induces an antitumor effect in an osteosarcoma tumor, it cannot prevent bone destruction. Zoledronic acid (ZOL) is a clinically available agent that inhibits bone destruction. In this study, we investigated the potential of combination therapy with OBP-301 and ZOL against osteosarcomas with bone destruction. The antitumor activity of OBP-301 and ZOL in monotherapy or combination therapy was assessed using three human osteosarcoma cell lines (143B, MNNG/HOS, SaOS-2). The cytotoxic effect of OBP-301 and/or ZOL was measured by assay of cell apoptosis. The effect of OBP-301 and ZOL on osteoclast activation was investigated. The potential of combination therapy against tumor growth and bone destruction was analyzed using an orthotopic 143B osteosarcoma xenograft tumor model. OBP-301 and ZOL decreased the viability of human osteosarcoma cells. Combination therapy with OBP-301 and ZOL displayed a synergistic antitumor effect, in which OBP-301 promoted apoptosis through suppression of anti-apoptotic myeloid cell leukemia 1 (MCL1). Combination therapy significantly inhibited tumor-mediated osteoclast activation, tumor growth and bone destruction compared to monotherapy. These results suggest that combination therapy of OBP-301 and ZOL suppresses osteosarcoma progression via suppression of MCL1 and osteoclast activation.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/terapia , Difosfonatos/farmacología , Imidazoles/farmacología , Osteosarcoma/terapia , Adenoviridae/genética , Animales , Apoptosis , Neoplasias Óseas/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Terapia Combinada , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Ratones , Ratones Desnudos , Viroterapia Oncolítica , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Osteosarcoma/patología , Ligando RANK/farmacología , Células RAW 264.7 , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Ácido Zoledrónico
17.
Mol Ther ; 23(7): 1182-1188, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25896244

RESUMEN

Fluorescence-guided surgery (FGS) of cancer is an area of intense current interest. However, although benefits have been demonstrated with FGS, curative strategies need to be developed. Glioblastoma multiforme (GBM) is one of the most invasive of cancers and is not totally resectable using standard bright-light surgery (BLS) or current FGS strategies. We report here a curative strategy for FGS of GBM. In this study, telomerase-dependent adenovirus OBP-401 infection brightly and selectively labeled GBM with green fluorescent protein (GFP) for FGS in orthotopic nude mouse models. OBP-401-based FGS enabled curative resection of GBM without recurrence for at least 150 days, compared to less than 30 days with BLS.


Asunto(s)
Fluorescencia , Glioblastoma/cirugía , Proteínas Fluorescentes Verdes/uso terapéutico , Recurrencia Local de Neoplasia/cirugía , Adenoviridae , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Supervivencia sin Enfermedad , Glioblastoma/patología , Proteínas Fluorescentes Verdes/química , Humanos , Ratones , Ratones Desnudos , Recurrencia Local de Neoplasia/patología , Imagen Óptica
18.
Mol Ther ; 23(3): 501-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25523761

RESUMEN

Currently, early gastrointestinal cancers are treated endoscopically, as long as there are no lymph node metastases. However, once a gastrointestinal cancer invades the submucosal layer, the lymph node metastatic rate rises to higher than 10%. Therefore, surgery is still the gold standard to remove regional lymph nodes containing possible metastases. Here, to avoid prophylactic surgery, we propose a less-invasive biological ablation of lymph node metastasis in submucosally invaded gastrointestinal cancer patients. We have established an orthotopic early rectal cancer xenograft model with spontaneous lymph node metastasis by implantation of green fluorescent protein (GFP)-labeled human colon cancer cells into the submucosal layer of the murine rectum. A solution containing telomerase-specific oncolytic adenovirus was injected into the peritumoral submucosal space, followed by excision of the primary rectal tumors mimicking the endoscopic submucosal dissection (ESD) technique. Seven days after treatment, GFP signals had completely disappeared indicating that sentinel lymph node metastasis was selectively eradicated. Moreover, biologically treated mice were confirmed to be relapse-free even 4 weeks after treatment. These results indicate that virus-mediated biological ablation selectively targets lymph node metastasis and provides a potential alternative to surgery for submucosal invasive gastrointestinal cancer patients.


Asunto(s)
Adenoviridae/genética , Neoplasias del Colon/terapia , Neoplasias Gastrointestinales/terapia , Ganglios Linfáticos/metabolismo , Recurrencia Local de Neoplasia/prevención & control , Viroterapia Oncolítica/métodos , Animales , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias del Colon/cirugía , Modelos Animales de Enfermedad , Femenino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Mucosa Gástrica/cirugía , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/patología , Neoplasias Gastrointestinales/cirugía , Expresión Génica , Genes Reporteros , Vectores Genéticos/administración & dosificación , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ganglios Linfáticos/patología , Ganglios Linfáticos/cirugía , Metástasis Linfática , Ratones , Ratones Desnudos , Ratones Transgénicos , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Trasplante de Neoplasias , Biopsia del Ganglio Linfático Centinela , Telomerasa/genética , Telomerasa/metabolismo
19.
Gut ; 64(4): 627-35, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24870621

RESUMEN

BACKGROUND: Molecular-based companion diagnostic tests are being used with increasing frequency to predict their clinical response to various drugs, particularly for molecularly targeted drugs. However, invasive procedures are typically required to obtain tissues for this analysis. Circulating tumour cells (CTCs) are novel biomarkers that can be used for the prediction of disease progression and are also important surrogate sources of cancer cells. Because current CTC detection strategies mainly depend on epithelial cell-surface markers, the presence of heterogeneous populations of CTCs with epithelial and/or mesenchymal characteristics may pose obstacles to the detection of CTCs. METHODS: We developed a new approach to capture live CTCs among millions of peripheral blood leukocytes using a green fluorescent protein (GFP)-expressing attenuated adenovirus, in which the telomerase promoter regulates viral replication (OBP-401, TelomeScan). RESULTS: Our biological capturing system can image epithelial and mesenchymal tumour cells with telomerase activities as GFP-positive cells. After sorting, direct sequencing or mutation-specific PCR can precisely detect different mutations in KRAS, BRAF and KIT genes in epithelial, mesenchymal or epithelial-mesenchymal transition-induced CTCs, and in clinical blood samples from patients with colorectal cancer. CONCLUSIONS: This fluorescence virus-guided viable CTC capturing method provides a non-invasive alternative to tissue biopsy or surgical resection of primary tumours for companion diagnostics.


Asunto(s)
Adenoviridae , Neoplasias Colorrectales/patología , Proteínas Fluorescentes Verdes , Células Neoplásicas Circulantes/patología , Adenoviridae/metabolismo , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/biosíntesis , Humanos , Técnicas de Diagnóstico Molecular/métodos
20.
Sci Rep ; 14(1): 10199, 2024 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702437

RESUMEN

In pancreatic ductal adenocarcinoma (PDAC) patients, the importance of peritoneal lavage cytology, which indicates unresectability, remains controversial. This study sought to determine whether positive peritoneal lavage cytology (CY+) precludes pancreatectomy. Furthermore, we propose a novel liquid biopsy using peritoneal lavage fluid to detect viable peritoneal tumor cells (v-PTCs) with TelomeScan F35, a telomerase-specific replication-selective adenovirus engineered to express green fluorescent protein. Resectable cytologically or histologically proven PDAC patients (n = 53) were enrolled. CY was conducted immediately following laparotomy. The resulting fluid was examined by conventional cytology (conv-CY; Papanicolaou staining and MOC-31 immunostaining) and by the novel technique (Telo-CY; using TelomeScan F35). Of them, 5 and 12 were conv-CY+ and Telo-CY+, respectively. All underwent pancreatectomy. The two double-CY+ (conv-CY+ and Telo-CY+) patients showed early peritoneal recurrence (P-rec) postoperatively, despite adjuvant chemotherapy. None of the three conv-CY+ Telo-CY- patients exhibited P-rec. Six of the 10 Telo-CY+ conv-CY- patients (60%) relapsed with P-rec. Of the remaining 38 double-CY- [conv-CY-, Telo-CY-, conv-CY± (Class III)] patients, 3 (8.3%) exhibited P-rec. Although conv-CY+ status predicted poor prognosis and a higher risk of P-rec, Telo-CY was more sensitive for detecting v-PTC. Staging laparoscopy and performing conv-CY and Telo-CY are needed to confirm the indication for pancreatectomy.


Asunto(s)
Carcinoma Ductal Pancreático , Pancreatectomía , Neoplasias Pancreáticas , Lavado Peritoneal , Humanos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/cirugía , Masculino , Femenino , Anciano , Persona de Mediana Edad , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/diagnóstico , Citodiagnóstico/métodos , Anciano de 80 o más Años , Recurrencia Local de Neoplasia/patología , Biopsia Líquida/métodos , Neoplasias Peritoneales/patología , Neoplasias Peritoneales/diagnóstico , Adulto , Citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA