280 results on '"oncolytic virus therapy"'
Search Results
2. Durable complete response after combined treatment with tumor-infiltrating lymphocytes and oncolytic adenovirus (TILT-123) in a patient with metastatic mucosal melanoma
- Author
-
T.J. Monberg, T. Kudling, B. Albieri, S. Pakola, E. Ellebaek, M. Donia, R.L. Eefsen, C. von Buchwald, C. Kistler, J.M. Santos, J. Clubb, L. Haybout, M.C.W. Westergaard, D.C.A. Quixabeira, E. Jirovec, R. Havunen, S. Sorsa, V. Cervera-Carrascon, A. Hemminki, and I.M. Svane
- Subjects
case report ,mucosal melanoma ,tumor-infiltrating lymphocytes ,oncolytic virus therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Despite significant advancements in the treatment of malignant melanoma, metastatic mucosal melanoma remains a therapeutic challenge due to its complex pathogenesis, distinct pathological characteristics, and limited response to immunotherapy. Combining different immunotherapeutic approaches offers a potential strategy to address these challenges. Tumor-infiltrating lymphocyte (TIL) therapy and oncolytic virus therapy represent promising treatment modalities that may synergize with each other. Patient and methods: We present a case of a 48-year-old woman with metastatic sinonasal mucosal melanoma who achieved a durable complete pathological response following treatment with multiple injections of the oncolytic virus TILT-123 (igrelimogene litadenorepvec) and a single infusion of TILs, without preconditioning chemotherapy or postconditioning interleukin-2. Results: Immunohistochemical analysis and single-cell sequencing revealed interesting alterations in injected and noninjected tumors as well as in peripheral blood, during the treatment course, suggesting that TILT-123 facilitated TIL engraftment into the tumor, ultimately leading to a complete response. Conclusions: This case underscores the potential of combined immunotherapeutic approaches as a promising strategy for patients with metastatic mucosal melanoma.
- Published
- 2024
- Full Text
- View/download PDF
3. Current and future immunotherapeutic approaches in pancreatic cancer treatment.
- Author
-
Farhangnia, Pooya, Khorramdelazad, Hossein, Nickho, Hamid, and Delbandi, Ali-Akbar
- Subjects
- *
PANCREATIC cancer , *CANCER treatment , *KILLER cells , *IMMUNE checkpoint inhibitors , *PANCREATIC tumors , *CHIMERIC antigen receptors - Abstract
Pancreatic cancer is a major cause of cancer-related death, but despondently, the outlook and prognosis for this resistant type of tumor have remained grim for a long time. Currently, it is extremely challenging to prevent or detect it early enough for effective treatment because patients rarely exhibit symptoms and there are no reliable indicators for detection. Most patients have advanced or spreading cancer that is difficult to treat, and treatments like chemotherapy and radiotherapy can only slightly prolong their life by a few months. Immunotherapy has revolutionized the treatment of pancreatic cancer, yet its effectiveness is limited by the tumor's immunosuppressive and hard-to-reach microenvironment. First, this article explains the immunosuppressive microenvironment of pancreatic cancer and highlights a wide range of immunotherapy options, including therapies involving oncolytic viruses, modified T cells (T-cell receptor [TCR]-engineered and chimeric antigen receptor [CAR] T-cell therapy), CAR natural killer cell therapy, cytokine-induced killer cells, immune checkpoint inhibitors, immunomodulators, cancer vaccines, and strategies targeting myeloid cells in the context of contemporary knowledge and future trends. Lastly, it discusses the main challenges ahead of pancreatic cancer immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. A viral attack on brain tumors: the potential of oncolytic virus therapy.
- Author
-
Mokhtarpour, Kasra, Akbarzadehmoallemkolaei, Milad, and Rezaei, Nima
- Subjects
- *
ONCOLYTIC virotherapy , *BRAIN tumors , *IMMUNE checkpoint inhibitors , *GENE therapy , *BRAIN cancer - Abstract
Managing malignant brain tumors remains a significant therapeutic hurdle that necessitates further research to comprehend their treatment potential fully. Oncolytic viruses (OVs) offer many opportunities for predicting and combating tumors through several mechanisms, with both preclinical and clinical studies demonstrating potential. OV therapy has emerged as a potent and effective method with a dual mechanism. Developing innovative and effective strategies for virus transduction, coupled with immune checkpoint inhibitors or chemotherapy drugs, strengthens this new technique. Furthermore, the discovery and creation of new OVs that can seamlessly integrate gene therapy strategies, such as cytotoxic, anti-angiogenic, and immunostimulatory, are promising advancements. This review presents an overview of the latest advancements in OVs transduction for brain cancer, focusing on the safety and effectiveness of G207, G47Δ, M032, rQNestin34.5v.2, C134, DNX-2401, Ad-TD-nsIL12, NSC-CRAd-S-p7, TG6002, and PVSRIPO. These are evaluated in both preclinical and clinical models of various brain tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Tumor-targeted delivery of copper-manganese biomineralized oncolytic adenovirus for colorectal cancer immunotherapy.
- Author
-
Li, Yi-Shu, Ye, Lu-Yi, Luo, Yan-Xi, Zheng, Wen-Jie, Si, Jing-Xing, Yang, Xue, Zhang, You-Ni, Wang, Shi-Bing, Zou, Hai, Jin, Ke-Tao, Ge, Tong, Cai, Yu, and Mou, Xiao-Zhou
- Subjects
ONCOLYTIC virotherapy ,COLORECTAL cancer ,IMMUNOTHERAPY ,COPPER ions ,GENE amplification ,ANTIGEN presentation ,COPPER ,MANGANESE - Abstract
Oncolytic viral therapy (OVT) is a novel anti-tumor immunotherapy approach, specifically replicating within tumor cells. Currently, oncolytic viruses are mainly administered by intratumoral injection. However, achieving good results for distant metastatic tumors is challenging. In this study, a multifunctional oncolytic adenovirus, OA@CuMnCs, was developed using bimetallic ions copper and manganese. These metal cations form a biomineralized coating on the virus's surface, reducing immune clearance. It is known that viruses upregulate the expression of PD-L1. Copper ions in OA@CuMnCs can decrease the PD-L1 expression of tumor cells, thereby promoting immune cell-related factor release. This process involves antigen presentation and the combination of immature dendritic cells, transforming them into mature dendritic cells. It changes "cold" tumors into "hot" tumors, further inducing immunogenic cell death. While oncolytic virus replication requires oxygen, manganese ions in OA@CuMnCs can react with endogenous hydrogen peroxide. This reaction produces oxygen, enhancing the virus's replication ability and the tumor lysis effect. Thus, this multifunctionally coated OA@CuMnCs demonstrates potent amplification in immunotherapy efficacy, and shows great potential for further clinical OVT. Oncolytic virus therapy (OVs) is a new anti-tumor immunotherapy method that can specifically replicate in tumor cells. Although the oncolytic virus can achieve a therapeutic effect on some non-metastatic tumors through direct intratumoral injection, there are still three major defects in the treatment of metastatic tumors: immune response, hypoxia effect, and administration route. Various studies have shown that the immune response in vivo can be overcome by modifying or wrapping the surface protein of the oncolytic virus. In this paper, a multifunctional coating of copper and manganese was prepared by combining the advantages of copper and manganese ions. The coating has a simple preparation method and mild conditions, and can effectively enhance tumor immunotherapy. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. New hopes for the breast cancer treatment: perspectives on the oncolytic virus therapy.
- Author
-
Chowaniec, Hanna, Ślubowska, Antonina, Mroczek, Magdalena, Borowczyk, Martyna, Braszka, Małgorzata, Dworacki, Grzegorz, Dobosz, Paula, and Wichtowski, Mateusz
- Subjects
ONCOLYTIC virotherapy ,HEAD & neck cancer ,GYNECOLOGIC cancer ,CANCER treatment ,BREAST cancer ,HUMAN herpesvirus 1 ,IMMUNE checkpoint inhibitors - Abstract
Oncolytic virus (OV) therapy has emerged as a promising frontier in cancer treatment, especially for solid tumours. While immunotherapies like immune checkpoint inhibitors and CAR-T cells have demonstrated impressive results, their limitations in inducing complete tumour regression have spurred researchers to explore new approaches targeting tumours resistant to current immunotherapies. OVs, both natural and genetically engineered, selectively replicate within cancer cells, inducing their lysis while sparing normal tissues. Recent advancements in clinical research and genetic engineering have enabled the development of targeted viruses that modify the tumour microenvironment, triggering anti-tumour immune responses and exhibiting synergistic effects with other cancer therapies. Several OVs have been studied for breast cancer treatment, including adenovirus, protoparvovirus, vaccinia virus, reovirus, and herpes simplex virus type I (HSV-1). These viruses have been modified or engineered to enhance their tumour-selective replication, reduce toxicity, and improve oncolytic properties.Newer generations of OVs, such as Oncoviron and Delta-24-RGD adenovirus, exhibit heightened replication selectivity and enhanced anticancer effects, particularly in breast cancer models. Clinical trials have explored the efficacy and safety of various OVs in treating different cancers, including melanoma, nasopharyngeal carcinoma, head and neck cancer, and gynecologic malignancies. Notably, Talimogene laherparepvec (T-VEC) and Oncorine have. been approved for advanced melanoma and nasopharyngeal carcinoma, respectively. However, adverse effects have been reported in some cases, including flu-like symptoms and rare instances of severe complications such as fistula formation. Although no OV has been approved specifically for breast cancer treatment, ongoing preclinical clinical trials focus on four groups of viruses. While mild adverse effects like low-grade fever and nausea have been observed, the effectiveness of OV monotherapy in breast cancer remains insufficient. Combination strategies integrating OVs with chemotherapy, radiotherapy, or immunotherapy, show promise in improving therapeutic outcomes. Oncolytic virus therapy holds substantial potential in breast cancer treatment, demonstrating safety in trials. Multi-approach strategies combining OVs with conventional therapies exhibit more promising therapeutic effects than monotherapy, signalling a hopeful future for OV-based breast cancer treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Neoadjuvant use of oncolytic herpes virus G47Δ prevents local recurrence after insufficient resection in tongue cancer models
- Author
-
Kosuke Inoue, Hirotaka Ito, Miwako Iwai, Minoru Tanaka, Yoshiyuki Mori, and Tomoki Todo
- Subjects
tongue cancer ,G47Δ ,oncolytic virus therapy ,immunotherapy ,neoadjuvant therapy ,local recurrence ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
A complete resection of tongue cancer is often difficult. We investigate the usefulness of administering G47Δ (teserpaturev), a triple-mutated oncolytic herpes simplex virus type 1, prior to resection. G47Δ exhibits good cytopathic effects and replication capabilities in all head and neck cancer cell lines tested. In an orthotopic SCCVII tongue cancer model of C3H/He mice, an intratumoral inoculation with G47Δ significantly prolongs the survival. Further, mice with orthotopic tongue cancer received neoadjuvant G47Δ (or mock) therapy with or without “hemilateral” resection, the maximum extent avoiding surgical deaths. Neoadjuvant G47Δ and resection led to 10/10 survival (120 days), whereas the survivals for G47Δ alone and resection alone were 6/10 and 5/10, respectively: all control animals died by day 11. Furthermore, 100% survival was achieved with neoadjuvant G47Δ therapy even when the resection area was narrowed to “partial,” providing insufficient resection margins, whereas hemilateral resection alone caused death by local recurrence in half of the animals. G47Δ therapy caused increased number of tumor-infiltrating CD8+ and CD4+ cells, increased F4/80+ cells within the residual tongues, and increased expression of immune-related genes in and around the tumor. These results imply that neoadjuvant use of G47Δ is useful for preventing local recurrence after tongue cancer surgery.
- Published
- 2023
- Full Text
- View/download PDF
8. New hopes for the breast cancer treatment: perspectives on the oncolytic virus therapy
- Author
-
Hanna Chowaniec, Antonina Ślubowska, Magdalena Mroczek, Martyna Borowczyk, Małgorzata Braszka, Grzegorz Dworacki, Paula Dobosz, and Mateusz Wichtowski
- Subjects
breast cancer ,immunotherapy ,oncolytic virus therapy ,tumour microenvironment ,TME ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Oncolytic virus (OV) therapy has emerged as a promising frontier in cancer treatment, especially for solid tumours. While immunotherapies like immune checkpoint inhibitors and CAR-T cells have demonstrated impressive results, their limitations in inducing complete tumour regression have spurred researchers to explore new approaches targeting tumours resistant to current immunotherapies. OVs, both natural and genetically engineered, selectively replicate within cancer cells, inducing their lysis while sparing normal tissues. Recent advancements in clinical research and genetic engineering have enabled the development of targeted viruses that modify the tumour microenvironment, triggering anti-tumour immune responses and exhibiting synergistic effects with other cancer therapies. Several OVs have been studied for breast cancer treatment, including adenovirus, protoparvovirus, vaccinia virus, reovirus, and herpes simplex virus type I (HSV-1). These viruses have been modified or engineered to enhance their tumour-selective replication, reduce toxicity, and improve oncolytic properties.Newer generations of OVs, such as Oncoviron and Delta-24-RGD adenovirus, exhibit heightened replication selectivity and enhanced anticancer effects, particularly in breast cancer models. Clinical trials have explored the efficacy and safety of various OVs in treating different cancers, including melanoma, nasopharyngeal carcinoma, head and neck cancer, and gynecologic malignancies. Notably, Talimogene laherparepvec (T-VEC) and Oncorine have. been approved for advanced melanoma and nasopharyngeal carcinoma, respectively. However, adverse effects have been reported in some cases, including flu-like symptoms and rare instances of severe complications such as fistula formation. Although no OV has been approved specifically for breast cancer treatment, ongoing preclinical clinical trials focus on four groups of viruses. While mild adverse effects like low-grade fever and nausea have been observed, the effectiveness of OV monotherapy in breast cancer remains insufficient. Combination strategies integrating OVs with chemotherapy, radiotherapy, or immunotherapy, show promise in improving therapeutic outcomes. Oncolytic virus therapy holds substantial potential in breast cancer treatment, demonstrating safety in trials. Multi-approach strategies combining OVs with conventional therapies exhibit more promising therapeutic effects than monotherapy, signalling a hopeful future for OV-based breast cancer treatments.
- Published
- 2024
- Full Text
- View/download PDF
9. A Review of Current and Pipeline Drugs for Treatment of Melanoma.
- Author
-
Natarelli, Nicole, Aleman, Sarah J., Mark, Isabella M., Tran, Jasmine T., Kwak, Sean, Botto, Elizabeth, Aflatooni, Shaliz, Diaz, Michael J., and Lipner, Shari R.
- Subjects
- *
ONCOLYTIC virotherapy , *CANCER chemotherapy , *LYMPHADENECTOMY , *DRUGS , *RADIOTHERAPY , *BRAF genes , *SURGICAL robots - Abstract
Malignant melanoma is the most aggressive form of skin cancer. Standard treatment options include surgery, radiation therapy, systemic chemotherapy, targeted therapy, and immunotherapy. Combining these modalities often yields better responses. Surgery is suitable for localized cases, sometimes involving lymph node dissection and biopsy, to assess the spread of the disease. Radiation therapy may be sometimes used as a standalone treatment or following surgical excision. Systemic chemotherapy, while having low response rates, is utilized as part of combination treatments or when other methods fail. The development of resistance to systemic chemotherapies and associated side effects have prompted further research and clinical trials for novel approaches. In the case of advanced-stage melanoma, a comprehensive approach may be necessary, incorporating targeted therapies and immunotherapies that demonstrate significant antitumor activity. Targeted therapies, including inhibitors targeting BRAF, MEK, c-KIT, and NRAS, are designed to block the specific molecules responsible for tumor growth. These therapies show promise, particularly in patients with corresponding mutations. Combination therapy, including BRAF and MEK inhibitors, has been evidenced to improve progression-free survival; however, concerns about resistance and cutaneous toxicities highlight the need for close monitoring. Immunotherapies, leveraging tumor-infiltrating lymphocytes and CAR T cells, enhance immune responses. Lifileucel, an FDA-approved tumor-infiltrating lymphocyte therapy, has demonstrated improved response rates in advanced-stage melanoma. Ongoing trials continue to explore the efficacy of CAR T-cell therapy for advanced melanoma. Checkpoint inhibitors targeting CTLA-4 and PD-1 have enhanced outcomes. Emerging IL-2 therapies boost dendritic cells, enhancing anticancer immunity. Oncolytic virus therapy, approved for advanced melanoma, augments treatment efficacy in combination approaches. While immunotherapy has significantly advanced melanoma treatment, its success varies, prompting research into new drugs and factors influencing outcomes. This review provides insights into current melanoma treatments and recent therapeutic advances. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. 免疫治疗在实体瘤中的应用现状.
- Author
-
赵璐, 张正凤, 王大臻, 杨柳, 刘泽, 综述, 娄长杰, and 审校
- Subjects
TUMOR treatment ,ONCOLYTIC virotherapy ,PROGRAMMED death-ligand 1 ,IMMUNOGLOBULINS ,CELLULAR therapy ,IMMUNE checkpoint inhibitors ,MEDICAL practice - Abstract
Copyright of Practical Oncology Journal is the property of Journal of Practical Oncology Editorial Office and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
11. Current Clinical Landscape of Immunotherapeutic Approaches in Pancreatic Cancer Treatment
- Author
-
Farhangnia, Pooya, Mollazadeh Ghomi, Shamim, Mollazadehghomi, Shabnam, Delbandi, Ali-Akbar, Rezaei, Nima, Series Editor, Ahmed, Atif A., Editorial Board Member, Aguiar, Rodrigo, Editorial Board Member, Ambrosio, Maria R., Editorial Board Member, Artac, Mehmet, Editorial Board Member, Augustine, Tanya N., Editorial Board Member, Bambauer, Rolf, Editorial Board Member, Bhat, Ajaz Ahmad, Editorial Board Member, Bertolaccini, Luca, Editorial Board Member, Bianchini, Chiara, Editorial Board Member, Cavic, Milena, Editorial Board Member, Chakrabarti, Sakti, Editorial Board Member, Cho, William C. S., Editorial Board Member, Czarnecka, Anna M., Editorial Board Member, Domingues, Cátia, Editorial Board Member, Eşkazan, A. Emre, Editorial Board Member, Fares, Jawad, Editorial Board Member, Fonseca Alves, Carlos E., Editorial Board Member, Fru, Pascaline, Editorial Board Member, Da Gama Duarte, Jessica, Editorial Board Member, García, Mónica C., Editorial Board Member, Gener, Melissa A.H., Editorial Board Member, Estrada Guadarrama, José Antonio, Editorial Board Member, Hargadon, Kristian M., Editorial Board Member, Holvoet, Paul, Editorial Board Member, Jurisic, Vladimir, Editorial Board Member, Kabir, Yearul, Editorial Board Member, Katsila, Theodora, Editorial Board Member, Kleeff, Jorg, Editorial Board Member, Liang, Chao, Editorial Board Member, Tan, Mei Lan, Editorial Board Member, Li, Weijie, Editorial Board Member, Prado López, Sonia, Editorial Board Member, Macha, Muzafar A., Editorial Board Member, Malara, Natalia, Editorial Board Member, Orhan, Adile, Editorial Board Member, Prado-Garcia, Heriberto, Editorial Board Member, Pérez-Velázquez, Judith, Editorial Board Member, Rashed, Wafaa M., Editorial Board Member, Sanguedolce, Francesca, Editorial Board Member, Sorrentino, Rosalinda, Editorial Board Member, Shubina, Irina Zh., Editorial Board Member, de Araujo, Heloisa Sobreiro Selistre, Editorial Board Member, Torres-Suárez, Ana Isabel, Editorial Board Member, Włodarczyk, Jakub, Editorial Board Member, Yeong, Joe Poh Sheng, Editorial Board Member, Toscano, Marta A., Editorial Board Member, Wong, Tak-Wah, Editorial Board Member, Yin, Jun, Editorial Board Member, and Yu, Bin, Editorial Board Member
- Published
- 2023
- Full Text
- View/download PDF
12. Overcoming resistance of stroma-rich pancreatic cancer with focal adhesion kinase inhibitor combined with G47Δ and immune checkpoint inhibitors
- Author
-
Tomoharu Yamada, Ryosuke Tateishi, Miwako Iwai, Minoru Tanaka, Hideaki Ijichi, Makoto Sano, Kazuhiko Koike, and Tomoki Todo
- Subjects
pancreatic ductal adenocarcinoma ,G47Δ ,oncolytic virus therapy ,focal adhesion kinase inhibitor ,immune checkpoint inhibitor ,teserpaturev ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease known for its dense tumor stroma. Focal adhesion kinase inhibitor (FAKi), a non-receptor type tyrosine kinase inhibitor, reduces the tumor stroma. G47Δ, a third-generation oncolytic herpes simplex virus type 1, destroys tumor cells selectively and induces antitumor immune responses. This study evaluates the efficacy of FAKi and G47Δ in PDAC models in combination with or without immune checkpoint inhibitors. G47Δ was effective in human PDAC cell lines in vitro and in subcutaneous as well as orthotopic tumor models. Transgenic mouse-derived #146 cells were used to generate subcutaneous PDAC tumors with rich stroma in immunocompetent mice. In this #146 tumor model, the efficacy of FAKi was synergistically augmented when combined with G47Δ, which reflected not only a decreased stromal content but also a significant shifting of the tumor microenvironment toward immune stimulation. In transgenic autochthonous PKF mice, a rare model that develops stroma-rich PDAC with a 100% penetrance and resembles human PDAC in various aspects, the prolongation of survival compared with FAKi alone was achieved only when FAKi was combined with G47Δ and immune checkpoint inhibitors. The FAKi combination therapy may be useful to overcome the treatment resistance of stroma-rich PDAC.
- Published
- 2023
- Full Text
- View/download PDF
13. Encouraging probiotics for the prevention and treatment of immune-related adverse events in novel immunotherapies against malignant glioma
- Author
-
Sayuri Yoshikawa, Kurumi Taniguchi, Haruka Sawamura, Yuka Ikeda, Ai Tsuji, and Satoru Matsuda
- Subjects
glioma ,oncolytic virus therapy ,immune-related adverse events ,gut microbiota ,gut-brain-immune axis ,engram ,immune-related disorders ,reactive oxygen species ,Internal medicine ,RC31-1245 - Abstract
Among the malignant tumors in the central nervous system (CNS), glioma is the most challenging tumor to the public society, which accounts for the majority of intracranial malignant tumors with impaired brain function. In general, conventional therapies are still unable to provide an effective cure. However, novel immunotherapies have changed the treatment scene giving patients a greater potential to attain long term survival, improved quality of life. Having shown favorable results in solid tumors, those therapies are now at a cancer research hotspot, which could even shrink the growth of glioma cells without causing severe complications. However, it is important to recognize that the therapy may be occasionally associated with noteworthy adverse action called immune-related adverse events (IRAEs) which have emerged as a potential limitation of the therapy. Multiple classes of mediators have been developed to enhance the ability of immune system to target malignant tumors including glioma but may also be associated with the IRAEs. In addition, it is probable that it would take long time after the therapy to exhibit severe immune-related disorders. Gut microbiota could play an integral role in optimal immune development and/or appropriate function for the cancer therapy, which is a vital component of the multidirectional communication between immune system, brain, and gut, also known as gut-brain-immune axis. Here, we show the potential effects of the gut-brain-immune axis based on an “engram theory” for the innovative treatment of IRAEs.
- Published
- 2022
- Full Text
- View/download PDF
14. Regulatory Issues: PMDA – Review of Sakigake Designation Products: Oncolytic Virus Therapy with Delytact Injection (Teserpaturev) for Malignant Glioma.
- Author
-
Maruyama, Yoshiaki, Sakurai, Akira, Noda, Shinichi, Fujiwara, Yasuhiro, Okura, Narumi, Takagi, Toshinori, Asano, Junichi, and Honda, Futaba
- Subjects
DRUG approval ,INTRAVENOUS therapy ,GENETIC mutation ,GLIOMAS ,DRUG design ,MARKETING ,COMMERCIAL product evaluation ,CELL lines ,T cells ,ONCOLYTIC virotherapy - Abstract
In June 2021, the Ministry of Health, Labor and Welfare approved Delytact Injection as a regenerative medical product for oncolytic virus therapy. The active substance of Delytact Injection is teserpaturev, a genetically engineered herpes simplex virus type 1 (strain F) in which the α47 gene and both copies of the γ34.5 gene have been deleted and the infected cell protein 6 (ICP6) gene has been inactivated by the insertion of the lacZ gene from Escherichia coli. Delytact Injection, when intratumorally administered to patients with malignant glioma, is expected to exert the following effects: (1) the mutant virus selectively replicates in tumor cells and destroys the infected cells through the replication process, exerting a cytocidal effect, and (2) the administration leads to induction of tumor-responsive T cells, which activates antitumor immunity and thus prolongs the survival of patients with malignant glioma. A Japanese phase II study (Study GD01) was conducted in patients with glioblastoma who had residual or recurrent tumors after radiotherapy with concomitant temozolomide. In Study GD01, however, stable disease continued for an extended period in some patients with glioblastoma. Hence, Delytact Injection is expected to be effective to a certain level. In line with this, Delytact Injection has been approved as an option for the treatment of malignant glioma, with one of the 3 approval conditions including conducting a use-results comparison survey and resubmission of the marketing authorization application within the granted time period of 7 years, under the conditional and time-limited approval scheme described in Article 23–26 of Act on Securing Quality, Efficacy and Safety of Products Including Pharmaceuticals and Medical Devices. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. Exploring the Interactions of Oncolytic Viral Therapy and Immunotherapy of Anti-CTLA-4 for Malignant Melanoma Mice Model.
- Author
-
Yu, Jui-Ling, Jang, Sophia R.-J., and Liu, Kwei-Yan
- Subjects
- *
ONCOLYTIC virotherapy , *MELANOMA , *T cells , *ANIMAL disease models , *IMMUNE checkpoint inhibitors , *IMMUNOTHERAPY - Abstract
Oncolytic ability to direct target and lyse tumor cells makes oncolytic virus therapy (OVT) a promising approach to treating cancer. Despite its therapeutic potential to stimulate anti-tumor immune responses, it also has immunosuppressive effects. The efficacy of OVTs as monotherapies can be enhanced by appropriate adjuvant therapy such as anti-CTLA-4. In this paper, we propose a mathematical model to explore the interactions of combined therapy of oncolytic viruses and a checkpoint inhibitor, anti-CTLA-4. The model incorporates both the susceptible and infected tumor populations, natural killer cell population, virus population, tumor-specific immune populations, virus-specific immune populations, tumor suppressive cytokine IFN-g, and the effect of immune checkpoint inhibitor CTLA-4. In particular, we distinguish the tumor-specific immune abilities of CD8+ T, NK cells, and CD4+ T cells and describe the destructive ability of cytokine on tumor cells as well as the inhibitory capacity of CTLA-4 on various components. Our model is validated through the experimental results. We also investigate various dosing strategies to improve treatment outcomes. Our study reveals that tumor killing rate by cytokines, cytokine decay rate, and tumor growth rate play important roles on both the OVT monotherapy and the combination therapy. Moreover, parameters related to CD8+ T cell killing have a large impact on treatment outcomes with OVT alone, whereas parameters associated with IFN-g strongly influence treatment responses for the combined therapy. We also found that virus killing by NK cells may halt the desired spread of OVs and enhance the probability of tumor escape during the treatment. Our study reveals that it is the activation of host anti-tumor immune system responses rather than its direct destruction of the tumor cells plays a major biological function of the combined therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. Manipulating T-cell metabolism to enhance immunotherapy in solid tumor.
- Author
-
Chen Chen, Zehua Wang, Yi Ding, and Yanru Qin
- Subjects
T cells ,IMMUNE checkpoint inhibitors ,METABOLISM ,AEROBIC metabolism ,CELL metabolism - Abstract
Cellular metabolism is not only essential for tumor cells to sustain their rapid growth and proliferation, but also crucial to maintain T cell fitness and robust immunity. Dysregulated metabolism has been recognized as a hallmark of cancer, which provides survival advantages for tumor cells under stress conditions. Also, emerging evidence suggests that metabolic reprogramming impacts the activation, differentiation, function, and exhaustion of T cells. Normal stimulation of resting T cells promotes the conversion of catabolic and oxidative metabolism to aerobic glycolysis in effector T cells, and subsequently back to oxidative metabolism in memory T cells. These metabolic transitions profoundly affect the trajectories of T-cell differentiation and fate. However, these metabolic events of T cells could be dysregulated by their interplays with tumor or the tumor microenvironment (TME). Importantly, metabolic competition in the tumor ecosystem is a new mechanism resulting in strong suppression of effector T cells. It is appreciated that targeting metabolic reprogramming is a promising way to disrupt the hypermetabolic state of tumor cells and enhance the capacity of immune cells to obtain nutrients. Furthermore, immunotherapies, such as immune checkpoint inhibitor (ICI), adoptive cell therapy (ACT), and oncolytic virus (OV) therapy, have significantly refashioned the clinical management of solid tumors, they are not sufficiently effective for all patients. Understanding how immunotherapy affects T cell metabolism provides a bright avenue to better modulate T cell anti-tumor response. In this review, we provide an overview of the cellular metabolism of tumor and T cells, provide evidence on their dynamic interaction, highlight how metabolic reprogramming of tumor and T cells regulate the anti-tumor responses, describe T cell metabolic patterns in the context of ICI, ACT, and OV, and propose hypothetical combination strategies to favor potent T cell functionality. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
17. Bistability in a model of tumor-immune system interactions with an oncolytic viral therapy
- Author
-
G. V. R. K. Vithanage, Hsiu-Chuan Wei, and Sophia R-J Jang
- Subjects
tumor ,immune system ,immune impairment ,oncolytic virus therapy ,global sensitivity analysis ,numerical bifurcation analysis ,Biotechnology ,TP248.13-248.65 ,Mathematics ,QA1-939 - Abstract
A mathematical model of tumor-immune system interactions with an oncolytic virus therapy for which the immune system plays a twofold role against cancer cells is derived. The immune cells can kill cancer cells but can also eliminate viruses from the therapy. In addition, immune cells can either be stimulated to proliferate or be impaired to reduce their growth by tumor cells. It is shown that if the tumor killing rate by immune cells is above a critical value, the tumor can be eradicated for all sizes, where the critical killing rate depends on whether the immune system is immunosuppressive or proliferative. For a reduced tumor killing rate with an immunosuppressive immune system, that bistability exists in a large parameter space follows from our numerical bifurcation study. Depending on the tumor size, the tumor can either be eradicated or be reduced to a size less than its carrying capacity. However, reducing the viral killing rate by immune cells always increases the effectiveness of the viral therapy. This reduction may be achieved by manipulating certain genes of viruses via genetic engineering or by chemical modification of viral coat proteins to avoid detection by the immune cells.
- Published
- 2022
- Full Text
- View/download PDF
18. Efficacy and safety of a third-generation oncolytic herpes virus G47Δ in models of human esophageal carcinoma
- Author
-
Shoh Yajima, Kotaro Sugawara, Miwako Iwai, Minoru Tanaka, Yasuyuki Seto, and Tomoki Todo
- Subjects
oncolytic virus therapy ,G47Δ ,esophageal cancer ,preclinical safety ,orthotopic tumor model ,herpes simplex virus ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Treatment options are limited for esophageal carcinoma (EC). G47Δ, a triple-mutated, conditionally replicating herpes simplex virus type 1 (HSV-1), exhibits enhanced killing of tumor cells with high safety features. Here, we studied the efficacy of G47Δ using preclinical models of human EC. In vitro, G47Δ showed efficient cytopathic effects and replication capabilities in all eight human esophageal cancer cell lines tested. In athymic mice harboring subcutaneous tumors of human EC (KYSE180, TE8, and OE19), two intratumoral injections with G47Δ significantly inhibited the tumor growth. To mimic the clinical treatment situations, we established an orthotopic EC model using luciferase-expressing TE8 cells (TE8-luc). An intratumoral injection with G47Δ markedly inhibited the growth of orthotopic TE8-luc tumors in athymic mice. Furthermore, we evaluated the safety of applying G47Δ to the esophagus in mice. A/J mice inoculated intraesophageally or administered orally with G47Δ (107 plaque-forming units [pfu]) survived for more than 2 months without remarkable symptoms, whereas the majority with wild-type HSV-1 (106 pfu) deteriorated within 10 days. PCR analyses showed that the G47Δ DNA was confined to the esophagus after intraesophageal inoculation and was not detected in major organs after oral administration. Our results provide a rationale for the clinical use of G47Δ for treating EC.
- Published
- 2021
- Full Text
- View/download PDF
19. Oncolytic herpes virus G47Δ works synergistically with CTLA-4 inhibition via dynamic intratumoral immune modulation
- Author
-
Kotaro Sugawara, Miwako Iwai, Hirotaka Ito, Minoru Tanaka, Yasuyuki Seto, and Tomoki Todo
- Subjects
oncolytic virus therapy ,G47Δ ,CTLA-4 ,PD-1 ,immune-checkpoint inhibitors ,herpes simplex virus type 1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Oncolytic virus therapy can increase the immunogenicity of tumors and remodel the immunosuppressive tumor microenvironment, leading to an increased antitumor response to immune-checkpoint inhibitors. Here, we investigated the therapeutic potential of G47Δ, a third-generation oncolytic herpes simplex virus type 1, in combination with immune-checkpoint inhibitors using various syngeneic murine subcutaneous tumor models. Intratumoral inoculations with G47Δ and systemic anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibody administration caused an enhanced antitumor activity when combined and worked synergistically. Conversely, the efficacy of G47Δ in combination with anti-programmed cell death protein-1 (PD-1) antibody was equivalent to that of the anti-PD-1 antibody alone in all murine models examined. The combination of intratumoral G47Δ and systemic anti-CTLA-4 antibody was shown to recruit effector T cells into the tumor efficiently while decreasing regulatory T cells. Furthermore, a wide range of gene signatures related to inflammation, lymphoid lineage, and T cell activation was highly upregulated with the combination therapy, suggesting the conversion of immune-insusceptible tumors to immune susceptible. The therapeutic effect proved tumor specific and long lasting. Immune cell subset depletion studies demonstrated that CD4+ T cells were required for synergistic curative activity. The results depict the dynamics of immune modulation of the tumor microenvironment and provide a clinical rationale for using G47Δ with immune checkpoint inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
20. Oncolytic herpes virus G47Δ injected into tongue cancer swiftly traffics in lymphatics and suppresses metastasis
- Author
-
Toshihiro Uchihashi, Hirokazu Nakahara, Hiroshi Fukuhara, Miwako Iwai, Hirotaka Ito, Akinari Sugauchi, Minoru Tanaka, Mikihiko Kogo, and Tomoki Todo
- Subjects
G47Δ ,oncolytic virus therapy ,lymph node metastasis ,tongue cancer ,antitumor immunity ,oral squamous cell carcinoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The prognosis of oral squamous cell carcinoma (OSCC) largely depends on the control of lymph node metastases. We evaluate the therapeutic efficacy of G47Δ, a third-generation oncolytic herpes simplex virus type 1 (HSV-1), in mouse tongue cancer models. Intratumoral injection with G47Δ prolonged the survival in all orthotopic models investigated. In both athymic and immunocompetent models, G47Δ injected into the tongue cancer swiftly traffics to the draining cervical lymph nodes and suppresses lymph node metastases. In the immunocompetent KLN205-MUC1 model, in which the metastatic cascade that tongue cancer patients commonly experience is reproduced, intratumoral G47Δ injection even immediately prior to a tumor resection prolonged survival. Cervical lymph nodes 18 h after G47Δ treatment showed the presence of G47Δ infection and an increase in CD69-positive cells, indicating an immediate activation of T cells. Furthermore, G47Δ injected directly into enlarged metastatic lymph nodes significantly prolonged the survival at an advanced stage. Whereas intratumorally injected oncolytic HSV-1 does not readily circulate in the blood stream, G47Δ is shown to traffic in the lymphatics swiftly. The use of G47Δ can lead to entirely new treatment strategies for tongue cancer and other OSCC at all clinical stages.
- Published
- 2021
- Full Text
- View/download PDF
21. On a mathematical model of tumor-immune system interactions with an oncolytic virus therapy.
- Author
-
Jang, Sophia R-J and Wei, Hsiu-Chuan
- Subjects
ONCOLYTIC virotherapy ,MATHEMATICAL models ,GLOBAL asymptotic stability ,NATURAL immunity ,VIRUS diseases - Abstract
We investigate a mathematical model of tumor–immune system interactions with oncolytic virus therapy (OVT). Susceptible tumor cells may become infected by viruses that are engineered specifically to kill cancer cells but not healthy cells. Once the infected cancer cells are destroyed by oncolysis, they release new infectious virus particles to help kill surrounding tumor cells. The immune system constructed includes innate and adaptive immunities while the adaptive immunity is further separated into anti-viral or anti-tumor immune cells. The model is first analyzed by studying boundary equilibria and their stability. Numerical bifurcation analysis is performed to investigate the outcomes of the oncolytic virus therapy. The model has a unique tumor remission equilibrium, which is unlikely to be stable based on the parameter values given in the literature. Multiple stable positive equilibria with tumor sizes close to the carrying capacity coexist in the system if the tumor is less antigenic. However, as the viral infection rate increases, the OVT becomes more effective in the sense that the tumor can be dormant for a longer period of time even when the tumor is weakly antigenic. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. An Extensive Review on Preclinical and Clinical Trials of Oncolytic Viruses Therapy for Pancreatic Cancer.
- Author
-
Nisar, Maryum, Paracha, Rehan Zafar, Adil, Sidra, Qureshi, Sumair Naseem, and Janjua, Hussnain Ahmed
- Subjects
ONCOLYTIC virotherapy ,PANCREATIC cancer ,BEGOMOVIRUSES ,CLINICAL trials ,TUMOR antigens ,CANCER treatment ,PANCREATIC tumors - Abstract
Chemotherapy resistance and peculiar tumor microenvironment, which diminish or mitigate the effects of therapies, make pancreatic cancer one of the deadliest malignancies to manage and treat. Advanced immunotherapies are under consideration intending to ameliorate the overall patient survival rate in pancreatic cancer. Oncolytic viruses therapy is a new type of immunotherapy in which a virus after infecting and lysis the cancer cell induces/activates patients' immune response by releasing tumor antigen in the blood. The current review covers the pathways and molecular ablation that take place in pancreatic cancer cells. It also unfolds the extensive preclinical and clinical trial studies of oncolytic viruses performed and/or undergoing to design an efficacious therapy against pancreatic cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
23. Intravenous Oncolytic Vaccinia Virus Therapy Results in a Differential Immune Response between Cancer Patients.
- Author
-
West, Emma J., Scott, Karen J., Tidswell, Emma, Bendjama, Kaidre, Stojkowitz, Nicolas, Lusky, Monika, Kurzawa, Marta, Prasad, Raj, Toogood, Giles, Ralph, Christy, Anthoney, D. Alan, Melcher, Alan A., Collinson, Fiona J., and Samson, Adel
- Subjects
- *
EXPERIMENTAL design , *VIRUSES , *IMMUNOHISTOCHEMISTRY , *CANCER patients , *BIOTHERAPY , *DESCRIPTIVE statistics , *TUMORS , *DATA analysis software , *IMMUNOTHERAPY - Abstract
Simple Summary: Oncolytic viruses (OVs) have been extensively studied as an immunotherapeutic agent against a variety of cancers with some successes. Immunotherapeutic strategies, such as OVs, aim to transform an immunologically 'cold' tumour microenvironment into a more favourable inflammatory 'hot' tumour. However, it is evident that not all patients have a favourable response to treatment. Furthermore, reliable biomarkers able to predict a patient's response to therapy have not yet been elucidated. We show evidence of a distinct immunologically exhausted profile in patients who do not respond to OV, which may pave the way for the development of predictive biomarkers leading to a more personalised approach to cancer treatment using combination therapies. Pexa-Vec is an engineered Wyeth-strain vaccinia oncolytic virus (OV), which has been tested extensively in clinical trials, demonstrating enhanced cytotoxic T cell infiltration into tumours following treatment. Favourable immune consequences to Pexa-Vec include the induction of an interferon (IFN) response, followed by inflammatory cytokine/chemokine secretion. This promotes tumour immune infiltration, innate and adaptive immune cell activation and T cell priming, culminating in targeted tumour cell killing, i.e., an immunologically 'cold' tumour microenvironment is transformed into a 'hot' tumour. However, as with all immunotherapies, not all patients respond in a uniformly favourable manner. Our study herein, shows a differential immune response by patients to intravenous Pexa-Vec therapy, whereby some patients responded to the virus in a typical and expected manner, demonstrating a significant IFN induction and subsequent peripheral immune activation. However, other patients experienced a markedly subdued immune response and appeared to exhibit an exhausted phenotype at baseline, characterised by higher baseline immune checkpoint expression and regulatory T cell (Treg) levels. This differential baseline immunological profile accurately predicted the subsequent response to Pexa-Vec and may, therefore, enable the development of predictive biomarkers for Pexa-Vec and OV therapies more widely. If confirmed in larger clinical trials, these immunological biomarkers may enable a personalised approach, whereby patients with an exhausted baseline immune profile are treated with immune checkpoint blockade, with the aim of reversing immune exhaustion, prior to or alongside OV therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
24. Nonhuman Primate Adenoviruses of the Human Adenovirus B Species Are Potent and Broadly Acting Oncolytic Vector Candidates.
- Author
-
Bots, Selas T.F., Kemp, Vera, Cramer, Steve J., van den Wollenberg, Diana J.M., Hornsveld, Marten, Lamfers, Martine L.M., van der Pluijm, Gabri, and Hoeben, Rob C.
- Subjects
- *
ADENOVIRUSES , *HUMAN adenoviruses , *SPECIES , *HERD immunity , *HOMINIDS , *PRIMATES - Abstract
The use of human adenoviruses (hAds) as oncolytic agents has demonstrated considerable potential. However, their efficacy in clinical studies is generally moderate and often varies between patients. This may, in part, be attributable to variable pre-existing neutralizing immunity in patients, which can impact the antitumor efficacy and lead to response heterogeneity. Our aim was to isolate new Ads for the development of oncolytic vectors with low prevalence of neutralizing immunity in the human population. To this end, we isolated a collection of new nonhuman primate (nhp) Ads from stool samples of four great ape species held captive. We elected 12 isolates comprising the broadest genetic variability for further characterization. For three new nhpAds, all classified as the human adenovirus B (HAdV-B) species, no neutralizing activity could be detected when exposed to a preparation of immunoglobulins isolated from a pool of >1,000 donors as a surrogate of population immunity. In addition, the nhpAds of the HAdV-B species showed enhanced oncolytic potency compared to nhpAds of the HAdV-C species as well as to human adenovirus type 5 (HAdV-C5) in vitro when tested in a panel of 29 human cancer cell lines. Next-generation sequencing of the viral genomes revealed higher sequence similarity between hAds and nhpAds of HAdV-B compared to HAdV-C, which might underlie the differences in oncolytic ability. As a proof-of-concept, the Rb-binding domain of the E1A protein of the gorilla-derived HAdV-B nhpAd-lumc007 was deleted, thereby creating a new oncolytic derivative, which demonstrated increased oncolytic potential compared to HAdV-C5. Collectively, our data demonstrate that nhpAds of the HAdV-B species can serve as an alternative for the development of potent oncolytic Ad vectors with limited pre-existing neutralizing immunity in humans. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. Novel recombinant coxsackievirus B3 with genetically inserted basic peptide elicits robust antitumor activity against lung cancer
- Author
-
Ligang Cai and Zhiyi Liu
- Subjects
coxsackievirus B3 ,cytokines ,immune responses ,lung cancer ,Oncolytic virus therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Cancer therapy that utilizes oncolytic virus may offer an exciting alternative, and coxsackievirus B3 (CVB3) is a potent oncolytic virus. This study was to assess the oncolytic activities of novel recombinant CVB3 with genetically inserted basic peptides in lung cancer. Recombinant CVB3 was produced in Vero cells, with or without genetically inserted basic peptides. In vitro and in vivo experiments with nude mouse models bearing human lung carcinoma xenografts were performed to examine the antitumor activities. Cytokines and immune responses to the recombinant CVB3 were determined in cynomolgus monkeys. Recombinant CVB3 with genetically inserted basic peptides was associated with significantly higher pH values within tumors. Mice treated with recombinant CVB3 showed significantly less tumor progression, and recombinant CVB3 with genetically inserted basic peptides appeared to enhance tumor suppression. Recombinant CVB3 was associated with significantly less proliferation of various lung cancer cells without affecting proliferation of normal lung fibroblasts. The cytokine profiles of the cynomolgus monkeys were comparable among control group (normal saline solution) and those given recombinant CVB3 with or without fused basic peptides, with no induction of excessive cytokine or immune responses. In conclusions, recombinant CVB3, especially those with fused basic peptides, possess strong antitumor activities without eliciting excessive immune responses.
- Published
- 2020
- Full Text
- View/download PDF
26. Efficacy of a Third-Generation Oncolytic Herpes Virus G47Δ in Advanced Stage Models of Human Gastric Cancer
- Author
-
Kotaro Sugawara, Miwako Iwai, Shoh Yajima, Minoru Tanaka, Kazuyoshi Yanagihara, Yasuyuki Seto, and Tomoki Todo
- Subjects
oncolytic virus therapy ,G47Δ ,gastric cancer ,scirrhous-type gastric cancer ,herpes simplex virus type 1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Advanced gastric cancer, especially scirrhous gastric cancer with peritoneal dissemination, remains refractory to conventional therapies. G47Δ, a third-generation oncolytic herpes simplex virus type 1, is an attractive novel therapeutic agent for solid cancer. In this study, we investigated the therapeutic potential of G47Δ for human gastric cancer. In vitro, G47Δ showed good cytopathic effects and replication capabilities in nine human gastric cancer cell lines tested. In vivo, intratumoral inoculations with G47Δ (2 × 105 or 1 × 106 plaque-forming units [PFU]) significantly inhibited the growth of subcutaneous tumors (MKN45, MKN74, and 44As3). To evaluate the efficacy of G47Δ for advanced-stage models of gastric cancer, we generated an orthotopic tumor model and peritoneal dissemination models of human scirrhous gastric cancer (MKN45-luc and 44As3Luc), which have features mimicking intractable scirrhous cancer patients. G47Δ (1 × 106 PFU) was constantly efficacious whether administered intratumorally or intraperitoneally in the clinically relevant models. Notably, G47Δ injected intraperitoneally readily distributed to, and selectively replicated in, disseminated tumors. Furthermore, flow cytometric analyses of tumor-infiltrating cells in subcutaneous tumors revealed that intratumoral G47Δ injections markedly decreased M2 macrophages while increasing M1 macrophages and natural killer (NK) cells. These findings indicate the usefulness of G47Δ for treating human gastric cancer, including scirrhous gastric cancer and the ones in advanced stages.
- Published
- 2020
- Full Text
- View/download PDF
27. An Extensive Review on Preclinical and Clinical Trials of Oncolytic Viruses Therapy for Pancreatic Cancer
- Author
-
Maryum Nisar, Rehan Zafar Paracha, Sidra Adil, Sumair Naseem Qureshi, and Hussnain Ahmed Janjua
- Subjects
pancreatic cancer ,immunotherapy ,oncolytic virus therapy ,Adenovirus ,recombinant virus ,clinical trials ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Chemotherapy resistance and peculiar tumor microenvironment, which diminish or mitigate the effects of therapies, make pancreatic cancer one of the deadliest malignancies to manage and treat. Advanced immunotherapies are under consideration intending to ameliorate the overall patient survival rate in pancreatic cancer. Oncolytic viruses therapy is a new type of immunotherapy in which a virus after infecting and lysis the cancer cell induces/activates patients’ immune response by releasing tumor antigen in the blood. The current review covers the pathways and molecular ablation that take place in pancreatic cancer cells. It also unfolds the extensive preclinical and clinical trial studies of oncolytic viruses performed and/or undergoing to design an efficacious therapy against pancreatic cancer.
- Published
- 2022
- Full Text
- View/download PDF
28. A general non-local delay model on oncolytic virus therapy.
- Author
-
Wang, Zizi, Zhang, Qian, and Luo, Yong
- Subjects
- *
ONCOLYTIC virotherapy , *IMPLICIT functions , *ONCOGENIC viruses , *CONTINUOUS functions , *DELAY differential equations - Abstract
• A general non-local delay model was developed by age-infected law. • Global stability and uniformly persistence are studied. • Mathematical result support that viruses therapy can decrease the tumor load. • Bayesian information criterion was adopted to select a better model when fitting the experimental data. The oncolytic virus is regarded as a novel, powerful, and biologically safe method of cancer treatment. A general delay differential system was driven by the age-dependent model better to understand the interaction between tumor cells and viruses. General continuous functions F (x , y) and G (x) depict the tumor proliferation rate and virus infection rate. The critical threshold value R 0 was calculated that plays a determinant role in whether virus therapy occurs. The non-local delay term ∫ t − τ t β G (x (θ)) v (θ) e − α (t − θ) d θ makes our model hard to analyze when using the traditional eigenvalue method. The method combining implicit function theorem and comparison theorem is used to overcome this problem. Furthermore, we support the fact that virotherapy can lead to tumor remission by using the fluctuation method. Lastly, Bayesian information criterion was adopted to select a better model when fitting the experimental data. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
29. Age-structure model for oncolytic virotherapy.
- Author
-
Ding, Chuying, Wang, Zizi, and Zhang, Qian
- Subjects
- *
ONCOLYTIC virotherapy , *SENSITIVITY analysis - Abstract
A non-local delay differential system was developed by age-infected consideration to understand the interaction between tumor cells and oncolytic virus. The critical threshold value R 1 was obtained that dominates the dynamical behaviors of our model. By strongly ρ -persistent and Comparison theorem, the global stability and uniformly strongly persistent were investigated. Our global sensitivity analyses indicate that R 1 is most sensitive to virus clearance rate d while is least sensitive to viral life cycle τ. Finally, our model was used to fit with experimental data which indicates that viral life cycle τ plays a key role in data fitting. And the trained model shows perfect prediction in the testing data set with the coefficient of determination R 2 = 9 5. 5 5 %. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
30. 脑胶质瘤的基因治疗与病毒治疗研究进展.
- Author
-
张国栋, 鞠海涛, 黄平, 岳鹏, 万方, and 窦长武
- Abstract
Glioma is a highly lethal primary brain tumor. Due to the drug resistance of cancer stem cell subsets, the difficulty of drugs crossing the blood-brain barrier and immunosuppression, the conventional treatment has not significantly improved the survival time and quality of life of patients. Therefore, with the support of advances in molecular biology, the arduous exploration of new therapies continues. Gene therapy involves the delivery of therapeutic genetic material to tumor cells to produce a specific anti-tumor response. In addition, viruses are highly cytotoxic and easy to modify. The inherent characteristics of horizontal transfer of genetic material make them valuable tools for genetic engineering. This article reviews the application of various viral vectors in glioma gene therapy and oncolytic virus therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
31. Novel recombinant coxsackievirus B3 with genetically inserted basic peptide elicits robust antitumor activity against lung cancer.
- Author
-
Cai, Ligang and Liu, Zhiyi
- Subjects
LUNG cancer ,KRA ,SALINE solutions ,IMMUNE response ,CANCER invasiveness - Abstract
Cancer therapy that utilizes oncolytic virus may offer an exciting alternative, and coxsackievirus B3 (CVB3) is a potent oncolytic virus. This study was to assess the oncolytic activities of novel recombinant CVB3 with genetically inserted basic peptides in lung cancer. Recombinant CVB3 was produced in Vero cells, with or without genetically inserted basic peptides. In vitro and in vivo experiments with nude mouse models bearing human lung carcinoma xenografts were performed to examine the antitumor activities. Cytokines and immune responses to the recombinant CVB3 were determined in cynomolgus monkeys. Recombinant CVB3 with genetically inserted basic peptides was associated with significantly higher pH values within tumors. Mice treated with recombinant CVB3 showed significantly less tumor progression, and recombinant CVB3 with genetically inserted basic peptides appeared to enhance tumor suppression. Recombinant CVB3 was associated with significantly less proliferation of various lung cancer cells without affecting proliferation of normal lung fibroblasts. The cytokine profiles of the cynomolgus monkeys were comparable among control group (normal saline solution) and those given recombinant CVB3 with or without fused basic peptides, with no induction of excessive cytokine or immune responses. In conclusions, recombinant CVB3, especially those with fused basic peptides, possess strong antitumor activities without eliciting excessive immune responses. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
32. Optimal timing of PD-1 blockade in combination with oncolytic virus therapy
- Author
-
Hong-My Nguyen, Ann W. Silk, Praveen K. Bommareddy, and Dipongkor Saha
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Combination therapy ,T cell ,Priming (immunology) ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Neoplasms ,Internal medicine ,Tumor Microenvironment ,Humans ,Medicine ,Oncolytic Virotherapy ,Tumor microenvironment ,business.industry ,Oncolytic virus ,Blockade ,Oncolytic Viruses ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Oncolytic Virus Therapy ,business - Abstract
Anti-PD-1 and oncolytic viruses (OVs) have non-overlapping anti-tumor mechanisms, since each agent works at different steps of the cancer-immunity cycle. Evidence suggests that OVs improve therapeutic responses to anti-PD-1 therapy by reversing immunosuppressive factors, increasing the number and diversity of infiltrating lymphocytes, and promoting PD-L1 expression in both injected and non-injected tumors. Many studies in preclinical models suggest that the timing of anti-PD-1 administration influences the therapeutic success of the combination therapy (anti-PD-1 + OV). Therefore, determining the appropriate sequencing of agents is of critical importance to designing a rationale OV-based combinational clinical trial. Currently, the combination of anti-PD-1 and OVs are being delivered using various schedules, and we have classified the timing of administration of anti-PD-1 and OVs into five categories: (i) anti-PD-1 lead-in → OV; (ii) concurrent administration; (iii) OV lead-in → anti-PD-1; (iv) concurrent therapy lead-in → anti-PD-1; and (v) OV lead-in → concurrent therapy. Based on the reported preclinical and clinical literature, the most promising treatment strategy to date is hypothesized to be OV lead-in → concurrent therapy. In the OV lead-in → concurrent therapy approach, initial OV treatment results in T cell priming and infiltration into tumors and an immunologically hot tumor microenvironment (TME), which can be counterbalanced by engagement of PD-L1 to PD-1 receptor on immune cells, leading to T cell exhaustion. Therefore, after initial OV therapy, concurrent use of both OV and anti-PD-1 is critical through which OV maintains T cell priming and an immunologically hot TME, whereas PD-1 blockade helps to overcome PD-L1/PD-1-mediated T cell exhaustion. It is important to note that the hypothetical conclusion drawn in this review is based on thorough literature review on current understanding of OV + anti-PD-1 combination therapies and rhythm of treatment-induced cancer-immunity cycle. A variety of confounding factors such as tumor types, OV types, presence or absence of cytokine transgenes carried by an OV, timing of treatment initiation, varying dosages and treatment frequencies/duration of OV and anti-PD-1, etc. may affect the validity of our conclusion that will need to be further examined by future research (such as side-by-side comparative studies using all five treatment schedules in a given tumor model).
- Published
- 2022
- Full Text
- View/download PDF
33. Neuroimmune crosstalk and its impact on cancer therapy and research
- Author
-
Sharawy, Iman
- Published
- 2022
- Full Text
- View/download PDF
34. HIV-Encoded Gene Therapy as Anti-cancer Therapeutics: A Narrative Review.
- Author
-
Balakrishnan P, Sathish S, and Saravanan S
- Abstract
Recently, there has been interest in using viruses as cancer treatments. Oncolytic virology was founded by scientists who noticed that viruses might preferentially lyse cancer cells over healthy ones. Oncolytic virotherapy has similar obstacles as other treatment approaches, gaining entry into the specific tumour cell, encountering antiviral immune responses, off-target infection and many other unfavourable circumstances in the tumour microenvironment, and a lack of unique therapeutic and predictive biomarkers. However, oncolytic viruses have emerged as the main players in the biological treatment for cancer with the use of vectors such as human adenoviruses in oncolytic virotherapy. Recent large-scale research has shown that other viruses, such as the measles virus and the herpes simplex virus (HSV), may potentially be viable options for cancer treatment. The FDA has cleared T-VEC, an HSV-based oncolytic virus, for use in biological cancer treatment after its successful completion of human clinical trials. Furthermore, the measles virus vaccine strain has shown remarkable outcomes in pre-clinical and clinical testing. The use of such modified viruses in biological cancer treatment holds promise for groundbreaking discoveries in the field of cancer research because of their therapeutic effectiveness, fewer side effects, and safety. Several other newer approaches have been used in recent years. HIV-encoded proteins are also hypothesized to promote mitochondrial homeostasis causing bystander-induced apoptosis. We provide an overview of the most recent developments in the clinical use of oncolytic virus-based biological cancer treatment in this study. This evaluation also assesses the advantages and disadvantages of the viral candidates and provides insight into their potential in the future., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2024, Balakrishnan et al.)
- Published
- 2024
- Full Text
- View/download PDF
35. A Comparative Safety Profile Assessment of Oncolytic Virus Therapy Based on Clinical Trials.
- Author
-
Takuma Matsuda, Hiroyo Karube, and Atsushi Aruga
- Subjects
THERAPEUTIC use of antineoplastic agents ,CLINICAL trials ,IN vivo studies ,FEVER ,NAUSEA ,ONCOGENES ,RISK assessment ,SHIVERING ,TOXICITY testing ,TUMORS ,ADVERSE health care events ,FATIGUE (Physiology) ,ONCOLYTIC virotherapy ,PATIENT safety - Abstract
Oncolytic virus therapy (OVT) represents a new class of therapeutic agents in cancer treatment. The molecular and cellular mechanisms of action of OVTs have been evaluated in nonclinical/clinical phase trials. Various genetically modified viruses have been developed as oncolytic agents, and the first approval of an OVT for clinical use was issued by the US Food and Drug Administration in 2015. In this context, more and more clinical development of OVTs is anticipated in the future. This article provides a risk assessment of OVT based on the safety data obtained from all clinical trials to date using a publicly available database. The most common adverse events (AEs) observed in clinical trials have been infection-related symptoms such as fatigue, chills, fever, and nausea; few serious AEs have been observed, regardless of the kind of virus or transfected genes. In vivo systemic infusion of OVTs demonstrated a high percentage of AEs, but most AEs were manageable using common drugs. This paper describes OVTs' specific safety/toxicity profiles and encourages the performance of further clinical trials of OVTs to address the most serious challenges anticipated in the development of OVTs as a new class of drugs for the treatment of cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
36. The ever‐expanding landscape of cancer therapeutic approaches
- Author
-
Christine J. Watson
- Subjects
medicine.medical_specialty ,Glycosylation ,business.industry ,Cancer ,Cell Biology ,Disease ,Precision medicine ,medicine.disease ,Biochemistry ,RNA Precursors ,medicine ,Humans ,Oncolytic Virus Therapy ,Calcium ,Intensive care medicine ,business ,Molecular Biology - Abstract
Cancer is a leading cause of death and a major health problem worldwide, particularly in more developed countries. There is, therefore, an urgent clinical need to develop more effective therapies to treat cancer and metastatic disease. In this Editorial, the content of The FEBS Journal's Special Issue on Cancer Therapeutics is outlined. The interesting collection of recent articles in this issue covers a wide repertoire of cancer therapeutic approaches. While some of the articles discuss broad-spectrum applications such as immunotherapy and oncolytic virus therapy, others focus on a particular type of cancer or a signalling pathway that has gone awry such as aberrant Ca2+ signalling, glycosylation or pre-mRNA processing. Finally, an article featured in this issue reviews our current understanding of how cancer cells can become dormant, often for decades, and which pathways reactivate these cells to cause relapse. I am sure there is something for everyone in this issue.
- Published
- 2021
- Full Text
- View/download PDF
37. Evaluation of the oncolytic property of recombinant Newcastle disease virus strain R2B in 4T1 and B16-F10 cells in-vitro
- Author
-
Vikram N. Vakharia, Asok Kumar Mariappan, Sohini Dey, Madhan Mohan Chellappa, Narayan Ramamurthy, Ajai Lawrence D’Silva, Dinesh C. Pathak, and Rahul Batheja
- Subjects
Oncolytic Virotherapy ,General Veterinary ,Membrane permeability ,viruses ,Newcastle disease virus ,Apoptosis ,Mammary Neoplasms, Animal ,Biology ,Recombinant virus ,Molecular biology ,Virus ,Oncolytic virus ,Rodent Diseases ,Mice ,Cell culture ,Cell Line, Tumor ,Cancer cell ,Animals ,Oncolytic Virus Therapy ,Melanoma - Abstract
Recombinant Newcastle disease virus vectors have gained a lot of interest for its oncolytic virus therapy and cancer immune therapeutic properties due to its selective replication to high titers in cancer cells. The aim of this study was to find out the oncolytic effects of mesogenic recombinant NDV strain R2B-GFP on murine mammary tumor cell line 4T1 and murine melanoma cell line B16-F10. The anti-tumor effects of R2B-GFP virus were studied via expression of virus transgene GFP in cancer cells, evaluating its cytotoxicity and cell migration efficacies by MTT and wound healing assays respectively. In addition, the underlying apoptotic mechanism of R2B-GFP virus was estimated by TUNEL assay, colorimetric estimation of Caspase-3, 8 and 9 and the estimation of Bax to Bcl-2 ratio. The results showed a significant decrease in viability of both 4T1 and B16-F10 cells infected with R2B-GFP virus at 0.1 and 1 MOI. R2B-GFP virus could significantly induce apoptosis in the 4T1 and B16-F10 cells as compared to the uninfected control. Further, a flow cytometry analysis on apoptotic cells percentage and mitochondria membrane permeability test was also studied in R2B-GFP virus treated 4T1 and B16-F10 cell lines. The R2B-GFP virus caused an increase in loss of mitochondrial membrane permeability in both 4T1 and B16-F10 cells indicating the involvement of mitochondrial regulated cell death. Thus, the recombinant virus R2B-GFP virus proved to be a valid candidate for oncolytic viral therapy in 4T1 and B16-F10 cells.
- Published
- 2021
- Full Text
- View/download PDF
38. Oncolytic herpes virus G47Δ works synergistically with CTLA-4 inhibition via dynamic intratumoral immune modulation
- Author
-
Miwako Iwai, Minoru Tanaka, Kotaro Sugawara, Yasuyuki Seto, Tomoki Todo, and Hirotaka Ito
- Subjects
Cancer Research ,antitumor immunity ,immune-checkpoint inhibitors ,herpes simplex virus type 1 ,Oncolytic herpes virus ,regulatory T cells ,esophageal carcinoma ,Immune system ,PD-1 ,Medicine ,tumor microenvironment ,Pharmacology (medical) ,RC254-282 ,G47Δ ,Tumor microenvironment ,biology ,business.industry ,Immunogenicity ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncolytic virus ,oncolytic virus therapy ,Oncology ,CTLA-4 ,biology.protein ,Cancer research ,Molecular Medicine ,Oncolytic Virus Therapy ,Original Article ,Antibody ,business - Abstract
Oncolytic virus therapy can increase the immunogenicity of tumors and remodel the immunosuppressive tumor microenvironment, leading to an increased antitumor response to immune-checkpoint inhibitors. Here, we investigated the therapeutic potential of G47Δ, a third-generation oncolytic herpes simplex virus type 1, in combination with immune-checkpoint inhibitors using various syngeneic murine subcutaneous tumor models. Intratumoral inoculations with G47Δ and systemic anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibody administration caused an enhanced antitumor activity when combined and worked synergistically. Conversely, the efficacy of G47Δ in combination with anti-programmed cell death protein-1 (PD-1) antibody was equivalent to that of the anti-PD-1 antibody alone in all murine models examined. The combination of intratumoral G47Δ and systemic anti-CTLA-4 antibody was shown to recruit effector T cells into the tumor efficiently while decreasing regulatory T cells. Furthermore, a wide range of gene signatures related to inflammation, lymphoid lineage, and T cell activation was highly upregulated with the combination therapy, suggesting the conversion of immune-insusceptible tumors to immune susceptible. The therapeutic effect proved tumor specific and long lasting. Immune cell subset depletion studies demonstrated that CD4+ T cells were required for synergistic curative activity. The results depict the dynamics of immune modulation of the tumor microenvironment and provide a clinical rationale for using G47Δ with immune checkpoint inhibitors., Graphical abstract, G47Δ, a third-generation oncolytic HSV-1, works synergistically with CTLA-4 inhibition, but not PD-1, in murine tumor models. The combination therapy causes increased effector T cells, decreased Tregs, and upregulated gene signatures related to immunogenicity within tumors. The therapeutic effect proved tumor specific and long lasting and requires CD4+ cells for synergy.
- Published
- 2021
39. Positron emission tomography imaging with 89Zr-labeled anti-CD8 cys-diabody reveals CD8+ cell infiltration during oncolytic virus therapy in a glioma murine model
- Author
-
Anna M. Wu, Jason M. Warram, Jianmei W. Leavenworth, Anna G. Sorace, Felix B. Salazar, Jennifer M. Coleman, Benjamin B. Kasten, James M. Markert, Adriana V.F. Massicano, Suzanne E. Lapi, Hailey A. Houson, G. Yancey Gillespie, and Richard Tavaré
- Subjects
0301 basic medicine ,T cell ,medicine.medical_treatment ,CD8 Antigens ,Science ,Standardized uptake value ,CD8-Positive T-Lymphocytes ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Glioma ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Simplexvirus ,Author Correction ,Cancer ,Oncolytic Virotherapy ,Radioisotopes ,Multidisciplinary ,Molecular medicine ,medicine.diagnostic_test ,business.industry ,Immunotherapy ,medicine.disease ,Oncolytic virus ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Positron emission tomography ,030220 oncology & carcinogenesis ,Cancer research ,Oncolytic Virus Therapy ,Medicine ,Zirconium ,business ,Tomography, X-Ray Computed ,Biomarkers ,Ex vivo - Abstract
Determination of treatment response to immunotherapy in glioblastoma multiforme (GBM) is a process which can take months. Detection of CD8+ T cell recruitment to the tumor with a noninvasive imaging modality such as positron emission tomography (PET) may allow for tumor characterization and early evaluation of therapeutic response to immunotherapy. In this study, we utilized 89Zr-labeled anti-CD8 cys-diabody-PET to provide proof-of-concept to detect CD8+ T cell immune response to oncolytic herpes simplex virus (oHSV) M002 immunotherapy in a syngeneic GBM model. Immunocompetent mice (n = 16) were implanted intracranially with GSC005 GBM tumors, and treated with intratumoral injection of oHSV M002 or saline control. An additional non-tumor bearing cohort (n = 4) receiving oHSV M002 treatment was also evaluated. Mice were injected with 89Zr-labeled anti-CD8 cys-diabody seven days post oHSV administration and imaged with a preclinical PET scanner. Standardized uptake value (SUV) was quantified. Ex vivo tissue analyses included autoradiography and immunohistochemistry. PET imaging showed significantly higher SUV in tumors which had been treated with M002 compared to those without M002 treatment (p = 0.0207) and the non-tumor bearing M002 treated group (p = 0.0021). Accumulation in target areas, especially the spleen, was significantly reduced by blocking with the non-labeled diabody (p + cell trafficking patterns after oHSV treatment. This PET imaging strategy could aid in distinguishing responders from non-responders during immunotherapy of GBM.
- Published
- 2021
40. Triple‐mutated oncolytic herpes virus for treating both fast‐ and slow‐growing tumors
- Author
-
Hiroshi Fukuhara, Yuta Takeshima, and Tomoki Todo
- Subjects
0301 basic medicine ,Cancer Research ,Chromosomes, Artificial, Bacterial ,viruses ,herpes simplex virus type 1 ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Oncolytic herpes virus ,Immediate-Early Proteins ,03 medical and health sciences ,Mice ,Viral Proteins ,0302 clinical medicine ,DU145 ,Cell Line, Tumor ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Telomerase reverse transcriptase ,Promoter Regions, Genetic ,Telomerase ,Vero Cells ,oncolytic virus ,Oncolytic Virotherapy ,Bacterial artificial chromosome ,Brain Neoplasms ,Cancer ,Epidemiology and Prevention ,General Medicine ,Original Articles ,medicine.disease ,Xenograft Model Antitumor Assays ,Oncolytic virus ,Oncolytic Viruses ,030104 developmental biology ,Herpes simplex virus ,Oncology ,030220 oncology & carcinogenesis ,G47∆ ,Mutation ,Cancer research ,Oncolytic Virus Therapy ,Original Article ,Female ,hTERT ,Glioblastoma ,ICP6 - Abstract
Oncolytic virus therapy has emerged as a promising treatment option against cancer. To date, oncolytic viruses have been developed for malignant tumors, but the need for this new therapeutic modality also exists for benign and slow‐growing tumors. G47∆ is an oncolytic herpes simplex virus type 1 (HSV‐1) with an enhanced replication capability highly selective to tumor cells due to genetically engineered, triple mutations in the γ34.5, ICP6 and α47 genes. To create a powerful, but safe oncolytic HSV‐1 that replicates efficiently in tumors regardless of growth speed, we used a bacterial artificial chromosome system that allows a desired promoter to regulate the expression of the ICP6 gene in the G47∆ backbone. Restoration of the ICP6 function in a tumor‐specific manner using the hTERT promoter led to a highly capable oncolytic HSV‐1. T‐hTERT was more efficacious in the slow‐growing OS‐RC‐2 and DU145 tumors than the control viruses, while retaining a high efficacy in the fast‐growing U87MG tumors. The safety features are also retained, as T‐hTERT proved safe when inoculated into the brain of HSV‐1 sensitive A/J mice. This new technology should facilitate the use of oncolytic HSV‐1 for all tumors irrespective of growth speed., We report a powerful oncolytic HSV‐1 that can replicate explosively in any tumor regardless of its growth speed using clinically used, third‐generation HSV‐1, G47∆, as the backbone. Tumor‐specific restoration of the ICP6 function led to a highly capable oncolytic HSV‐1 with intended features including safety. This new technology should facilitate the use of oncolytic HSV‐1 for all tumors whether benign or malignant.
- Published
- 2021
41. Dynamics and optimal immunotherapy for models of tumor-immune system interactions with oncolytic viruses
- Author
-
Vithana, Rohana Kumara and Vithana, Rohana Kumara
- Abstract
Immunotherapy has become a valuable weapon for combating cancer of various types. Oncolytic virus therapy in particular is regarded as the next major breakthrough in cancer treatment following the success of immune checkpoint inhibitors. The aim of this dissertation is to investigate the efficacy of oncolytic viral therapy by constructing mathematical models that take into account tumor suppressive effects. Therein the immune cells kill not only cancer cells but also viruses from the therapy. We first evaluate therapy effectiveness with respect to long term dynamical behavior of the tumor. Without tumor saturation effects, it is shown that if the tumor killing rate by immune cells is above a critical value, the tumor can be eradicated for all sizes. For a reduced tumor killing rate with a tumor-suppressed immune response, that bistability exists in a large parameter space and reducing the viral killing rate by immune cells always increases the effectiveness of the viral therapy. For the model with tumor saturation effects, it is proven that the tumor-immune system interaction can support at most three positive equilibria where two of them are stable and the other is a saddle point. Moreover, an immunosuppressive microenvironment may enhance viral therapy efficacy. We also apply optimal control techniques to devise best therapeutic strategies by minimizing susceptible tumor cells as well as the costs associated with the treatment over a short time period.
- Published
- 2022
42. Oncolytic reovirus-mediated recruitment of early innate immune responses reverses immunotherapy resistance in prostate tumors
- Author
-
M. Denyer, Nicola E. Annels, Guy Simpson, Kevin J. Harrington, Richard G. Vile, Mehreen Arif, Hardev Pandha, Alan Melcher, and Matthew C. Coffey
- Subjects
0301 basic medicine ,Cancer Research ,Oncolytic virus ,medicine.medical_treatment ,BTLA ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Interferon ,medicine ,Pharmacology (medical) ,Prostate cancer ,Innate immune system ,business.industry ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immune checkpoint ,Immunotherapy resistance ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Oncolytic Virus Therapy ,Original Article ,business ,medicine.drug - Abstract
Prostate cancers are considered “cold” tumors characterized by minimal T cell infiltrates, absence of a type I interferon (IFN) signature, and the presence of immunosuppressive cells. This non-inflamed phenotype is likely responsible for the lack of sensitivity of prostate cancer patients to immune checkpoint blockade (ICB) therapy. Oncolytic virus therapy can potentially overcome this resistance to immunotherapy in prostate cancers by transforming cold tumors into “hot,” immune cell-infiltrated tumors. We investigated whether the combination of intratumoral oncolytic reovirus, followed by targeted blockade of Programmed cell death protein 1 (PD-1) checkpoint inhibition and/or the immunomodulatory CD73/Adenosine system can enhance anti-tumor immunity. Treatment of subcutaneous TRAMP-C2 prostate tumors with combined intratumoral reovirus and anti-PD-1 or anti-CD73 antibody significantly enhanced survival of mice compared with reovirus or either antibody therapy alone. Only combination therapy led to rejection of pre-established tumors and protection from tumor re-challenge. This therapeutic effect was dependent on CD4+ T cells and natural killer (NK) cells. NanoString immune profiling of tumors confirmed that reovirus increased tumor immune cell infiltration and revealed an upregulation of the immune-regulatory receptor, B- and T-lymphocyte attenuator (BTLA). This expression of BTLA on innate antigen-presenting cells (APCs) and its ligand, Herpesvirus entry mediator (HVEM), on T cells from reovirus-infected tumors was in keeping with a role for the HVEM-BTLA pathway in promoting the potent anti-tumor memory response observed., Graphical Abstract, Immunotherapy in prostate cancer is limited because of a lack of immune cells within the tumor. Annels et al. used oncolytic viruses to recruit immune cells into the tumor and showed that in combination with checkpoint inhibitors, which take the brake off of the immune system, established tumors were rejected.
- Published
- 2021
- Full Text
- View/download PDF
43. A novel vaccinia virus enhances anti-tumor efficacy and promotes a long-term anti-tumor response in a murine model of colorectal cancer
- Author
-
Na Wang, Louisa S Chard Dunmall, Hua Cao, Jiwei Wang, Zhe Zhang, Yaohe Wang, Yongchao Chu, and Wenli Yan
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,medicine.medical_treatment ,lcsh:RC254-282 ,Virus ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Immunity ,Medicine ,Pharmacology (medical) ,business.industry ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,Cytokine ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Oncolytic Virus Therapy ,Original Article ,Vaccinia ,business - Abstract
Colorectal cancer (CRC) is one of the leading causes of mortality and morbidity in the world, and there remains an urgent need to develop long-lasting therapies to treat CRC and prevent recurrence in patients. Oncolytic virus therapy (OVT) has demonstrated remarkable efficacy in a number of different cancer models. Here, we report a novel vaccinia virus (VV)-based OVT for treatment of CRC. The novel VV, based on the recently reported novel VVLΔTKΔN1L virus, was armed with the pleiotropic cytokine interleukin-21 (IL-21) to enhance anti-tumor immune responses stimulated after viral infection of tumor cells. Compared with an unarmed virus, VVLΔTKΔN1L-mIL-21 had a superior anti-tumor efficacy in murine CMT93 subcutaneous CRC models in vivo, mediated mainly by CD8+ T cells. Treatment resulted in development of long-term immunity against CMT93 tumor cells, as evidenced by prevention of disease recurrence. These results demonstrate that VVLΔTKΔN1L-mIL-21 is a promising therapeutic agent for treatment of CRC., Graphical Abstract, A novel tumor-targeted replicating vaccinia virus expressing IL-21 demonstrates a superior antitumor efficacy for murine colorectal cancer, mediated mainly by immune cells. The virus treatment also results in development of long-term tumor-specific immunity to prevent recurrence. These results provide a promising therapeutic agent for treatment of colorectal cancer.
- Published
- 2021
- Full Text
- View/download PDF
44. Suppression of CCDC6 sensitizes tumor to oncolytic virus M1
- Author
-
Guangmei Yan, Ying Liu, Jiankai Liang, Ke Li, Xincheng Liu, Wentao Huang, Cheng Hu, Yuan Lin, Jing Cai, Hao Zhang, Liang Peng, Wenbo Zhu, and Jianguang Qiu
- Subjects
0301 basic medicine ,Cancer Research ,Oncolytic virus ,ISG, interferon-stimulated genes ,viruses ,Gene Expression ,Apoptosis ,CCDC6, Coiled-coil-domain containing 6 ,Mice ,0302 clinical medicine ,Interferon pathway ,Neoplasms ,DNA-PKI, DNA-PK inhibitor ,RNA, Small Interfering ,Oncolytic Virotherapy ,Gene knockdown ,M1 virus ,HRP, horseradish peroxidase ,Transfection ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Endoplasmic Reticulum Stress ,ZAP, zinc finger antiviral protein ,Oncolytic Viruses ,XBP1, X-box binding protein 1 ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,RT, room temperature ,Oncolytic Virus Therapy ,Female ,Original article ,DAUCs, area under the curve ,PVDF, polyvinylidene fluoride ,siRNA, Small interfering RNAs ,Genetic Vectors ,Alphavirus ,Biology ,lcsh:RC254-282 ,Virus ,03 medical and health sciences ,DNA-PK, DNA-dependent protein kinase ,Cell Line, Tumor ,Animals ,Humans ,Genetic Therapy ,biology.organism_classification ,Xenograft Model Antitumor Assays ,IFNs, Interferons ,VCP, valosin-containing protein ,Cytoskeletal Proteins ,Disease Models, Animal ,030104 developmental biology ,CCDC6 ,Cancer cell ,Cancer research ,ATM, ataxia telangiectasia-mutated - Abstract
Oncolytic virus is an effective therapeutic strategy for cancer treatment, which exploits natural or manipulated viruses to selectively target and kill cancer cells. However, the innate antiviral system of cancer cells may resistant to the treatment of oncolytic virus. M1 virus is a newly identified oncolytic virus belonging to alphavirus species, but the molecular mechanisms underlying its anticancer activity are largely unknown. Cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assays. RNA seq analysis was used to analyze the gene alternation after M1 virus infection. Small interfering RNAs transfection for gene knockdown was used for gene functional tests. Caspase-3/7 activity was detected by Caspase-Glo Assay Systems. A mice model of orthotopic bladder tumor was established to determine the oncolytic effectiveness of the M1 virus. The expression of cleaved-Caspase 3 as well as Ki-67 in tumor cells were detected by immunohistochemical analysis. To further define the molecular factors involved in M1 virus-mediated biological function, we knocked down genes related to alphavirus’ activity and found that CCDC6 plays an important role in the oncolytic activity of M1 virus. Moreover, knocked down of CCDC6 augments the reproduction of M1 virus and resulted in endoplasmic reticulum (ER) stress-induced cell apoptosis in vitro as well as in vivo orthotopic bladder cancer model. Our research provides a rational new target for developing new compounds to promote the efficacy of oncolytic virus therapy.
- Published
- 2021
- Full Text
- View/download PDF
45. Immunotherapy in patients with cervical cancer
- Author
-
A. G. Kedrova
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Durvalumab ,cervical cancer ,medicine.medical_treatment ,Ipilimumab ,Pembrolizumab ,immune checkpoint inhibitors ,Avelumab ,03 medical and health sciences ,0302 clinical medicine ,Atezolizumab ,Internal medicine ,medicine ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging ,business.industry ,Obstetrics and Gynecology ,Gynecology and obstetrics ,Immunotherapy ,030104 developmental biology ,030220 oncology & carcinogenesis ,RG1-991 ,Oncolytic Virus Therapy ,Surgery ,immunotherapy ,pembrolizumab ,Nivolumab ,business ,medicine.drug - Abstract
Immunotherapy, also known as therapy with immune checkpoint inhibitors, has shown good results in the treatment of both solid tumors and hematological malignancies. Patients with diseases that were considered incurable earlier now have an opportunity for long-term disease stabilization and high frequency of clinical remissions. This review focuses on clinical benefits and toxicity profiles of immune checkpoint inhibitors used for cervical cancer, as well as on the ways to improve prognosis and indications for immunotherapy. Correct choice of biomarkers for predicting the response to immunotherapy will ensure more precise selection of patients. This review of immunotherapy methods aims to help clinicians with the indications for this relatively new treatment which has revolutionized treatment standards. Immunotherapy has many forms, including oncolytic virus therapy, chimeric antigen receptor T-cell therapy (CAR), cancer vaccines, and adoptive T-cell therapy, in particular, immune checkpoint inhibitors, first generation of which includes monoclonal antibodies against PD-1 (pembrolizumab, nivolumab, and cemiplimab), against PD-L1 (atezolizumab, avelumab, and durvalumab), and against CTLA-4 protein (ipilimumab).
- Published
- 2020
- Full Text
- View/download PDF
46. Membrane Derived Vesicles as Biomimetic Carriers for Targeted Drug Delivery System
- Author
-
Xue Yang, Hong-Ying Pan, Hong Chen, Shi-Bing Wang, Le-Yi Zhang, and Zhi-Ming Hu
- Subjects
Cell Survival ,Antineoplastic Agents ,02 engineering and technology ,Cell membrane ,Extracellular Vesicles ,03 medical and health sciences ,Drug Delivery Systems ,Biomimetic Materials ,Neoplasms ,Drug Discovery ,medicine ,Humans ,030304 developmental biology ,Drug Carriers ,0303 health sciences ,Photosensitizing Agents ,Chemistry ,Cell Membrane ,Lipid bilayer fusion ,General Medicine ,Photothermal therapy ,021001 nanoscience & nanotechnology ,Microvesicles ,Cell biology ,medicine.anatomical_structure ,Photochemotherapy ,Targeted drug delivery ,Drug delivery ,Tissue tropism ,Oncolytic Virus Therapy ,0210 nano-technology - Abstract
Extracellular vesicles (EVs) are membrane vesicles (MVs) playing important roles in various cellular and molecular functions in cell-to-cell signaling and transmitting molecular signals to adjacent as well as distant cells. The preserved cell membrane characteristics in MVs derived from live cells, give them great potential in biological applications. EVs are nanoscale particulates secreted from living cells and play crucial roles in several important cellular functions both in physiological and pathological states. EVs are the main elements in intercellular communication in which they serve as carriers for various endogenous cargo molecules, such as RNAs, proteins, carbohydrates, and lipids. High tissue tropism capacity that can be conveniently mediated by surface molecules, such as integrins and glycans, is a unique feature of EVs that makes them interesting candidates for targeted drug delivery systems. The cell-derived giant MVs have been exploited as vehicles for delivery of various anticancer agents and imaging probes and for implementing combinational phototherapy for targeted cancer treatment. Giant MVs can efficiently encapsulate therapeutic drugs and deliver them to target cells through the membrane fusion process to synergize photodynamic/photothermal treatment under light exposure. EVs can load diagnostic or therapeutic agents using different encapsulation or conjugation methods. Moreover, to prolong the blood circulation and enhance the targeting of the loaded agents, a variety of modification strategies can be exploited. This paper reviews the EVs-based drug delivery strategies in cancer therapy. Biological, pharmacokinetics and physicochemical characteristics, isolation techniques, engineering, and drug loading strategies of EVs are discussed. The recent preclinical and clinical progresses in applications of EVs and oncolytic virus therapy based on EVs, the clinical challenges and perspectives are discussed.
- Published
- 2020
- Full Text
- View/download PDF
47. The recombinant Newcastle disease virus Anhinga strain expressing human TRAIL exhibit antitumor effects on a glioma nude mice model
- Author
-
Qingzhong Yu, Lin Cui, Ying An, Jianying Qi, Yunzhou Wu, Jiechao Yin, Jinjiao He, Kai Yang, Xiaohui Jing, Mingyao Liu, Hui Dong, Bo Qu, Shijun Yan, and Deshan Li
- Subjects
viruses ,Newcastle disease virus ,Mice, Nude ,Apoptosis ,Virus Replication ,Recombinant virus ,Newcastle disease ,Virus ,law.invention ,TNF-Related Apoptosis-Inducing Ligand ,Mice ,03 medical and health sciences ,0302 clinical medicine ,law ,Cell Line, Tumor ,Virology ,Animals ,Humans ,Cytotoxic T cell ,030212 general & internal medicine ,Oncolytic Virotherapy ,biology ,Glioma ,biology.organism_classification ,Oncolytic virus ,Disease Models, Animal ,Oncolytic Viruses ,Infectious Diseases ,Recombinant DNA ,Oncolytic Virus Therapy ,Female ,030211 gastroenterology & hepatology - Abstract
Oncolytic virus therapy is perhaps the next major breakthrough in cancer treatment following the success in immunotherapy using immune checkpoint inhibitors. However, the potential oncolytic ability of the recombinant newcastle disease virus (NDV) Anhinga strain carried with tumor necrosis factor-related apoptosis inducing ligand (TRAIL) has not been fully explored at present. In the present study, the recombinant NDV/Anh-TRAIL that secretes soluble TRAIL was constructed and the experiment results suggested NDV/Anh-TRAIL as a promising candidate for glioma therapy. Growth kinetic and TRAIL secreted quantity of recombinant NDV/Anh-TRAIL virus were measured. Cytotoxic and cell apoptosis were analyzed for its anti-glioma therapy in vitro. Nude mice were used for the in vivo evaluation. Both tumor volume and mice behavior after injection were observed. The recombinant virus replicated with the same kinetics as the parental virus and the highest expression of TRAIL (77.8 ng/L) was found at 48 hours. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, a tetrazole and flow cytometry data revealed that the recombinant NDV/Anh-TRAIL (56.1 ± 8.2%) virus could induce a more severe apoptosis rate, when compared with the NDV wild type (37.2 ± 7.0%) and mock (7.0 ± 1.8%) groups (P < .01), in U251 cells. Furthermore, in the present animal study, the average tumor volume was smaller in the NDV/Anh-TRAIL group (97.21 mm3 ), when compared with the NDV wild type (205.03 mm3 , P < .05) and PBS (310.30 mm3 , P < .01) groups.
- Published
- 2020
- Full Text
- View/download PDF
48. Oncolytic virus combined with traditional treatment versus traditional treatment alone in patients with cancer: a meta-analysis
- Author
-
Tianyu Tang, Wei Song, Xueli Bai, Zifan Yang, Tingbo Liang, Xiaozhen Zhang, Meng Wang, Yuwei Li, Zengwei Tang, and Yinan Shen
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Neutropenia ,Combination therapy ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Internal medicine ,medicine ,Humans ,Adverse effect ,Survival rate ,Fatigue ,Oncolytic Virotherapy ,business.industry ,Cancer ,Anemia ,Hematology ,General Medicine ,Odds ratio ,medicine.disease ,Combined Modality Therapy ,Oncolytic virus ,Survival Rate ,Oncolytic Viruses ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Meta-analysis ,Oncolytic Virus Therapy ,Surgery ,business - Abstract
Oncolytic virus therapy has shown benefits for multiple cancers, while limitations remain for traditional treatment. However, few studies have concentrated on comparing whether oncolytic virus combined with traditional treatment is better than traditional treatment alone in patients with cancer. We conducted a meta-analysis of the curative effect and safety of oncolytic virus combination therapy. We searched the PubMed, Embase, Cochrane Library, and Web of Science databases comprehensively for articles comparing oncolytic virus combined with traditional treatment to traditional treatment alone in patients with cancer. A meta-analysis and trial sequential analysis were performed. A total of 12 studies involving 1494 patients (combination therapy group, 820 patients; traditional treatment group, 674 patients) were included in the study. Compared with traditional treatment alone, combination therapy was significantly associated with high objective response rate [odds ratio (OR) 1.35, 95% confidence interval (CI) 1.01–1.82, p = 0.04]. There were no significant differences for other outcomes such as 1- and 2-year survival rate, and 4- and 12-month progression-free survival rate. Combination therapy was significantly associated with high incidence of grade ≥ 3 adverse effects (OR 1.47, 95% CI 1.06–2.05, p = 0.02) and high incidence of grade ≥ 3 neutropenia (OR 1.65, 95% CI 1.13–2.43, p = 0.01). There were no significant differences for other grade ≥ 3 adverse effects, e.g., gastrointestinal adverse effects, influenza-like illness, fatigue, anemia, and thrombocytopenia. Despite partially increased toxicity, the combination therapy improves the effectiveness of cancer treatment. However, high-quality, large-scale studies are needed to evaluate its effectiveness and safety.
- Published
- 2020
- Full Text
- View/download PDF
49. Targeted Metabolic Reprogramming to Improve the Efficacy of Oncolytic Virus Therapy
- Author
-
Barry E. Kennedy, Shashi Gujar, and Maryanne Sadek
- Subjects
Genetic Vectors ,Review ,Biology ,Oxidative Phosphorylation ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Neoplasms ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Glycolysis ,Molecular Biology ,030304 developmental biology ,Oncolytic Virotherapy ,Pharmacology ,0303 health sciences ,Tumor microenvironment ,Cancer ,Genetic Therapy ,Cellular Reprogramming ,medicine.disease ,Oncolytic virus ,Oncolytic Viruses ,Metabolic pathway ,Treatment Outcome ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Molecular Medicine ,Oncolytic Virus Therapy ,Energy Metabolism ,Metabolic Networks and Pathways - Abstract
Oncolytic viruses (OVs) represent a promising new class of cancer therapeutics and cause antitumor effects by two major mechanisms: (1) directly killing cancer cells in a process known as oncolysis, or (2) initiating a powerful antitumor immune response. Interestingly, energy metabolism, within either cancer cells or immune cells, plays a pivotal role in defining the outcome of OV-mediated antitumor effects. Following therapeutic administration, OVs must hijack host cell metabolic pathways to acquire building blocks such as nucleotides, lipids, and amino acids for the process of replication that is necessary for oncolysis. Additionally, OV-stimulated antitumor immune responses are highly dependent on the metabolic state within the tumor microenvironment. Thus, metabolic reprogramming strategies bear the potential to enhance the efficacy of both OV-mediated oncolysis and antitumor immune responses.
- Published
- 2020
- Full Text
- View/download PDF
50. Combination immunotherapy of oncolytic virus nanovesicles and PD-1 blockade effectively enhances therapeutic effects and boosts antitumour immune response
- Author
-
Wei Ji, Lihua Qiu, Lanfang Li, Zhengzi Qian, Shiyong Zhou, Huilai Zhang, and Peiqi Zhao
- Subjects
Skin Neoplasms ,Immune checkpoint inhibitors ,Programmed Cell Death 1 Receptor ,Pharmaceutical Science ,02 engineering and technology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Combination immunotherapy ,skin and connective tissue diseases ,Melanoma ,neoplasms ,Oncolytic Virotherapy ,business.industry ,Therapeutic effect ,021001 nanoscience & nanotechnology ,medicine.disease ,Combined Modality Therapy ,Oncolytic virus ,Oncolytic Viruses ,030220 oncology & carcinogenesis ,Cancer research ,Nanoparticles ,Pd 1 blockade ,Oncolytic Virus Therapy ,sense organs ,0210 nano-technology ,business - Abstract
Immunotherapies are changing the landscape of melanoma treatment, but 70% of the melanoma patients have no response to immune checkpoint inhibitors or oncolytic virus therapy. Thus, novel formulations are needed to improve the population benefiting from immunotherapy. Here, we report a combined therapeutic modality based on oncolytic virus nanovesicles composed of CaCl
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.