Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.629
Filtrer
1.
Cancer Immunol Immunother ; 73(9): 173, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38953982

RÉSUMÉ

Recent studies have indicated that combining oncolytic viruses with CAR-T cells in therapy has shown superior anti-tumor effects, representing a promising approach. Nonetheless, the localized delivery method of intratumoral injection poses challenges for treating metastatic tumors or distal tumors that are difficult to reach. To address this obstacle, we employed HSV-1-infected CAR-T cells, which systemically delivery HSV into solid tumors. The biological function of CAR-T cells remained intact after loading them with HSV for a period of three days. In both immunocompromised and immunocompetent GBM orthotopic mouse models, B7-H3 CAR-T cells effectively delivered HSV to tumor lesions, resulting in enhanced T-cell infiltration and significantly prolonged survival in mice. We also employed a bilateral subcutaneous tumor model and observed that the group receiving intratumoral virus injection exhibited a significant reduction in tumor volume on the injected side, while the group receiving intravenous infusion of CAR-T cells carrying HSV displayed suppressed tumor growth on both sides. Hence, CAR-THSV cells offer notable advantages in the systemic delivery of HSV to distant tumors. In conclusion, our findings emphasize the potential of CAR-T cells as carriers for HSV, presenting significant advantages for oncolytic virotherapy targeting distant tumors.


Sujet(s)
Immunothérapie adoptive , Thérapie virale de cancers , Virus oncolytiques , Récepteurs chimériques pour l'antigène , Animaux , Souris , Thérapie virale de cancers/méthodes , Humains , Virus oncolytiques/immunologie , Virus oncolytiques/génétique , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Herpèsvirus humain de type 1/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Lignée cellulaire tumorale , Lymphocytes T/immunologie , Femelle , Glioblastome/thérapie , Glioblastome/immunologie
2.
Int Immunopharmacol ; 136: 112305, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38823178

RÉSUMÉ

The second-leading cause of death, cancer, poses a significant threat to human life. Innovations in cancer therapies are crucial due to limitations in traditional approaches. Newcastle disease virus (NDV), a nonpathogenic oncolytic virus, exhibits multifunctional anticancer properties by selectively infecting, replicating, and eliminating tumor cells. To enhance NDV's antitumor activity, four oncolytic NDV viruses were developed, incorporating IL24 and/or GM-CSF genes at different gene loci using reverse genetics. In vitro experiments revealed that oncolytic NDV virus augmented the antitumor efficacy of the parental virus rClone30, inhibiting tumor cell proliferation, inducing tumor cell fusion, and promoting apoptosis. Moreover, NDV carrying the IL24 gene inhibited microvessel formation in CAM experiments. Evaluation in a mouse model of liver cancer confirmed the therapeutic efficacy of oncolytic NDV viral therapy. Tumors in mice treated with oncolytic NDV virus significantly decreased in size, accompanied by tumor cell detachment and apoptosis evident in pathological sections. Furthermore, oncolytic NDV virus enhanced T cell and dendritic cell production and substantially improved the survival rate of mice with hepatocellular carcinoma, with rClone30-IL24(P/M) demonstrating significant therapeutic effects. This study establishes a basis for utilizing oncolytic NDV virus as an antitumor agent in clinical practice.


Sujet(s)
Interleukines , Virus de la maladie de Newcastle , Thérapie virale de cancers , Virus oncolytiques , Animaux , Virus de la maladie de Newcastle/génétique , Virus de la maladie de Newcastle/physiologie , Thérapie virale de cancers/méthodes , Virus oncolytiques/génétique , Virus oncolytiques/physiologie , Humains , Souris , Lignée cellulaire tumorale , Interleukines/génétique , Interleukines/métabolisme , Tumeurs du foie/thérapie , Souris de lignée BALB C , Carcinome hépatocellulaire/thérapie , Apoptose , Néovascularisation pathologique/thérapie , Prolifération cellulaire , Facteur de stimulation des colonies de granulocytes et de macrophages/génétique , Facteur de stimulation des colonies de granulocytes et de macrophages/métabolisme , Cellules dendritiques/immunologie , Lymphocytes T/immunologie
3.
Front Immunol ; 15: 1375413, 2024.
Article de Anglais | MEDLINE | ID: mdl-38895115

RÉSUMÉ

Introduction: Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas with unacceptably low cure rates occurring often in patients with neurofibromatosis 1 defects. To investigate oncolytic Herpes Simplex Virus (oHSV) as an immunotherapeutic approach, we compared viral replication, functional activity, and immune response between unarmed and interleukin 12 (IL-12)-armed oncolytic viruses in virus-permissive (B109) and -resistant (67C-4) murine MPNSTs. Methods: This study compared two attenuated IL-12-oHSVs with γ134.5 gene deletions (Δγ134.5) and the same transgene expression cassette. The primary difference in the IL-12-oHSVs was in their ability to counter the translational arrest response in infected cells. Unlike M002 (Δγ134.5, mIL-12), C002 (Δγ134.5, mIL-12, IRS1) expresses an HCMV IRS1 gene and evades dsRNA activated translational arrest in infected cells. Results and discussion: Our results show that oHSV replication and gene expression results in vitro were not predictive of oHSV direct oncolytic activity in vivo. Tumors that supported viral replication in cell culture studies resisted viral replication by both oHSVs and restricted M002 transgene expression in vivo. Furthermore, two IL-12-oHSVs with equivalent transcriptional activity differed in IL-12 protein production in vivo, and the differences in IL-12 protein levels were reflected in immune infiltrate activity changes as well as tumor growth suppression differences between the IL-12-oHSVs. C002-treated tumors exhibited sustained IL-12 production with improved dendritic cells, monocyte-macrophage activity (MHCII, CD80/CD86 upregulation) and a polyfunctional Th1-cell response in the tumor infiltrates. Conclusion: These results suggest that transgene protein production differences between oHSVs in vivo, in addition to replication differences, can impact OV-therapeutic activity.


Sujet(s)
Interleukine-12 , Thérapie virale de cancers , Virus oncolytiques , Transgènes , Réplication virale , Animaux , Interleukine-12/génétique , Interleukine-12/métabolisme , Souris , Thérapie virale de cancers/méthodes , Virus oncolytiques/génétique , Virus oncolytiques/immunologie , Lignée cellulaire tumorale , Immunothérapie/méthodes , Humains , Simplexvirus/génétique , Cellules dendritiques/immunologie , Femelle
4.
Nat Commun ; 15(1): 5442, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937436

RÉSUMÉ

Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.


Sujet(s)
Antigènes néoplasiques , Antigène CD274 , Lymphocytes T CD8+ , Carcinome hépatocellulaire , Tumeurs du foie , Thérapie virale de cancers , Virus oncolytiques , Microenvironnement tumoral , Virus oncolytiques/génétique , Virus oncolytiques/immunologie , Animaux , Thérapie virale de cancers/méthodes , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/immunologie , Microenvironnement tumoral/immunologie , Souris , Antigène CD274/métabolisme , Antigène CD274/génétique , Antigène CD274/immunologie , Humains , Tumeurs du foie/thérapie , Tumeurs du foie/immunologie , Antigènes néoplasiques/immunologie , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Interféron bêta/métabolisme , Interféron bêta/immunologie , Souris de lignée C57BL , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Lymphocytes T/immunologie , Femelle , Vesiculovirus/immunologie , Vesiculovirus/génétique
5.
Front Immunol ; 15: 1355566, 2024.
Article de Anglais | MEDLINE | ID: mdl-38835775

RÉSUMÉ

Dendritic cell (DC)-based vaccines have emerged as a promising strategy in cancer immunotherapy due to low toxicity. However, the therapeutic efficacy of DC as a monotherapy is insufficient due to highly immunosuppressive tumor environment. To address these limitations of DC as immunotherapeutic agent, we have developed a polymeric nanocomplex incorporating (1) oncolytic adenovirus (oAd) co-expressing interleukin (IL)-12 and granulocyte-macrophage colony-stimulating factor (GM-CSF) and (2) arginine-grafted bioreducible polymer with PEGylated paclitaxel (APP) to restore antitumor immune surveillance function in tumor milieu and potentiate immunostimulatory attributes of DC vaccine. Nanohybrid complex (oAd/APP) in combination with DC (oAd/APP+DC) induced superior expression level of antitumor cytokines (IL-12, GM-CSF, and interferon gamma) than either oAd/APP or DC monotherapy in tumor tissues, thus resulting in superior intratumoral infiltration of both endogenous and exogenous DCs. Furthermore, oAd/APP+DC treatment led superior migration of DC to secondary lymphoid organs, such as draining lymph nodes and spleen, in comparison with either monotherapy. Superior migration profile of DCs in oAd/APP+DC treatment group resulted in more prolific activation of tumor-specific T cells in these lymphoid organs and greater intratumoral infiltration of T cells. Additionally, oAd/APP+DC treatment led to lower subset of tumor infiltrating lymphocytes and splenocytes being immunosuppressive regulatory T cells than any other treatment groups. Collectively, oAd/APP+DC led to superior induction of antitumor immune response and amelioration of immunosuppressive tumor microenvironment to elicit potent tumor growth inhibition than either monotherapy.


Sujet(s)
Adenoviridae , Cellules dendritiques , Thérapie virale de cancers , Virus oncolytiques , Paclitaxel , Cellules dendritiques/immunologie , Animaux , Paclitaxel/pharmacologie , Adenoviridae/génétique , Souris , Virus oncolytiques/immunologie , Virus oncolytiques/génétique , Thérapie virale de cancers/méthodes , Association thérapeutique , Lignée cellulaire tumorale , Humains , Souris de lignée C57BL , Vaccins anticancéreux/immunologie , Immunothérapie/méthodes , Facteur de stimulation des colonies de granulocytes et de macrophages/métabolisme , Femelle , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques
6.
Sci Rep ; 14(1): 13108, 2024 06 07.
Article de Anglais | MEDLINE | ID: mdl-38849383

RÉSUMÉ

LHPP has been shown to be a new tumor suppressor, and has a tendency to be under-expressed in a variety of cancers. Oncolytic virotheray is a promising therapeutics for lung cancer in recent decade years. Here we successfully constructed a new recombinant oncolytic adenovirus GD55-LHPP and investigated the effect of GD55-LHPP on the growth of lung cancer cells in vitro and in vivo. The results showed that LHPP had lower expression in either lung cancer cells or clinical lung cancer tissues compared with normal cells or tissues, and GD55-LHPP effectively mediated LHPP expression in lung cancer cells. GD55-LHPP could effectively inhibit the proliferation of lung cancer cell lines and rarely affected normal cell growth. Mechanically, the oncolytic adenovirus GD55-LHPP was able to induce stronger apoptosis of lung cancer cells compared with GD55 through the activation of caspase signal pathway. Notably, GD55-LHPP also activated autophagy-related signal pathway. Further, GD55-LHPP efficiently inhibited tumor growth in lung cancer xenograft in mice and prolonged animal survival rate compared with the control GD55 or PBS. In conclusion, the novel construct GD55-LHPP provides a valuable strategy for lung cancer-targeted therapy and develop the role of tumor suppress gene LHPP in lung cancer gene therapy.


Sujet(s)
Adenoviridae , Apoptose , Tumeurs du poumon , Thérapie virale de cancers , Virus oncolytiques , Tests d'activité antitumorale sur modèle de xénogreffe , Tumeurs du poumon/thérapie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Humains , Animaux , Thérapie virale de cancers/méthodes , Adenoviridae/génétique , Virus oncolytiques/génétique , Souris , Lignée cellulaire tumorale , Prolifération cellulaire , Souris nude , Femelle , Autophagie
7.
Cell Biochem Funct ; 42(4): e4055, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38856033

RÉSUMÉ

The heterogeneity of the solid tumor microenvironment (TME) impairs the therapeutic efficacy of standard therapies and also reduces the infiltration of antitumor immune cells, all of which lead to tumor progression and invasion. In addition, self-renewing cancer stem cells (CSCs) support tumor dormancy, drug resistance, and recurrence, all of which might pose challenges to the eradication of malignant tumor masses with current therapies. Natural forms of oncolytic viruses (OVs) or engineered OVs are known for their potential to directly target and kill tumor cells or indirectly eradicate tumor cells by involving antitumor immune responses, including enhancement of infiltrating antitumor immune cells, induction of immunogenic cell death, and reprogramming of cold TME to an immune-sensitive hot state. More importantly, OVs can target stemness factors that promote tumor progression, which subsequently enhances the efficacy of immunotherapies targeting solid tumors, particularly the CSC subpopulation. Herein, we describe the role of CSCs in tumor heterogeneity and resistance and then highlight the potential and remaining challenges of immunotherapies targeting CSCs. We then review the potential of OVs to improve tumor immunogenicity and target CSCs and finally summarize the challenges within the therapeutic application of OVs in preclinical and clinical trials.


Sujet(s)
Immunothérapie , Tumeurs , Cellules souches tumorales , Thérapie virale de cancers , Virus oncolytiques , Humains , Cellules souches tumorales/immunologie , Virus oncolytiques/génétique , Virus oncolytiques/immunologie , Tumeurs/thérapie , Tumeurs/immunologie , Microenvironnement tumoral/immunologie , Animaux
8.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38928259

RÉSUMÉ

Oncolytic adenoviruses are in development as immunotherapeutic agents for solid tumors. Their efficacy is in part dependent on their ability to replicate in tumors. It is, however, difficult to obtain evidence for intratumoral oncolytic adenovirus replication if direct access to the tumor is not possible. Detection of systemic adenovirus DNA, which is sometimes used as a proxy, has limited value because it does not distinguish between the product of intratumoral replication and injected virus that did not replicate. Therefore, we investigated if detection of virus-associated RNA (VA RNA) by RT-qPCR on liquid biopsies could be used as an alternative. We found that VA RNA is expressed in adenovirus-infected cells in a replication-dependent manner and is secreted by these cells in association with extracellular vesicles. This allowed VA RNA detection in the peripheral blood of a preclinical in vivo model carrying adenovirus-injected human tumors and on liquid biopsies from a human clinical trial. Our results confirm that VA RNA detection in liquid biopsies can be used for minimally invasive assessment of oncolytic adenovirus replication in solid tumors in vivo.


Sujet(s)
Adenoviridae , Thérapie virale de cancers , Virus oncolytiques , ARN viral , Réplication virale , Humains , Virus oncolytiques/génétique , Virus oncolytiques/physiologie , ARN viral/génétique , Adenoviridae/génétique , Adenoviridae/physiologie , Animaux , Thérapie virale de cancers/méthodes , Souris , Lignée cellulaire tumorale , Tumeurs/thérapie , Tumeurs/génétique , Femelle
9.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891807

RÉSUMÉ

FOLFOXIRI chemotherapy is a first-line therapy for advanced or metastatic colorectal cancer (CRC), yet its therapeutic efficacy remains limited. Immunostimulatory therapies like oncolytic viruses can complement chemotherapies by fostering the infiltration of the tumor by immune cells and enhancing drug cytotoxicity. In this study, we explored the effect of combining the FOLFOXIRI chemotherapeutic agents with the oncolytic coxsackievirus B3 (CVB3) PD-H in the CRC cell line Colo320. Additionally, we examined the impact of the drugs on the expression of microRNAs (miRs), which could be used to increase the safety of oncolytic CVB3 containing corresponding miR target sites (miR-TS). The measurement of cytotoxic activity using the Chou-Talalay combination index approach revealed that PD-H synergistically enhanced the cytotoxic activity of oxaliplatin (OX), 5-fluorouracil (5-FU) and SN-38. PD-H replication was not affected by OX and SN-38 but inhibited by high concentrations of 5-FU. MiR expression levels were not or only slightly elevated by the drugs or with drug/PD-H combinations on Colo320 cells. Moreover, the drug treatment did not increase the mutation rate of the miR-TS inserted into the PD-H genome. The results demonstrate that the combination of FOLFOXIRI drugs and PD-H may be a promising approach to enhance the therapeutic effect of FOLFOXIRI therapy in CRC.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs colorectales , Fluorouracil , Leucovorine , microARN , Thérapie virale de cancers , Virus oncolytiques , Humains , Tumeurs colorectales/thérapie , Tumeurs colorectales/traitement médicamenteux , Lignée cellulaire tumorale , Fluorouracil/pharmacologie , Thérapie virale de cancers/méthodes , microARN/génétique , Virus oncolytiques/génétique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Leucovorine/pharmacologie , Leucovorine/usage thérapeutique , Composés organiques du platine/pharmacologie , Oxaliplatine/pharmacologie , Entérovirus humain B/effets des médicaments et des substances chimiques , Association thérapeutique , Irinotécan/pharmacologie
10.
Viruses ; 16(6)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38932177

RÉSUMÉ

Newcastle disease virus (NDV) is an avian pathogen with an unsegmented negative-strand RNA genome that belongs to the Paramyxoviridae family. While primarily pathogenic in birds, NDV presents no threat to human health, rendering it a safe candidate for various biomedical applications. Extensive research has highlighted the potential of NDV as a vector for vaccine development and gene therapy, owing to its transcriptional modularity, low recombination rate, and lack of a DNA phase during replication. Furthermore, NDV exhibits oncolytic capabilities, efficiently eliciting antitumor immune responses, thereby positioning it as a promising therapeutic agent for cancer treatment. This article comprehensively reviews the biological characteristics of NDV, elucidates the molecular mechanisms underlying its oncolytic properties, and discusses its applications in the fields of vaccine vector development and tumor therapy.


Sujet(s)
Vecteurs génétiques , Tumeurs , Virus de la maladie de Newcastle , Thérapie virale de cancers , Virus oncolytiques , Virus de la maladie de Newcastle/génétique , Virus de la maladie de Newcastle/immunologie , Animaux , Humains , Vecteurs génétiques/génétique , Tumeurs/thérapie , Tumeurs/immunologie , Thérapie virale de cancers/méthodes , Virus oncolytiques/génétique , Virus oncolytiques/immunologie , Thérapie génétique/méthodes , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Maladie de Newcastle/prévention et contrôle , Maladie de Newcastle/thérapie , Maladie de Newcastle/virologie , Maladie de Newcastle/immunologie , Développement de vaccin/méthodes
11.
Int J Cancer ; 155(5): E6, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38773742

RÉSUMÉ

EXPRESSION OF CONCERN: Nusrat Jahan, Jae M. Lee, Khalid Shah, Hiroaki Wakimoto, "Therapeutic targeting of chemoresistant and recurrent glioblastoma stem cells with a proapoptotic variant of oncolytic herpes simplex virus", International Journal of Cancer 141, no. 8 (2017): 1671-1681. https://doi.org/10.1002/ijc.30811. This Expression of Concern is for the above article, published online on 31 May 2017, in Wiley Online Library (wileyonlinelibrary.com), and has been published by agreement between the authors, the journal's Editor-in-Chief, Christoph Plass, the Union for International Cancer Control and John Wiley & Sons, Ltd. The Expression of Concern has been agreed following concerns raised regarding figure 4g and 5d. The Control (PBS) 4X DAPI image in figure 5d was found to contain elements similar to the Control (PBS) DAPI image presented in figure 4g. The authors admitted that they had mistakenly supplied the incorrect image to represent DAPI in figure 5d. Since the original DAPI image for figure 5d is no longer available, this issue could not be resolved. The journal has decided to issue an Expression of Concern to alert the readers.


Sujet(s)
Apoptose , Résistance aux médicaments antinéoplasiques , Glioblastome , Cellules souches tumorales , Thérapie virale de cancers , Virus oncolytiques , Simplexvirus , Glioblastome/thérapie , Glioblastome/anatomopathologie , Humains , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Thérapie virale de cancers/méthodes , Simplexvirus/génétique , Virus oncolytiques/génétique , Récidive tumorale locale , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/anatomopathologie
12.
J Immunother Cancer ; 12(5)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38821716

RÉSUMÉ

Cytokines are small proteins that regulate the growth and functional activity of immune cells, and several have been approved for cancer therapy. Oncolytic viruses are agents that mediate antitumor activity by directly killing tumor cells and inducing immune responses. Talimogene laherparepvec is an oncolytic herpes simplex virus type 1 (oHSV), approved for the treatment of recurrent melanoma, and the virus encodes the human cytokine, granulocyte-macrophage colony-stimulating factor (GM-CSF). A significant advantage of oncolytic viruses is the ability to deliver therapeutic payloads to the tumor site that can help drive antitumor immunity. While cytokines are especially interesting as payloads, the optimal cytokine(s) used in oncolytic viruses remains controversial. In this review, we highlight preliminary data with several cytokines and chemokines, including GM-CSF, interleukin 12, FMS-like tyrosine kinase 3 ligand, tumor necrosis factor α, interleukin 2, interleukin 15, interleukin 18, chemokine (C-C motif) ligand 2, chemokine (C-C motif) ligand 5, chemokine (C-X-C motif) ligand 4, or their combinations, and show how these payloads can further enhance the antitumor immunity of oHSV. A better understanding of cytokine delivery by oHSV can help improve clinical benefit from oncolytic virus immunotherapy in patients with cancer.


Sujet(s)
Cytokines , Immunothérapie , Tumeurs , Thérapie virale de cancers , Virus oncolytiques , Humains , Thérapie virale de cancers/méthodes , Virus oncolytiques/immunologie , Virus oncolytiques/génétique , Cytokines/métabolisme , Immunothérapie/méthodes , Tumeurs/thérapie , Tumeurs/immunologie , Animaux , Simplexvirus/immunologie , Simplexvirus/génétique , Herpèsvirus humain de type 1/immunologie
13.
Biochem Biophys Res Commun ; 718: 149931, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38723415

RÉSUMÉ

Oncolytic viruses (OVs) have shown potential in converting a "cold" tumor into a "hot" one and exhibit effectiveness in various cancer types. However, only a subset of patients respond to oncolytic virotherapy. It is important to understand the resistance mechanisms to OV treatment in pancreatic ductal adenocarcinoma (PDAC) to engineer oncolytic viruses. In this study, we used transcriptome RNA sequencing (RNA-seq) to identify Visfatin, which was highly expressed in the responsive tumors following OV treatment. To explore the antitumor efficacy, we modified OV-mVisfatin, which effectively inhibited tumor growth. For the first time, we revealed that Visfatin promoted the antitumor efficacy of OV by remodeling the tumor microenvironment, which involved enhancing CD8+ T cell and DC cell infiltration and activation, repolarizing macrophages towards the M1-like phenotype, and decreasing Treg cells using single-cell RNA sequencing (scRNA-seq) and flow cytometry. Furthermore, PD-1 blockade significantly enhanced OV-mVisfatin antitumor efficacy, offering a promising new therapeutic strategy for PDAC.


Sujet(s)
Herpèsvirus humain de type 1 , Nicotinamide phosphoribosyltransferase , Thérapie virale de cancers , Virus oncolytiques , Tumeurs du pancréas , Microenvironnement tumoral , Animaux , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Souris , Thérapie virale de cancers/méthodes , Nicotinamide phosphoribosyltransferase/génétique , Nicotinamide phosphoribosyltransferase/métabolisme , Herpèsvirus humain de type 1/génétique , Lignée cellulaire tumorale , Virus oncolytiques/génétique , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Souris de lignée C57BL , Humains , Lymphocytes T CD8+/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/génétique , Récepteur-1 de mort cellulaire programmée/métabolisme , Femelle
14.
Virology ; 595: 110093, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38692134

RÉSUMÉ

Oncolytic virotherapy stands out as a burgeoning and promising therapeutic paradigm, harnessing the intrinsic cytotoxicity of oncolytic viruses for selective replication and dissemination within tumors. The primary mode of action revolves around the direct eradication of tumor cells. In our previous investigations, we formulated an oncolytic herpes simplex virus type 2 (OH2) and substantiated its anti-tumor efficacy both in vivo and in vitro. Subsequently, we embarked on a phase I/II clinical trial in China (NMPA, 2018L02743) and the USA (FDA, IND 27137) to assess OH2's safety, biodistribution, and anti-tumor activity as a standalone agent in patients with advanced solid tumors. In this investigation, our primary focus was to comprehend the influence of the major capsid protein VP5 of OH2 on its efficacy as an antitumor agent. Our findings underscore that the VP5 protein significantly amplifies OH2's oncolytic impact on A549 cells. Additionally, we observed that VP5 actively promotes the induction of apoptosis in A549 cells, both in vivo and in vitro. Through comprehensive transcriptional sequencing, we further authenticated that the VP5 protein triggers apoptosis-related signaling pathways and Gene Ontology (GO) terms in A549 cells. Moreover, we scrutinized differentially expressed genes in the p53-dependent apoptosis pathway and conducted meticulous in vitro validation of these genes. Subsequently, we delved deeper into unraveling the functional significance of the TP53I3 gene and conclusively affirmed that the VP5 protein induces apoptosis in A549 cells through the TP53I3 gene. These revelations illuminate the underlying mechanisms of OH2's antitumor activity and underscore the pivotal role played by the VP5 protein. The outcomes of our study harbor promising implications for the formulation of effective oncolytic virotherapy strategies in cancer treatment.


Sujet(s)
Apoptose , Herpèsvirus humain de type 2 , Thérapie virale de cancers , Virus oncolytiques , Humains , Virus oncolytiques/génétique , Virus oncolytiques/physiologie , Cellules A549 , Thérapie virale de cancers/méthodes , Animaux , Herpèsvirus humain de type 2/physiologie , Herpèsvirus humain de type 2/génétique , Protéines de capside/génétique , Protéines de capside/métabolisme , Souris , Tests d'activité antitumorale sur modèle de xénogreffe
15.
Biochim Biophys Acta Rev Cancer ; 1879(4): 189110, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38754793

RÉSUMÉ

Oncolytic viruses (OVs) are increasingly recognized as potent tools in cancer therapy, effectively targeting and eradicating oncogenic conditions while sparing healthy cells. They enhance antitumor immunity by triggering various immune responses throughout the cancer cycle. Genetically engineered OVs swiftly destroy cancerous tissues and activate the immune system by releasing soluble antigens like danger signals and interferons. Their ability to stimulate both innate and adaptive immunity makes them particularly attractive in cancer immunotherapy. Recent advancements involve combining OVs with other immune therapies, yielding promising results. Transgenic OVs, designed to enhance immunostimulation and specifically target cancer cells, further improve immune responses. This review highlights the intrinsic mechanisms of OVs and underscores their synergistic potential with other immunotherapies. It also proposes strategies for optimizing armed OVs to bolster immunity against tumors.


Sujet(s)
Immunothérapie , Tumeurs , Thérapie virale de cancers , Virus oncolytiques , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Virus oncolytiques/immunologie , Virus oncolytiques/génétique , Thérapie virale de cancers/méthodes , Immunothérapie/méthodes , Animaux
16.
Adv Protein Chem Struct Biol ; 140: 419-492, 2024.
Article de Anglais | MEDLINE | ID: mdl-38762277

RÉSUMÉ

Discovering a therapeutic that can counteract the aggressiveness of this disease's mechanism is crucial for improving survival rates for cancer patients and for better understanding the most different types of cancer. In recent years, using these viruses as an anticancer therapy has been thought to be successful. They mostly work by directly destroying cancer cells, activating the immune system to fight cancer, and expressing exogenous effector genes. For the treatment of tumors, oncolytic viruses (OVs), which can be modified to reproduce only in tumor tissues and lyse them while preserving the healthy non-neoplastic host cells and reinstating antitumor immunity which present a novel immunotherapeutic strategy. OVs can exist naturally or be created in a lab by altering existing viruses. These changes heralded the beginning of a new era of less harmful virus-based cancer therapy. We discuss three different types of oncolytic viruses that have already received regulatory approval to treat cancer as well as clinical research using oncolytic adenoviruses. The primary therapeutic applications, mechanism of action of oncolytic virus updates, future views of this therapy will be covered in this chapter.


Sujet(s)
Immunothérapie , Tumeurs , Thérapie virale de cancers , Virus oncolytiques , Humains , Virus oncolytiques/immunologie , Virus oncolytiques/génétique , Tumeurs/thérapie , Tumeurs/immunologie , Immunothérapie/méthodes , Thérapie virale de cancers/méthodes , Animaux
17.
Breast Cancer Res ; 26(1): 78, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750591

RÉSUMÉ

BACKGROUND: Metastatic breast cancer is a leading cause of cancer death in woman. Current treatment options are often associated with adverse side effects and poor outcomes, demonstrating the need for effective new treatments. Immunotherapies can provide durable outcomes in many cancers; however, limited success has been achieved in metastatic triple negative breast cancer. We tested whether combining different immunotherapies can target metastatic triple negative breast cancer in pre-clinical models. METHODS: Using primary and metastatic 4T1 triple negative mammary carcinoma models, we examined the therapeutic effects of oncolytic vesicular stomatitis virus (VSVΔM51) engineered to express reovirus-derived fusion associated small transmembrane proteins p14 (VSV-p14) or p15 (VSV-p15). These viruses were delivered alone or in combination with natural killer T (NKT) cell activation therapy mediated by adoptive transfer of α-galactosylceramide-loaded dendritic cells. RESULTS: Treatment of primary 4T1 tumors with VSV-p14 or VSV-p15 alone increased immunogenic tumor cell death, attenuated tumor growth, and enhanced immune cell infiltration and activation compared to control oncolytic virus (VSV-GFP) treatments and untreated mice. When combined with NKT cell activation therapy, oncolytic VSV-p14 and VSV-p15 reduced metastatic lung burden to undetectable levels in all mice and generated immune memory as evidenced by enhanced in vitro recall responses (tumor killing and cytokine production) and impaired tumor growth upon rechallenge. CONCLUSION: Combining NKT cell immunotherapy with enhanced oncolytic virotherapy increased anti-tumor immune targeting of lung metastasis and presents a promising treatment strategy for metastatic breast cancer.


Sujet(s)
Cellules T tueuses naturelles , Thérapie virale de cancers , Virus oncolytiques , Animaux , Femelle , Souris , Cellules T tueuses naturelles/immunologie , Thérapie virale de cancers/méthodes , Humains , Lignée cellulaire tumorale , Virus oncolytiques/génétique , Virus oncolytiques/immunologie , Immunothérapie/méthodes , Virus de la stomatite vésiculeuse de type Indiana/génétique , Virus de la stomatite vésiculeuse de type Indiana/immunologie , Tumeurs du sein triple-négatives/thérapie , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/anatomopathologie , Association thérapeutique , Métastase tumorale , Vesiculovirus/génétique , Cellules dendritiques/immunologie , Tumeurs du sein/thérapie , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Modèles animaux de maladie humaine
18.
Nat Commun ; 15(1): 3669, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38693119

RÉSUMÉ

Oncolytic viruses (OVs) show promise as a cancer treatment by selectively replicating in tumor cells and promoting antitumor immunity. However, the current immunogenicity induced by OVs for tumor treatment is relatively weak, necessitating a thorough investigation of the mechanisms underlying its induction of antitumor immunity. Here, we show that HSV-1-based OVs (oHSVs) trigger ZBP1-mediated PANoptosis (a unique innate immune inflammatory cell death modality), resulting in augmented antitumor immune effects. Mechanistically, oHSV enhances the expression of interferon-stimulated genes, leading to the accumulation of endogenous Z-RNA and subsequent activation of ZBP1. To further enhance the antitumor potential of oHSV, we conduct a screening and identify Fusobacterium nucleatum outer membrane vesicle (Fn-OMV) that can increase the expression of PANoptosis execution proteins. The combination of Fn-OMV and oHSV demonstrates potent antitumor immunogenicity. Taken together, our study provides a deeper understanding of oHSV-induced antitumor immunity, and demonstrates a promising strategy that combines oHSV with Fn-OMV.


Sujet(s)
Fusobacterium nucleatum , Herpèsvirus humain de type 1 , Thérapie virale de cancers , Virus oncolytiques , Protéines de liaison à l'ARN , Herpèsvirus humain de type 1/immunologie , Herpèsvirus humain de type 1/génétique , Virus oncolytiques/génétique , Virus oncolytiques/immunologie , Animaux , Humains , Thérapie virale de cancers/méthodes , Souris , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/immunologie , Lignée cellulaire tumorale , Fusobacterium nucleatum/immunologie , Tumeurs/thérapie , Tumeurs/immunologie , Femelle , Immunité innée , Souris de lignée BALB C
19.
Adv Exp Med Biol ; 1451: 369-381, 2024.
Article de Anglais | MEDLINE | ID: mdl-38801591

RÉSUMÉ

Despite the significant advancement of new tools and technology in the field of medical biology and molecular biology, the challenges in the treatment of most cancer types remain constant with the problem of developing resistance toward drugs and no substantial enhancement in the overall survival rate of cancer patients. Immunotherapy has shown the most promising results in different clinical and preclinical trials in the treatment of various cancer due to its higher efficacy and minimum collateral damage in many cancer patients as compared to conventional chemotherapy and radiotherapy. An oncolytic virus is a new class of immunotherapy that can selectively replicate in tumor cells and destroy them by the process of cell lysis while exerting minimum or no effect on a normal cell. Besides this, it can also activate the host's innate immune system, which generates an anti-tumor immune response to eliminate the tumor cells. Several wild types and genetically modified viruses have been investigated to show oncolytic behavior. Vaccinia virus has been studied extensively and tested for its promising oncolytic nature on various model systems and clinical trials. Recently, several engineered vaccinia viruses have been developed that express the desired genes encoded for selective penetration in tumor cells and enhanced activation of the immune system for generating anti-tumor immunity. However, further investigation is required to prove their potential and enhance their therapeutic efficacy.


Sujet(s)
Tumeurs , Thérapie virale de cancers , Virus oncolytiques , Poxviridae , Humains , Thérapie virale de cancers/méthodes , Tumeurs/thérapie , Tumeurs/immunologie , Virus oncolytiques/génétique , Virus oncolytiques/physiologie , Animaux , Poxviridae/génétique , Poxviridae/physiologie , Immunothérapie/méthodes , Virus de la vaccine/génétique , Virus de la vaccine/immunologie , Virus de la vaccine/physiologie
20.
Mol Med Rep ; 30(1)2024 07.
Article de Anglais | MEDLINE | ID: mdl-38757346

RÉSUMÉ

Ovarian cancer is a multifactorial and deadly disease. Despite significant advancements in ovarian cancer therapy, its incidence is on the rise and the molecular mechanisms underlying ovarian cancer invasiveness, metastasis and drug resistance remain largely elusive, resulting in poor prognosis. Oncolytic viruses armed with therapeutic transgenes of interest offer an attractive alternative to chemical drugs, which often face innate and acquired drug resistance. The present study constructed a novel oncolytic adenovirus carrying ERCC1 short interfering (si)RNA, regulated by hTERT and HIF promoters, termed Ad­siERCC1. The findings demonstrated that this oncolytic adenovirus effectively inhibits the proliferation, migration and invasion of ovarian cancer cells. Furthermore, the downregulation of ERCC1 expression by siRNA ameliorates drug resistance to cisplatin (DDP) chemotherapy. It was found that Ad­siERCC1 blocks the cell cycle in the G1 phase and enhances apoptosis through the PI3K/AKT­caspase­3 signaling pathways in SKOV3 cells. The results of the present study highlighted the critical effect of oncolytic virus Ad­siERCC1 in inhibiting the survival of ovarian cancer cells and increasing chemotherapy sensitivity to DDP. These findings underscore the potent antitumor effect of Ad­siERCC1 on ovarian cancers in vivo.


Sujet(s)
Adenoviridae , Apoptose , Prolifération cellulaire , Cisplatine , Protéines de liaison à l'ADN , Endonucleases , Thérapie virale de cancers , Virus oncolytiques , Tumeurs de l'ovaire , Petit ARN interférent , Humains , Femelle , Tumeurs de l'ovaire/thérapie , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Adenoviridae/génétique , Lignée cellulaire tumorale , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Endonucleases/génétique , Endonucleases/métabolisme , Apoptose/génétique , Thérapie virale de cancers/méthodes , Virus oncolytiques/génétique , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Cisplatine/pharmacologie , Cisplatine/usage thérapeutique , Mouvement cellulaire/génétique , Résistance aux médicaments antinéoplasiques/génétique , Vecteurs génétiques/génétique , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal , Protéines proto-oncogènes c-akt/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...