Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 805
Filtrar
1.
Front Immunol ; 15: 1473288, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39430750

RESUMEN

Lung cancer accounts for the highest cancer-related mortality worldwide. While immunotherapies targeting anti-tumor immune responses have demonstrated efficacy in clinical practice, the demand for novel treatment modalities remains urgent. Oncolytic viruses (OVs), which selectively kill tumor cells while stimulating an anti-tumor immune response, represent a potential breakthrough in lung cancer therapy. The induction of anti-tumor immunity by OVs is central to their overall therapeutic effectiveness. Many natural receptors on the surface of cancer cells are dysregulated, providing potential entry points for OVs. Furthermore, the inherent dysregulation of some key signaling pathways in lung cancer cells promotes proliferation, progression and metastasis, which may facilitate selective viral replication. In this review, we explore the application of OVs in lung cancer by analyzing several major OVs and their corresponding entry receptors. Then, we also examine the key signaling pathways and molecules with the potential to synergize with OVs in modulating the immune tumor microenvironment. Finally, we discuss the combination and administration strategies that warrant further clinical trials for validation. Despite certain limitations, the tolerability of OVs positions virotherapy as a promising avenue in the future of lung cancer treatment.


Asunto(s)
Neoplasias Pulmonares , Viroterapia Oncolítica , Virus Oncolíticos , Transducción de Señal , Internalización del Virus , Humanos , Viroterapia Oncolítica/métodos , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/inmunología , Virus Oncolíticos/inmunología , Virus Oncolíticos/fisiología , Animales , Microambiente Tumoral/inmunología
2.
Exp Oncol ; 46(2): 81-86, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39396176

RESUMEN

Oncolytic viruses (OVs) are emerging as novel tools in cancer therapy. Oncolytic virotherapy offers an attractive therapeutic combination of tumor-specific killing and immune co-stimulation, therefore amplifying the host immune response against tumors. Moreover, OVs can be engineered for the expression of different immunostimulatory molecules to optimize and enhance the efficacy of oncolytic virotherapy. The effectiveness of OVs has been demonstrated in many preclinical studies for different types of cancers to achieve the aim of personalized cancer therapy. Human respiratory syncytial virus (RSV), an RNA virus of the Pneumoviridae family causes severe lower respiratory tract infections in infants and immunocompromised individuals. Interestingly, the oncolytic activity of RSV demonstrated in human prostate, hepatocellular, and dermal cancer cells is mostly mediated via apoptotic cell death associated with the impaired NF-κB activation or with the defect of the IFNα/ß-induced STAT-1 activation. At the same time, the studies on cervical cancer revealed that RSV infection resulted in autophagy activation and apoptosis through the ROS-BAX and TNF- α-mediated pathways. The rational combinations of OVs, including RSV, with other approaches may benefit patients whose response to conventional therapies is limited. Here, we discuss the oncolytic activity of RSV and its potential use against different types of cancer.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Virus Sincitial Respiratorio Humano , Humanos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Neoplasias/terapia , Animales , Infecciones por Virus Sincitial Respiratorio/terapia , Infecciones por Virus Sincitial Respiratorio/virología
3.
Virol J ; 21(1): 245, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39369233

RESUMEN

BACKGROUND: Cervical squamous cell carcinoma (CSCC) is a prevalent gynecological malignancy worldwide. Current treatments for CSCC can impact fertility and cause long-term complications, underscoring the need for new therapeutic strategies. Oncolytic virotherapy has emerged as a promising option for cancer treatment. Previous research has demonstrated the oncolytic activity of the coxsackievirus B3 strain 2035 A (CVB3/2035A) against various tumor types. This study aims to evaluate the clinical viability of CVB3/2035A for CSCC treatment, focusing on its oncolytic effect in patient-derived CSCC organoids. METHODS: The oncolytic effects of CVB3/2035A were investigated using human CSCC cell lines in vitro and mouse xenograft models in vivo. Preliminary tests for tumor-selectivity were conducted on patient-derived CSCC tissue samples and compared to normal cervical tissues ex vivo. Three patient-derived CSCC organoid lines were developed and treated with CVB3/2035A alone and in combination with paclitaxel. Both cytotoxicity and virus replication were evaluated in vitro. RESULTS: CVB3/2035A exhibited significant cytotoxic effects in human CSCC cell lines and xenograft mouse models. The virus selectively induced oncolysis in patient-derived CSCC tissue samples while sparing normal cervical tissues ex vivo. In patient-derived CSCC organoids, which retained the immunohistological characteristics of the original tumors, CVB3/2035A also demonstrated significant cytotoxic effects and efficient replication, as evidenced by increased viral titers and presence of viral nucleic acids and proteins. Notably, the combination of CVB3/2035A and paclitaxel resulted in enhanced cytotoxicity and viral replication. CONCLUSIONS: CVB3/2035A showed oncolytic activity in CSCC cell lines, xenografts, and patient-derived tissue cultures and organoids. Furthermore, the virus exhibited synergistic anti-tumor effects with paclitaxel against CSCC. These results suggest CVB3/2035A could serve as an alternative or adjunct to current CSCC chemotherapy regimens.


Asunto(s)
Carcinoma de Células Escamosas , Enterovirus Humano B , Viroterapia Oncolítica , Virus Oncolíticos , Organoides , Paclitaxel , Neoplasias del Cuello Uterino , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Neoplasias del Cuello Uterino/virología , Neoplasias del Cuello Uterino/terapia , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Femenino , Organoides/virología , Ratones , Enterovirus Humano B/fisiología , Enterovirus Humano B/efectos de los fármacos , Viroterapia Oncolítica/métodos , Carcinoma de Células Escamosas/virología , Carcinoma de Células Escamosas/terapia , Carcinoma de Células Escamosas/tratamiento farmacológico , Virus Oncolíticos/fisiología , Línea Celular Tumoral , Replicación Viral/efectos de los fármacos
4.
Cell Commun Signal ; 22(1): 514, 2024 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-39434159

RESUMEN

BACKGROUND: Gastric cancer (GC) is a leading malignant disease in numerous countries, including Taiwan with limited therapeutic options. Animal viruses including oncolytic avian reovirus (ARV) have the possibility to avoid pre-existing immunity in humans, while being safe and immunostimulatory. Here, we provide a novel insight into oncolytic ARV and UV-ARV-sensitized patient's peripheral blood mononuclear cells (P-PBMCs) and tumor infiltrating lymphocytes (TILs) killing primary GC (PGC) cells through the surface TLR3 and TRAIL/DR4/DR5 immunogenic apoptosis pathway. METHODS: We conducted a comprehensive study to reveal whether ARV- or UV-inactivated ARV (UV-ARV)-modulated P-PBMCs or TILs killing ARV- and UV-ARV-sensitized AGS cells and PGC cells derived from clinical patients and to investigate the regulation of surface TLR3 receptor and upstream signaling pathways. Apoptosis analysis by flow cytometry and Western blot, suppression of signal pathway by specific inhibitors, in situ proximity ligation assay (PLA), time-resolved flurometry and lactate dehydrogenase (LDH) cytotoxicity assays, and an in vitro co-culture model were established to study the interplay between ARV- and UV-ARV-sensitized P-PBMCs and TILs to kill PGC cells and their upstream pathways. RESULTS: Our results reveal that increased levels of DR4 and DR5 were observed in ARV and UV-ARV sensitized PGC cells through the TLR3/p38/p53 signaling pathway. Importantly, we found that the σC protein of ARV or UV-ARV interacted with surface TLR3 of CD8+ TILs, thereby triggering the TLR3/NF-κB/IFN-γ/TRAIL signaling pathway which induces immunogenic apoptosis of PGC cells. This study sheds further light on the molecular basis behind ARV oncolysis and facilitates the ARV or UV-ARV as a cancer therapeutic. CONCLUSIONS: The study provides novel insights into ARV- or UV-ARV-sensitized P-PBMCs and CD8+ TILs to kill PGC cells through the immunogenic apoptosis pathway. We conclude that P-PBMCs can easily be obtained from GC patients and provide a rich source as TILs to kill PGC cells.


Asunto(s)
Apoptosis , Linfocitos T CD8-positivos , Linfocitos Infiltrantes de Tumor , Orthoreovirus Aviar , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Neoplasias Gástricas/virología , Linfocitos T CD8-positivos/inmunología , Orthoreovirus Aviar/fisiología , Orthoreovirus Aviar/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptor Toll-Like 3/metabolismo , Virus Oncolíticos/fisiología , Virus Oncolíticos/inmunología , Línea Celular Tumoral , Transducción de Señal , Viroterapia Oncolítica , Animales
5.
Int J Mol Sci ; 25(20)2024 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-39457007

RESUMEN

Treatment of glioblastoma is ineffective. Myx-M011L-KO/EGFP, a myxoma virus actively inducing apoptosis in BTICs linked to recurrence, offers innovative treatment. We loaded this construct into adipose-derived stem cells (ADSCs) to mitigate antiviral host responses and enable systemic delivery. The apoptotic and cytotoxic effects of the construct were studied using murine and human glioblastoma cell lines. Before implementing systemic delivery, we delivered the construct locally using ADSC to verify elimination of orthotopic murine glioma lesions. vMyx-M011L-KO/EGFP was cytotoxic to a murine cell line, preventing effective virus multiplication. In three human glioma cell lines, viral replication did occur, coupled with cell killing. The knock-out construct induced apoptotic cell death in these cultures. ADSCs infected ex vivo were shown to be sufficiently migratory to assure transfer of the therapeutic cargo to murine glioma lesions. Virus-loaded ADSCs applied to the artificial blood-brain barrier (BBB) yielded viral infection of glioma cells grown distally in the wells. Two rounds of local administration of this therapeutic platform starting 6 days post tumor implantation slowed down growth of orthotopic lesions and improved survival (total recovery < 20%). ADSCs infected ex vivo with vMyx-M011L-KO/EGFP show promise as a therapeutic tool in systemic elimination of glioma lesions.


Asunto(s)
Apoptosis , Barrera Hematoencefálica , Glioma , Myxoma virus , Viroterapia Oncolítica , Animales , Barrera Hematoencefálica/metabolismo , Myxoma virus/genética , Myxoma virus/fisiología , Ratones , Glioma/terapia , Glioma/patología , Humanos , Línea Celular Tumoral , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patología , Tejido Adiposo/citología , Células Madre/virología , Células Madre/citología
6.
Int J Mol Sci ; 25(20)2024 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-39457005

RESUMEN

Pancreatic cancer is one of the deadliest cancers globally, with limited success from existing therapies, including chemotherapies and immunotherapies like checkpoint inhibitors for patients with advanced pancreatic ductal adenocarcinoma (PDAC). A promising new approach is the use of oncolytic viruses (OV), a form of immunotherapy that has been demonstrated clinical effectiveness in various cancers. Here we investigated the potential of the oncolytic coxsackievirus B3 strain (CVB3) PD-H as a new treatment for pancreatic cancer. In vitro, PD-H exhibited robust replication, as measured by plaque assays, and potent lytic activity, as assessed by XTT assays, in most pancreatic tumor cell lines, outperforming two other coxsackievirus strains tested, H3N-375/1TS and CVA21. Thus, H3N-375/1TS showed efficient replication and lytic efficiency in distinctly fewer tumor cell lines, while most tumor cells were resistant to CVA21. The oncolytic efficiency of the three OV largely correlated with mRNA expression levels of viral receptors and their ability to induce apoptosis, as measured by cleaved caspase 3/7 activity in the tumor cells. In a syngeneic mouse model with subcutaneous pancreatic tumors, intratumoral administration of PD-H significantly inhibited tumor growth but did not completely stop tumor progression. Importantly, no virus-related side effects were observed. Although pancreatic tumors respond to PD-H treatment, its therapeutic efficacy is limited. Combining PD-H with other treatments, such as those aiming at reducing the desmoplastic stroma which impedes viral infection and spread within the tumor, may enhance its efficacy.


Asunto(s)
Enterovirus Humano B , Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias Pancreáticas , Animales , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/patología , Humanos , Viroterapia Oncolítica/métodos , Ratones , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Línea Celular Tumoral , Enterovirus Humano B/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto , Apoptosis , Replicación Viral , Carcinoma Ductal Pancreático/terapia , Carcinoma Ductal Pancreático/patología
7.
Int J Mol Sci ; 25(18)2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39337546

RESUMEN

Newcastle disease virus (NDV) is a highly pathogenic avian infectious disease agent and also a promising oncolytic virus with broad application prospects. The Endosomal Sorting Complex Required for Transport (ESCRT) machinery has been increasingly recognized for its crucial role in the life cycles of enveloped viruses, influencing processes such as viral entry, replication, and budding. In this study, we employed an RNA interference screening approach to identify key ESCRT components that regulate NDV replication in tumor cells. qPCR, immunofluorescence, and Western blot assays demonstrated that knockdown of HRS, CHMP4A, CHMP4B, and CHMP4C significantly impaired NDV replication in HeLa cells, with HRS exhibiting the most pronounced inhibitory effect. Additionally, HRS knockout significantly inhibited viral budding and suppressed NDV-induced cell death in HeLa cells. Notably, NDV infection was shown to significantly upregulate HRS gene and protein expression in a time-dependent manner. In conclusion, this study systematically identifies critical ESCRT components involved in NDV replication within tumor cells, with a particular focus on the role of HRS in promoting NDV's replication by promoting viral budding, offering new insights for the development of NDV-based oncolytic therapies.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte , Virus de la Enfermedad de Newcastle , Liberación del Virus , Replicación Viral , Virus de la Enfermedad de Newcastle/fisiología , Virus de la Enfermedad de Newcastle/genética , Humanos , Células HeLa , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Virus Oncolíticos/fisiología , Virus Oncolíticos/genética , Animales
8.
J Gen Virol ; 105(8)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39172037

RESUMEN

Rhabdoviral vectors can induce lysis of cancer cells. While studied almost exclusively at 37 °C, viruses are subject to a range of temperatures in vivo, including temperatures ≤31 °C. Despite potential implications, the effect of temperatures <37 °C on the performance of rhabdoviral vectors is unknown. We investigated the effect of low anatomical temperatures on two rhabdoviruses, vesicular stomatitis virus (VSV) and Maraba virus (MG1). Using a metabolic resazurin assay, VSV- and MG1-mediated oncolysis was characterized in a panel of cell lines at 28, 31, 34 and 37 °C. The oncolytic ability of both viruses was hindered at 31 and 28 °C. Cold adaptation of both viruses was attempted as a mitigation strategy. Viruses were serially passaged at decreasing temperatures in an attempt to induce mutations. Unfortunately, the cold-adaptation strategies failed to potentiate the oncolytic activity of the viruses at temperatures <37 °C. Interestingly, we discovered that viral replication was unaffected at low temperatures despite the abrogation of oncolytic activity. In contrast, the proliferation of cancer cells was reduced at low temperatures. Equivalent oncolytic effects could be achieved if cells at low temperatures were treated with viruses for longer times. This suggests that rhabdovirus-mediated oncolysis could be compromised at low temperatures in vivo where therapeutic windows are limited.


Asunto(s)
Frío , Virus Oncolíticos , Rhabdoviridae , Replicación Viral , Humanos , Rhabdoviridae/fisiología , Rhabdoviridae/genética , Animales , Virus Oncolíticos/fisiología , Virus Oncolíticos/genética , Vesiculovirus/fisiología , Vesiculovirus/genética , Viroterapia Oncolítica/métodos , Línea Celular , Vectores Genéticos/genética , Línea Celular Tumoral , Temperatura
9.
Viruses ; 16(8)2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39205238

RESUMEN

Proliferating cell nuclear antigen (PCNA) is a well-documented accessory protein of DNA repair and replication. It belongs to the sliding clamp family of proteins that encircle DNA and acts as a mobile docking platform for interacting proteins to mount and perform their metabolic tasks. PCNA presence is ubiquitous to all cells, and when located in the nucleus it plays a role in DNA replication and repair, cell cycle control and apoptosis in proliferating cells. It also plays a crucial role in the infectivity of some viruses, such as herpes simplex viruses (HSVs). However, more recently it has been found in the cytoplasm of immune cells such as neutrophils and macrophages where it has been shown to be involved in the development of a pro-inflammatory state. PCNA is also expressed on the surface of certain cancer cells and can play a role in preventing immune cells from killing tumours, as well as being associated with cancer virulence. Given the growing interest in oncolytic viruses (OVs) as a novel cancer therapeutic, this review considers the role of PCNA in healthy, cancerous, and immune cells to gain an understanding of how PCNA targeted therapy and oncolytic virotherapy may interact in the future.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Antígeno Nuclear de Célula en Proliferación , Humanos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Viroterapia Oncolítica/métodos , Neoplasias/terapia , Virus Oncolíticos/fisiología , Animales , Replicación del ADN
10.
Eur J Pharmacol ; 981: 176913, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39154830

RESUMEN

Tumor immunotherapy, especially immune checkpoint inhibitors (ICIs), has been applied in clinical practice, but low response to immune therapies remains a thorny issue. Oncolytic viruses (OVs) are considered promising for cancer treatment because they can selectively target and destroy tumor cells followed by spreading to nearby tumor tissues for a new round of infection. Immunogenic cell death (ICD), which is the major mechanism of OVs' anticancer effects, is induced by endoplasmic reticulum stress and reactive oxygen species overload after virus infection. Subsequent release of specific damage-associated molecular patterns (DAMPs) from different types of tumor cells can transform the tumor microenvironment from "cold" to "hot". In this paper, we broadly define ICD as those types of cell death that is immunogenic, and describe their signaling pathways respectively. Focusing on ICD, we also elucidate the advantages and disadvantages of recent combination therapies and their future prospects.


Asunto(s)
Muerte Celular Inmunogénica , Inmunoterapia , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Viroterapia Oncolítica/métodos , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Neoplasias/terapia , Neoplasias/inmunología , Inmunoterapia/métodos , Animales , Virus Oncolíticos/fisiología , Virus Oncolíticos/inmunología , Microambiente Tumoral/inmunología , Estrés del Retículo Endoplásmico/inmunología , Transducción de Señal
11.
Math Biosci ; 376: 109275, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39127095

RESUMEN

We model interactions between cancer cells and viruses during oncolytic viral therapy. One of our primary goals is to identify parameter regions that yield treatment failure or success. We show that the tumor size under therapy at a particular time is less than the size without therapy. Our analysis demonstrates two thresholds for the horizontal transmission rate: a "failure threshold" below which treatment fails, and a "success threshold" above which infection prevalence reaches 100% and the tumor shrinks to its smallest size. Moreover, we explain how changes in the virulence of the virus alter the success threshold and the minimum tumor size. Our study suggests that the optimal virulence of an oncolytic virus depends on the timescale of virus dynamics. We identify a threshold for the virulence of the virus and show how this threshold depends on the timescale of virus dynamics. Our results suggest that when the timescale of virus dynamics is fast, administering a more virulent virus leads to a greater reduction in the tumor size. Conversely, when the viral timescale is slow, higher virulence can induce oscillations with high amplitude in the tumor size. Furthermore, we introduce the concept of a "Hopf bifurcation Island" in the parameter space, an idea that has applications far beyond the results of this paper and is applicable to many mathematical models. We elucidate what a Hopf bifurcation Island is, and we prove that small Islands can imply very slowly growing oscillatory solutions.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Viroterapia Oncolítica/métodos , Humanos , Neoplasias/terapia , Neoplasias/virología , Virus Oncolíticos/fisiología , Modelos Biológicos , Virulencia , Conceptos Matemáticos
12.
Int J Mol Sci ; 25(15)2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39125874

RESUMEN

Oncolytic viruses and morbilliviruses in particular, represent an interesting therapeutic approach for tumors with a poor prognosis and frequent resistance to conventional therapies. Canine histiocytic sarcomas (HS) exemplify such a neoplasm in need for new curative approaches. Previous investigations demonstrated a limited success of an acute intratumoral application of canine distemper virus (CDV) on xenotransplanted canine histiocytic sarcoma cells (DH82 cells), while persistently CDV-infected DH82 cell transplants exhibited a complete spontaneous regression. Therefore, the present study focuses on an intratumoral application of persistently CDV vaccine strain Onderstepoort-infected DH82 (DH82 Ond p.i.) cells into non-infected subcutaneous DH82 cell transplants in a murine model. DH82 cell transplants that received 10 applications, two days apart, showed a transient growth retardation as well as larger areas of intratumoral necrosis, lower mitotic rates, and a decreased intratumoral vascularization compared to controls. Viral mRNA was detected in all neoplasms following application of DH82 Ond p.i. cells until 66 days after the last injection. Furthermore, infectious virus was present until 62 days after the last injection. Although complete regression was not achieved, the present application regimen provides promising results as a basis for further treatments, particularly with genetically modified viruses, to enhance the observed effects.


Asunto(s)
Virus del Moquillo Canino , Sarcoma Histiocítico , Viroterapia Oncolítica , Animales , Virus del Moquillo Canino/patogenicidad , Virus del Moquillo Canino/genética , Perros , Sarcoma Histiocítico/virología , Ratones , Viroterapia Oncolítica/métodos , Línea Celular Tumoral , Moquillo/virología , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología
13.
Viruses ; 16(7)2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39066301

RESUMEN

Primary bone malignancies, including osteosarcoma (OS), are rare but aggressive. Current OS treatment, involving surgical resection and chemotherapy, has improved survival for non-metastatic cases but remains ineffective for recurrent or metastatic OS. Oncolytic viral therapy (OVT) is a promising alternative, using naturally occurring or genetically modified viruses to selectively target and lyse cancer cells and induce a robust immune response against remaining OS cells. Various oncolytic viruses (OVs), such as adenovirus, herpes simplex virus, and measles virus, have demonstrated efficacy in preclinical OS models. Combining OVT with other therapeutics, such as chemotherapy or immunotherapy, may further improve outcomes. Despite these advances, challenges in reliability of preclinical models, safety, delivery, and immune response must be addressed to optimize OVT for clinical use. Future research should focus on refining delivery methods, exploring combination treatments, and clinical trials to ensure OVT's efficacy and safety for OS. Overall, OVT represents a novel approach with the potential to drastically improve survival outcomes for patients with OS.


Asunto(s)
Neoplasias Óseas , Viroterapia Oncolítica , Virus Oncolíticos , Osteosarcoma , Osteosarcoma/terapia , Viroterapia Oncolítica/métodos , Humanos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Neoplasias Óseas/terapia , Animales , Terapia Combinada
14.
Viruses ; 16(7)2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39066315

RESUMEN

To explore whether the p17 protein of oncolytic avian reovirus (ARV) mediates cell migration and invadopodia formation, we applied several molecular biological approaches for studying the involved cellular factors and signal pathways. We found that ARV p17 activates the p53/phosphatase and tensin homolog (PTEN) pathway to suppress the focal adhesion kinase (FAK)/Src signaling and downstream signal molecules, thus inhibiting cell migration and the formation of invadopodia in murine melanoma cancer cell line (B16-F10). Importantly, p17-induced formation of invadopodia could be reversed in cells transfected with the mutant PTENC124A. p17 protein was found to significantly reduce the expression levels of tyrosine kinase substrate 5 (TKs5), Rab40b, non-catalytic region of tyrosine kinase adaptor protein 1 (NCK1), and matrix metalloproteinases (MMP9), suggesting that TKs5 and Rab40b were transcriptionally downregulated by p17. Furthermore, we found that p17 suppresses the formation of the TKs5/NCK1 complex. Coexpression of TKs5 and Rab40b in B16-F10 cancer cells reversed p17-modulated suppression of the formation of invadopodia. This work provides new insights into p17-modulated suppression of invadopodia formation by activating the p53/PTEN pathway, suppressing the FAK/Src pathway, and inhibiting the formation of the TKs5/NCK1 complex.


Asunto(s)
Movimiento Celular , Quinasa 1 de Adhesión Focal , Orthoreovirus Aviar , Podosomas , Transducción de Señal , Animales , Ratones , Orthoreovirus Aviar/fisiología , Orthoreovirus Aviar/genética , Línea Celular Tumoral , Podosomas/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Virus Oncolíticos/fisiología , Virus Oncolíticos/genética , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Melanoma Experimental/terapia , Melanoma Experimental/patología , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética
15.
ACS Nano ; 18(28): 18282-18298, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38953884

RESUMEN

The therapeutic efficacy of oncolytic adenoviruses (OAs) relies on efficient viral transduction and replication. However, the limited expression of coxsackie-adenovirus receptors in many tumors, along with the intracellular antiviral signaling, poses significant obstacles to OA infection and oncolysis. Here, we present sonosensitizer-armed OAs (saOAs) that potentiate the antitumor efficacy of oncolytic virotherapy through sonodynamic therapy-augmented virus replication. The saOAs could not only efficiently infect tumor cells via transferrin receptor-mediated endocytosis but also exhibit enhanced viral replication and tumor oncolysis under ultrasound irradiation. We revealed that the sonosensitizer loaded on the viruses induced the generation of ROS within tumor cells, which triggered JNK-mediated autophagy, ultimately leading to the enhanced viral replication. In mouse models of malignant melanoma, the combination of saOAs and sonodynamic therapy elicited a robust antitumor immune response, resulting in significant inhibition of melanoma growth and improved host survival. This work highlights the potential of sonodynamic therapy in enhancing the effectiveness of OAs and provides a promising platform for fully exploiting the antitumor efficacy of oncolytic virotherapy.


Asunto(s)
Adenoviridae , Viroterapia Oncolítica , Virus Oncolíticos , Replicación Viral , Animales , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Adenoviridae/fisiología , Virus Oncolíticos/fisiología , Virus Oncolíticos/genética , Replicación Viral/efectos de la radiación , Ratones , Humanos , Línea Celular Tumoral , Terapia por Ultrasonido/métodos , Melanoma/terapia , Melanoma/patología
16.
Virology ; 598: 110171, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39018682

RESUMEN

In addition to chemotherapy, oncolytic viruses are an efficient treatment for acute myeloid leukemia (AML). Like other oncolytic viruses, the anti-tumor efficacy of reovirus when administered intravenously is reduced due to the presence of neutralizing antibodies. In this study, we evaluated the role of exosomes in human umbilical cord-derived mesenchymal stem cells (UC-MSCs) to deliver reovirus to AML cells. We show that UC-MSCs loaded with reovirus can deliver reovirus to tumor cells without cellular contact. We further demonstrate that the exosome inhibitor, GW4869, inhibits the release of exosomes as well as inhibited the transfer of reovirus from UC-MSCs to tumor cells. Mechanistically, we show that exosomes derived from reovirus-infected UC-MSCs (MSCREO-EXOs) have a tumor lysis effect and transmit reovirus to tumor cells mainly through clathrin-mediated endocytosis (CME) and macropinocytosis. In addition, we demonstrate the feasibility of using MSC-derived exosomes (MSC-EXOs) as a reovirus carrier to exert an anti-tumor effect on AML cells. Collectively, our data indicate that UC-MSCs transfer reovirus to AML cells via exosome release and prompt further study of MSC-EXOs as a potential reovirus carrier to treat AML.


Asunto(s)
Exosomas , Leucemia Mieloide Aguda , Células Madre Mesenquimatosas , Viroterapia Oncolítica , Virus Oncolíticos , Cordón Umbilical , Humanos , Exosomas/metabolismo , Células Madre Mesenquimatosas/virología , Células Madre Mesenquimatosas/metabolismo , Leucemia Mieloide Aguda/terapia , Cordón Umbilical/citología , Virus Oncolíticos/fisiología , Viroterapia Oncolítica/métodos , Línea Celular Tumoral , Reoviridae/fisiología , Compuestos de Anilina/farmacología , Endocitosis , Compuestos de Bencilideno
17.
Int J Mol Sci ; 25(13)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39000311

RESUMEN

Hepatocellular carcinoma is a refractory tumor with poor prognosis and high mortality. Many oncolytic viruses are currently being investigated for the treatment of hepatocellular carcinoma. Based on previous studies, we constructed a recombinant GM-CSF-carrying Sindbis virus, named SINV-GM-CSF, which contains a mutation (G to S) at amino acid 285 in the nsp1 protein of the viral vector. The potential of this mutated vector for liver cancer therapy was verified at the cellular level and in vivo, respectively, and the changes in the tumor microenvironment after treatment were also described. The results showed that the Sindbis virus could effectively infect hepatocellular carcinoma cell lines and induce cell death. Furthermore, the addition of GM-CSF enhanced the tumor-killing effect of the Sindbis virus and increased the number of immune cells in the intra-tumor microenvironment during the treatment. In particular, SINV-GM-CSF was able to efficiently kill tumors in a mouse tumor model of hepatocellular carcinoma by regulating the elevation of M1-type macrophages (which have a tumor-resistant ability) and the decrease in M2-type macrophages (which have a tumor-promoting capacity). Overall, SINV-GM-CSF is an attractive vector platform with clinical potential for use as a safe and effective oncolytic virus.


Asunto(s)
Carcinoma Hepatocelular , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Neoplasias Hepáticas , Viroterapia Oncolítica , Virus Oncolíticos , Virus Sindbis , Microambiente Tumoral , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Carcinoma Hepatocelular/terapia , Animales , Virus Sindbis/genética , Virus Sindbis/fisiología , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virología , Neoplasias Hepáticas/genética , Ratones , Viroterapia Oncolítica/métodos , Humanos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Macrófagos/metabolismo , Macrófagos/inmunología
18.
Bull Math Biol ; 86(8): 93, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38896363

RESUMEN

Virotherapy treatment is a new and promising target therapy that selectively attacks cancer cells without harming normal cells. Mathematical models of oncolytic viruses have shown predator-prey like oscillatory patterns as result of an underlying Hopf bifurcation. In a spatial context, these oscillations can lead to different spatio-temporal phenomena such as hollow-ring patterns, target patterns, and dispersed patterns. In this paper we continue the systematic analysis of these spatial oscillations and discuss their relevance in the clinical context. We consider a bifurcation analysis of a spatially explicit reaction-diffusion model to find the above mentioned spatio-temporal virus infection patterns. The desired pattern for tumor eradication is the hollow ring pattern and we find exact conditions for its occurrence. Moreover, we derive the minimal speed of travelling invasion waves for the cancer and for the oncolytic virus. Our numerical simulations in 2-D reveal complex spatial interactions of the virus infection and a new phenomenon of a periodic peak splitting. An effect that we cannot explain with our current methods.


Asunto(s)
Simulación por Computador , Conceptos Matemáticos , Modelos Biológicos , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Humanos , Neoplasias/terapia , Neoplasias/virología
19.
Int Immunopharmacol ; 137: 112393, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38852522

RESUMEN

Recombinant adenovirus serotype 5 (Ad5)-mediated virotherapy is a maturing technique in cancer treatment. However, the utility of adenovirus (Ad) has been limited by low expression of coxsackievirus and adenovirus receptor (CAR) in cancer cells resulting in poor infectivity of Ads. To overcome the problem, we aimed to develop a novel tropism-modified oncolytic adenovirus, ZD55-F-HI-sPD-1-EGFP, which contains the epitope of PD-1 (70-77aa) at the HI-loop of Ad fiber. Trimerization of Fiber-sPD-1 was confirmed by immunoblot analysis. ZD55-F-HI-sPD-1-EGFP shows a remarkable improvement in viral infection rate and gene transduction efficiency in the PD-L1-positive cancer cells. Competition assays with a PD-L1 protein reveals that cell internalization of ZD55-F-HI-sPD-1-EGFP is mediated by both CAR and PD-L1 at a high dose. The progeny virus production capacity showed that sPD-1 incorporated fiber-modified oncolytic Ad replication was not affected. Furthermore, treating with ZD55-F-HI-sPD-1-EGFP significantly increased viral infection rate and enhanced anti-tumor effect in vivo. This study demonstrates that the strategy to expand tropism of oncolytic Ad may significantly improve therapeutic profile for cancer treatment.


Asunto(s)
Adenoviridae , Antígeno B7-H1 , Viroterapia Oncolítica , Virus Oncolíticos , Tropismo Viral , Humanos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Animales , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Adenoviridae/genética , Adenoviridae/fisiología , Línea Celular Tumoral , Ratones , Neoplasias/terapia , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , Femenino , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Células HEK293
20.
Viruses ; 16(6)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38932212

RESUMEN

Oncolytic virotherapy, using viruses such as vesicular stomatitis virus (VSVΔ51) and Herpes Simplex Virus-1 (HSV-1) to selectively attack cancer cells, faces challenges such as cellular resistance mediated by the interferon (IFN) response. Dimethyl fumarate (DMF) is used in the treatment of multiple sclerosis and psoriasis and is recognized for its anti-cancer properties and has been shown to enhance both VSVΔ51 and HSV-1 oncolytic activity. Tepilamide fumarate (TPF) is a DMF analog currently undergoing clinical trials for the treatment of moderate-to-severe plaque psoriasis. The aim of this study was to evaluate the potential of TPF in enhancing the effectiveness of oncolytic viruses. In vitro, TPF treatment rendered 786-0 carcinoma cells more susceptible to VSVΔ51 infection, leading to increased viral replication. It outperformed DMF in both increasing viral infection and increasing the killing of these resistant cancer cells and other cancer cell lines tested. Ex vivo studies demonstrated TPF's selective boosting of oncolytic virus infection in cancer cells without affecting healthy tissues. Effectiveness was notably high in pancreatic and ovarian tumor samples. Our study further indicates that TPF can downregulate the IFN pathway through a similar mechanism to DMF, making resistant cancer cells more vulnerable to viral infection. Furthermore, TPF's impact on gene therapy was assessed, revealing its ability to enhance the transduction efficiency of vectors such as lentivirus, adenovirus type 5, and adeno-associated virus type 2 across various cell lines. This data underscore TPF's potential role in not only oncolytic virotherapy but also in the broader application of gene therapy. Collectively, these findings position TPF as a promising agent in oncolytic virotherapy, warranting further exploration of its therapeutic potential.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Replicación Viral , Humanos , Viroterapia Oncolítica/métodos , Línea Celular Tumoral , Virus Oncolíticos/fisiología , Replicación Viral/efectos de los fármacos , Fumaratos/farmacología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Dimetilfumarato/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA