Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.160
Filter
1.
Oncoimmunology ; 13(1): 2407532, 2024.
Article in English | MEDLINE | ID: mdl-39351443

ABSTRACT

Immunotherapy has emerged as a promising approach for cancer treatment, with oncolytic adenoviruses showing power as immunotherapeutic agents. In this study, we investigated the immunotherapeutic potential of an adenovirus construct expressing CXCL9, CXCL10, or IL-15 in clear cell renal cell carcinoma (ccRCC) tumor models. Our results demonstrated robust cytokine secretion upon viral treatment, suggesting effective transgene expression. Subsequent analysis using resistance-based transwell migration and microfluidic chip assays demonstrated increased T-cell migration in response to chemokine secretion by infected cells in both 2D and 3D cell models. Flow cytometry analysis revealed CXCR3 receptor expression across T-cell subsets, with the highest percentage found on CD8+ T-cells, underscoring their key role in immune cell migration. Alongside T-cells, we also detected NK-cells in the tumors of immunocompromised mice treated with cytokine-encoding adenoviruses. Furthermore, we identified potential immunogenic antigens that may enhance the efficacy and specificity of our armed oncolytic adenoviruses in ccRCC. Overall, our findings using ccRCC cell line, in vivo humanized mice, physiologically relevant PDCs in 2D and patient-derived organoids (PDOs) in 3D suggest that chemokine-armed adenoviruses hold promise for enhancing T-cell migration and improving immunotherapy outcomes in ccRCC. Our study contributes to the development of more effective ccRCC treatment strategies by elucidating immune cell infiltration and activation mechanisms within the tumor microenvironment (TME) and highlights the usefulness of PDOs for predicting clinical relevance and validating novel immunotherapeutic approaches. Overall, our research offers insights into the rational design and optimization of viral-based immunotherapies for ccRCC.


Subject(s)
Adenoviridae , Carcinoma, Renal Cell , Kidney Neoplasms , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/genetics , Humans , Animals , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Mice , Adenoviridae/genetics , Adenoviridae/immunology , Cell Line, Tumor , Xenograft Model Antitumor Assays , Oncolytic Virotherapy/methods , Immunotherapy/methods , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Chemokine CXCL9/immunology , Cell Movement , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL10/immunology , Cytokines/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Interleukin-15/genetics , Interleukin-15/metabolism , Interleukin-15/immunology , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , CD8-Positive T-Lymphocytes/immunology
2.
Virol J ; 21(1): 245, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39369233

ABSTRACT

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


Subject(s)
Carcinoma, Squamous Cell , Enterovirus B, Human , Oncolytic Virotherapy , Oncolytic Viruses , Organoids , Paclitaxel , Uterine Cervical Neoplasms , Xenograft Model Antitumor Assays , Humans , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/therapy , Uterine Cervical Neoplasms/drug therapy , Animals , Female , Organoids/virology , Mice , Enterovirus B, Human/physiology , Enterovirus B, Human/drug effects , Oncolytic Virotherapy/methods , Carcinoma, Squamous Cell/virology , Carcinoma, Squamous Cell/therapy , Carcinoma, Squamous Cell/drug therapy , Oncolytic Viruses/physiology , Cell Line, Tumor , Virus Replication/drug effects
3.
Cancer Immunol Immunother ; 73(12): 243, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39358654

ABSTRACT

The hemagglutinating virus of Japan envelope (HVJ-E) is an inactivated Sendai virus particle with antitumor effect and inducing antitumor immunity. However, its dosage and efficacy have not been verified. We conducted a phase I clinical study on chemotherapy-resistant malignant pleural mesothelioma (MPM) aiming to determine the recommended dosage for a phase II study through dose-limiting toxicity and evaluate HVJ-E's preliminary efficacy. HVJ-E was administered intratumorally and subcutaneously to the patients with chemotherapy-resistant MPM. While no serious adverse events occurred, known adverse events of HVJ-E were observed. In the preliminary antitumor efficacy using modified response evaluation criteria in solid tumors (RECIST) criteria, three low-dose patients exhibited progressive disease, while all high-dose patients achieved stable disease, yielding disease control rates (DCRs) of 0% and 100%, respectively. Furthermore, the dose-dependent effect of HVJ-E revealed on DCR modified by RECIST and the baseline changes in target lesion size (by CT and SUL-peak; p < 0.05). Comparing targeted lesions receiving intratumoral HVJ-E with non-injected ones, while no clear difference existed at the end of the study, follow-up cases suggested stronger antitumor effects with intratumoral administration. Our findings suggest that HVJ-E could be safely administered to patients with chemotherapy-resistant MPM at both study doses. HVJ-E exhibited some antitumor activity against chemotherapy-resistant MPM, and higher doses tended to have stronger antitumor effects than lower doses. Consequently, a phase II clinical trial with higher HVJ-E doses has been conducted for MPM treatment. Trial registration number: UMIN Clinical Trials Registry (#UMIN000019345).


Subject(s)
Drug Resistance, Neoplasm , Mesothelioma, Malignant , Pleural Neoplasms , Sendai virus , Humans , Male , Middle Aged , Female , Aged , Mesothelioma, Malignant/drug therapy , Mesothelioma, Malignant/pathology , Pleural Neoplasms/drug therapy , Injections, Subcutaneous , Oncolytic Virotherapy/methods , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Injections, Intralesional , Viral Envelope Proteins
4.
Sci Rep ; 14(1): 23671, 2024 10 10.
Article in English | MEDLINE | ID: mdl-39389985

ABSTRACT

Oncolytic virotherapy is emerging as a promising therapeutic avenue for cancer treatment, harnessing both innate and tumor-specific immune responses for targeted tumor elimination. In this study, we present a novel oncolytic virus (oHSV1-IL15B) derived from herpes simplex virus-1 (HSV-1), armed with IL-15/IL-15Rα complex, with a focus on treating colon cancer combined with oncolytic HSV-1 expressing anti-PD-1 antibody (oHSV1-aPD1). Results from our study reveal that recombinant oHSV-1 virus equipped with IL-15/IL-15Rα complex exhibited significant anti-tumor effects in a murine CT26 colon adenocarcinoma model. Notably, oHSV1-IL15B combined with oHSV-1-aPD1 demonstrates superior tumor inhibition and prolonged overall survival compared to oHSV1-mock and monotherapy groups. Further exploration highlights the impact of oHSV1-IL15B, oHSV-1-aPD1 and combined group on antitumor capacity, revealing a substantial increase in CD8+ T and CD4+ T cell proportions of CT26-bearing BALB/c mice and promoting apoptosis in tumor tissue. The study emphasizes the pivotal role of cytotoxic CD8+T cells in oncolytic virotherapy, demonstrating that recombinant oHSV1-IL15B combined with oncolytic HSV-1-aPD1 induces a robust tumor-specific T cell response. RNA sequence analysis highlighted oHSV1-IL15B combined with oHSV1-aPD1 improved tumors immune microenvironment on immune response, antiviral response-related genes and apoptosis-related genes, which contributed to anti-tumor immunotherapy. The findings underscore the promising antitumor activity achieved through the combination of IL-15/IL-15Rα complex and anti-PD-1 antibody with oHSV-1. This research opens avenues for diverse therapeutic strategies, suggesting the potential of synergistically utilizing cytokines and anti-PD-1 antibody with oncolytic viruses to enhance immunotherapy for cancer management.


Subject(s)
Colonic Neoplasms , Herpesvirus 1, Human , Interleukin-15 , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Herpesvirus 1, Human/genetics , Interleukin-15/genetics , Interleukin-15/immunology , Colonic Neoplasms/therapy , Colonic Neoplasms/immunology , Oncolytic Virotherapy/methods , Mice , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Cell Line, Tumor , Mice, Inbred BALB C , Humans , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/genetics , Interleukin-15 Receptor alpha Subunit/genetics , Female
5.
BMC Cancer ; 24(1): 1253, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39390431

ABSTRACT

BACKGROUND AND AIMS: Cervical cancer (CC) is a common cancer among women, often treated with Doxorubicin (Doxo). Research is underway to explore the use of oncolytic virus (OV) therapy as a means to improve drug efficacy and enhance the immune system's tumor-fighting capabilities. Hence, our study purposes to evaluate the therapeutic potential of Newcastle disease virus (NDV) in increasing the antitumor efficacy of Doxo in mouse models of CC. METHODS AND MATERIALS: TC1 cells were administered to C57BL/6 mice (Female) in a range of 6 to 8 weeks age (n = 40) to induce tumor growth. After tumor development, four treatment groups of mice were formed. Treatment were performed through NDV, Doxo, and a combination of both in three groups of treatment twice in a one-week intervention manner, while the control group treated with PBS. Following the last treatment, half of these mice were subjected to euthanize due to the immune-response assessment, and the other half were followed up till they died naturally in a certain period of time. RESULTS: Mice that underwent the combined treatment showed significantly improved survival rates and slower tumor progression in comparison with the control group. This combined treatment substantially elevated nitric oxide (NO) and lactate dehydrogenase (LDH) levels in the splenocytes cultures of mice bearing cervical tumors. Furthermore, combination therapy resulted in a notable elevation in TNF-α, IL-12, and IFN-γ, secretion alongside a reduction in the release of TGF-ß and IL-4 within the splenocytes in counter with the treatment of just NDV or Doxo. CONCLUSION: According to the findings of this study, it seems that utilizing NDV can improve the effectiveness of Doxo in a mouse model of CC, suggesting it can serve as an adjunct therapy alongside chemotherapy.


Subject(s)
Disease Models, Animal , Doxorubicin , Newcastle disease virus , Oncolytic Virotherapy , Uterine Cervical Neoplasms , Animals , Female , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/therapy , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Doxorubicin/therapeutic use , Doxorubicin/pharmacology , Mice , Oncolytic Virotherapy/methods , Combined Modality Therapy , Mice, Inbred C57BL , Cell Line, Tumor , Antibiotics, Antineoplastic/therapeutic use , Antibiotics, Antineoplastic/pharmacology , Humans , Oncolytic Viruses
6.
Asian Pac J Cancer Prev ; 25(9): 2991-2998, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39342575

ABSTRACT

OBJECTIVE: Leukemia is a group of hematologic malignancies in the bonemarrow that arise from the dysfunctional proliferation of developing leukocytes. It is classified as either acute or chronic based on the rapidity of proliferation and as myelocytic or lymphocytic based on the cell of origin. Newcastle Disease Virus (NDV) is an avian paramyxovirus, which has been demonstrated to possess significant oncolytic activity against mammalian cancers because its ability to kill tumor cells with limited toxicity to normal cells. METHODS:  In this study, the morphophical changes and apoptosis induction of WEHI 3B leukemia cell line treated with NDV strain AF2240 were studied by scanning electron microscopes and  transmission electron microscopes techniques. RESULT: Electron microscopy indicated that NDV strain AF 2240 significantly altered cell morphology and reduced cell viability. Furthermore,  early apoptosis was observed 6 h post-inoculation by fluorescence microscope. CONCLUSION: Our results suggest that NDV has ability to induce significant apoptoic structural changes in WEHI 3B leukemia cell line. These findings provide new insights into the mechanism of action of NDV virotherapy and could lead to the development of more effective treatments for leukemia.


Subject(s)
Apoptosis , Newcastle disease virus , Animals , Oncolytic Virotherapy/methods , Humans , Tumor Cells, Cultured , Mice , Microscopy, Electron, Transmission , Cell Proliferation , Cell Line, Tumor
7.
Asian Pac J Cancer Prev ; 25(9): 3111-3118, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39342590

ABSTRACT

OBJECTIVE: We hypothesized that attacking cancer cells by combining various modes of action can hinder them from taking the chance to evolve resistance to treatment. Incorporation of photodynamic therapy (PDT) with oncolytic virotherapy might be a promising dual approach to cancer treatment. METHODS: NDV AMHA1 strain as virotherapy in integration with aminolaevulinic acid (ALA) using low power He-Ne laser as PDT in the existing work was examined against breast cancer cells derived from Iraqi cancer patients named (AMJ13). This combination was evaluated using Chou-Talalay analysis. RESULTS: The results showed an increased killing rate when using both 0.01 and 0.1 Multiplicity of infection (MOI) of the virus when combined with a dose of 6172.8 photons/gm (ph/gm) of PDT focused on cancer cells. CONCLUSION: integration of the attenuated NDV-AMHA1 strain with photodynamic therapy has a synergistic killing effect on breast cancer cells in vitro, suggesting that this strategy could have clinical application to overcome breast cancer.


Subject(s)
Breast Neoplasms , Newcastle disease virus , Oncolytic Virotherapy , Photochemotherapy , Photosensitizing Agents , Humans , Photochemotherapy/methods , Breast Neoplasms/therapy , Breast Neoplasms/drug therapy , Female , Oncolytic Virotherapy/methods , Photosensitizing Agents/therapeutic use , Oncolytic Viruses , Tumor Cells, Cultured , Aminolevulinic Acid/therapeutic use , Aminolevulinic Acid/pharmacology , Combined Modality Therapy
8.
J Transl Med ; 22(1): 862, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334370

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most frequent and aggressive brain tumor in adults with the lowest survival rates five years post-diagnosis. Oncolytic viruses (OVs) selectively target and damage cancer cells, and for this reason they are being investigated as new therapeutic tools also against GBM. METHODS: An oncolytic herpes simplex virus type 1 (oHSV-1) with deletions in the γ34.5 neurovirulence gene and the US12 gene, expressing enhanced green fluorescent protein (EGFP-oHSV-1) as reporter gene was generated and tested for its capacity to infect and kill the murine GL261 glioblastoma (GBM) cell line. Syngeneic mice were orthotopically injected with GL261cells. Seven days post-implantation, EGFP-oHSV-1 was administered intratumorally. Twenty-one days after parental tumor challenge in the opposite brain hemisphere, mice were sacrified and their brains were analysed by immunohistochemistry to assess tumor presence and cell infiltrate. RESULTS: oHSV-1 replicates and induces cell death of GL261 cells in vitro. A single intracranial injection of EGFP-oHSV-1 in established GL261 tumors significantly prolongs survival in all treated mice compared to placebo treatment. Notably, 45% of treated mice became long-term survivors, and rejected GL261 cells upon rechallenge in the contralateral brain hemisphere, indicating an anamnestic antitumoral immune response. Post-mortem analysis revealed a profound modification of the tumor microenvironment with increased infiltration of CD4 + and CD8 + T lymphocytes, intertumoral vascular collapse and activation and redistribution of macrophage, microglia, and astroglia in the tumor area, with the formation of intense fibrotic tissue suggestive of complete rejection in long-term survivor mice. CONCLUSIONS: EGFP-oHSV1 demonstrates potent antitumoral activity in an immunocompetent GBM model as a monotherapy, resulting from direct cell killing combined with the stimulation of a protective adaptive immune response. These results open the way to possible application of our strategy in clinical setting.


Subject(s)
Adaptive Immunity , Glioblastoma , Herpesvirus 1, Human , Oncolytic Virotherapy , Animals , Glioblastoma/therapy , Glioblastoma/immunology , Glioblastoma/pathology , Cell Line, Tumor , Oncolytic Virotherapy/methods , Genetic Vectors , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Oncolytic Viruses/genetics , Mice, Inbred C57BL , Green Fluorescent Proteins/metabolism , Mice , Humans
9.
Hereditas ; 161(1): 36, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342391

ABSTRACT

BACKGROUND: The therapeutic potential of oncolytic measles virotherapy has been demonstrated across various malignancies. However, the effectiveness against human breast cancer (BC) and the underlying mechanisms of the recombinant measles virus vaccine strain Hu191 (rMeV-Hu191) remain unclear. METHODS: We utilized a range of methods, including cell viability assay, Western blot, flow cytometry, immunofluorescence, SA-ß-gal staining, reverse transcription quantitative real-time PCR, transcriptome sequencing, BC xenograft mouse models, and immunohistochemistry to evaluate the antitumor efficacy of rMeV-Hu191 against BC and elucidate the underlying mechanism. Additionally, we employed transcriptomics and gene set enrichment analysis to analyze the lipid metabolism status of BC cells following rMeV-Hu191 infection. RESULTS: Our study revealed the multifaceted antitumor effects of rMeV-Hu191 against BC. rMeV-Hu191 induced apoptosis, inhibited proliferation, and promoted senescence in BC cells. Furthermore, rMeV-Hu191 was associated with changes in oxidative stress and lipid homeostasis in infected BC cells. In vivo, studies using a BC xenograft mouse model confirmed a significant reduction in tumor growth following local injection of rMeV-Hu191. CONCLUSIONS: The findings highlight the potential of rMeV-Hu191 as a promising treatment for BC and provide valuable insights into the mechanisms underlying its oncolytic effect.


Subject(s)
Breast Neoplasms , Measles virus , Oncolytic Virotherapy , Animals , Breast Neoplasms/therapy , Breast Neoplasms/genetics , Humans , Mice , Female , Oncolytic Virotherapy/methods , Cell Line, Tumor , Measles virus/genetics , Xenograft Model Antitumor Assays , Apoptosis , Cell Proliferation , Measles Vaccine , Oncolytic Viruses/genetics , Cell Survival
10.
Genes (Basel) ; 15(9)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39336785

ABSTRACT

Oncolytic viruses (OVs) are promising cancer immunotherapy agents that stimulate anti-tumor immunity through the preferential infection and killing of tumor cells. OVs are currently under limited clinical usage, due in part to their restricted efficacy as monotherapies. Current efforts for enhancement of the therapeutic potency of OVs involve their combination with other therapy modalities, aiming at the concomitant exploitation of complementary tumor weaknesses. In this context, microtubule-targeting agents (MTAs) pose as an enticing option, as they perturb microtubule dynamics and function, induce cell-cycle arrest, and cause mitotic cell death. MTAs induce therapeutic benefit through cancer-cell-autonomous and non-cell-autonomous mechanisms and are a main component of the standard of care for different malignancies. However, off-target effects and acquired resistance involving distinct cellular and molecular mechanisms may limit the overall efficacy of MTA-based therapy. When combined, OVs and MTAs may enhance therapeutic efficacy through increases in OV infection and immunogenic cell death and a decreased probability of acquired resistance. In this review, we introduce OVs and MTAs, describe molecular features of their activity in cancer cells, and discuss studies and clinical trials in which the combination has been tested.


Subject(s)
Microtubules , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Oncolytic Virotherapy/methods , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/drug therapy , Oncolytic Viruses/genetics , Animals , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , Immunotherapy/methods , Combined Modality Therapy
11.
Int J Mol Sci ; 25(18)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39337402

ABSTRACT

Pancreatic cancer presents formidable challenges due to rapid progression and resistance to conventional treatments. Oncolytic viruses (OVs) selectively infect cancer cells and cause cancer cells to lyse, releasing molecules that can be identified by the host's immune system. Moreover, OV can carry immune-stimulatory payloads such as interleukin-12, which when delivered locally can enhance immune system-mediated tumor killing. OVs are very well tolerated by cancer patients due to their ability to selectively target tumors without affecting surrounding normal tissues. OVs have recently been combined with other therapies, including chemotherapy and immunotherapy, to improve clinical outcomes. Several OVs including adenovirus, herpes simplex viruses (HSVs), vaccinia virus, parvovirus, reovirus, and measles virus have been evaluated in preclinical and clinical settings for the treatment of pancreatic cancer. We evaluated the safety and tolerability of a replication-competent oncolytic adenoviral vector carrying two suicide genes (thymidine kinase, TK; and cytosine deaminase, CD) and human interleukin-12 (hIL12) in metastatic pancreatic cancer patients in a phase 1 trial. This vector was found to be safe and well-tolerated at the highest doses tested without causing any significant adverse events (SAEs). Moreover, long-term follow-up studies indicated an increase in the overall survival (OS) in subjects receiving the highest dose of the OV. Our encouraging long-term survival data provide hope for patients with advanced pancreatic cancer, a disease that has not seen a meaningful increase in OS in the last five decades. In this review article, we highlight several preclinical and clinical studies and discuss future directions for optimizing OV therapy in pancreatic cancer. We envision OV-based gene therapy to be a game changer in the near future with the advent of newer generation OVs that have higher specificity and selectivity combined with personalized treatment plans developed under AI guidance.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/immunology , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Oncolytic Virotherapy/methods , Animals , Immunotherapy/methods , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Interleukin-12/genetics , Combined Modality Therapy
12.
Cancer Immunol Immunother ; 73(11): 221, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39235531

ABSTRACT

BACKGROUND: Neuroblastoma is the most common extracranial solid tumor in children and accounts for 15% of pediatric cancer related deaths. Targeting neuroblastoma with immunotherapies has proven challenging due to a paucity of immune cells in the tumor microenvironment and the release of immunosuppressive cytokines by neuroblastoma tumor cells. We hypothesized that combining an oncolytic Herpes Simplex Virus (oHSV) with natural killer (NK) cells might overcome these barriers and incite tumor cell death. METHODS: We utilized MYCN amplified and non-amplified neuroblastoma cell lines, the IL-12 expressing oHSV, M002, and the human NK cell line, NK-92 MI. We assessed the cytotoxicity of NK cells against neuroblastoma with and without M002 infection, the effects of M002 on NK cell priming, and the impact of M002 and priming on the migratory capacity and CD107a expression of NK cells. To test clinical applicability, we then investigated the effects of M002 and NK cells on neuroblastoma in vivo. RESULTS: NK cells were more attracted to neuroblastoma cells that were infected with M002. There was an increase in neuroblastoma cell death with the combination treatment of M002 and NK cells both in vitro and in vivo. Priming the NK cells enhanced their cytotoxicity, migratory capacity and CD107a expression. CONCLUSIONS: To the best of our knowledge, these investigations are the first to demonstrate the effects of an oncolytic virus combined with self-maintaining NK cells in neuroblastoma and the priming effect of neuroblastoma on NK cells. The current studies provide a deeper understanding of the relation between NK cells and neuroblastoma and these data suggest that oHSV increases NK cell cytotoxicity towards neuroblastoma.


Subject(s)
Killer Cells, Natural , Neuroblastoma , Oncolytic Virotherapy , Neuroblastoma/therapy , Neuroblastoma/immunology , Killer Cells, Natural/immunology , Humans , Oncolytic Virotherapy/methods , Animals , Mice , Cell Line, Tumor , Oncolytic Viruses/immunology , Cytotoxicity, Immunologic , Simplexvirus/immunology , Xenograft Model Antitumor Assays
13.
Front Immunol ; 15: 1433315, 2024.
Article in English | MEDLINE | ID: mdl-39238638

ABSTRACT

Colorectal cancer is the third most common cancer and the second most lethal cancer in the world. The main cause of the disease is due to dietary and behavioral factors. The treatment of this complex disease is mainly based on traditional treatments, including surgery, radiotherapy, and chemotherapy. Due to its high prevalence and high morbidity, more effective treatments with fewer side effects are urgently needed. In recent years, immunotherapy has become a potential therapeutic alternative and one of the fastest-developing treatments. Immunotherapy inhibits tumor growth by activating or enhancing the immune system to recognize and attack cancer cells. This review presents the latest immunotherapies for immune checkpoint inhibitors, cell therapy, tumor-infiltrating lymphocytes, and oncolytic viruses. Some of these have shown promising results in clinical trials and are used in clinical treatment.


Subject(s)
Colorectal Neoplasms , Immune Checkpoint Inhibitors , Immunotherapy , Humans , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use , Animals , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology
14.
Commun Biol ; 7(1): 1132, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271928

ABSTRACT

ICVB-1042 is an oncolytic adenovirus containing modifications to enhance replication, lysis, and viral spreading in tumor cells. The anti-tumor activity, immune activation, tropism, selectivity, and mechanism of action were evaluated in preparation for a first-in-human study. ICVB-1042 was at least 100-fold more cytotoxic in A549 cells than in normal primary cells tested, demonstrating its high tumor selectivity and a low likelihood of targeting primary tissues. ICVB-1042 administered to mice intravenously or intratumorally was effective in reducing tumor burden. Its intravenous administration also inhibited tumor growth in orthotopic models. ICVB-1042 was well tolerated in mice compared to HAdV-C5 (Wt Ad5), with reduced liver sequestration, supporting safety of the drug for systemic delivery. These preclinical data demonstrating the safety and potency of ICVB-1042 for treatment of various solid tumors support the ongoing clinical investigation (NCT05904236).


Subject(s)
Adenoviridae , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Humans , Oncolytic Virotherapy/methods , Mice , Oncolytic Viruses/genetics , Neoplasms/therapy , Adenoviridae/genetics , Xenograft Model Antitumor Assays , Female , Cell Line, Tumor , A549 Cells , Virus Replication , Mice, Nude
15.
Oncotarget ; 15: 662-673, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39347716

ABSTRACT

INTRODUCTION: Many studies have highlighted the use of oncolytic viruses as a new class of therapeutic agents for central nervous system (CNS) tumors, especially glioblastomas (GMB). Zika Virus (ZIKV) proteins targeted to specific stem cells have been studied in vitro and animal models with promising results. MATERIALS AND METHODS: A systematic review was evaluated the efficacy and safety of the ZIKV use for CNS tumors treatment. Data were extracted and the in vivo studies were evaluated using the Robins-I tool. We assessed bias in each study using criteria such as selection bias, performance bias, detection bias, attrition bias, reporting bias, and others. According to Cochrane guidelines, bias was classified as high, low, or uncertain. High bias occurred when studies did not meet the criteria. Low bias was assigned when criteria were clearly met. Uncertain bias reflected insufficient information for a clear classification. RESULTS: The 14 included studies shown that ZIKV reduced cell viability or inhibited the growth, proliferation of glioma stem cells (GSCs), and Bcl2 expression - which could potentially enhance the effect of chemotherapy/radiotherapy; caused cytopathic effects, induced tumor cell damage, manifested oncolytic properties, and even selectively safely killed GSCs; ultimately, it led to significant tumor remission and enhanced long-term survival through enhanced T-cell response. CONCLUSIONS: Although current evidence suggests ZIKV as a promising treatment for CNS tumors and may improve survival when combined with surgery and radiotherapy. Despite limited human evidence, it shows potential benefits. Further research is needed to confirm safety, efficacy, and optimize treatment in humans.


Subject(s)
Brain Neoplasms , Oncolytic Virotherapy , Zika Virus Infection , Zika Virus , Humans , Brain Neoplasms/therapy , Brain Neoplasms/virology , Animals , Oncolytic Virotherapy/methods , Zika Virus Infection/therapy , Zika Virus Infection/virology , Neoplastic Stem Cells/virology , Oncolytic Viruses , Glioblastoma/therapy , Glioblastoma/virology , Cell Proliferation
16.
Neoplasia ; 57: 101056, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39276533

ABSTRACT

Single agent immune checkpoint inhibitors have been ineffective for patients with advanced stage and recurrent high grade serous ovarian cancer (HGSOC). Using pre-clinical models of HGSOC, we evaluated the anti-tumor and immune stimulatory effects of an oncolytic adenovirus, MEM-288. This conditionally replicative virus encodes a modified membrane stable CD40L and IFNß. We demonstrated this virus successfully infects HGSOC cell lines and primary human ascites samples in vitro. We evaluated the anti-tumor and immunostimulatory activity in vivo in immune competent mouse models. Intraperitoneal delivery of MEM-288 decreased ascites and solid tumor burden compared to controls, and treatment generated a systemic anti-tumor immune response. The tumor microenvironment had a higher proportion of anti-tumor macrophages and decreased markers of angiogenesis. MEM-288 is a promising immunotherapy agent in HGSOC, with further pre-clinical studies required to understand the mechanism of action in the peritoneal microenvironment and clinical activity in combination with other therapies.


Subject(s)
Adenoviridae , CD40 Ligand , Cystadenocarcinoma, Serous , Interferon-beta , Oncolytic Virotherapy , Oncolytic Viruses , Ovarian Neoplasms , Tumor Microenvironment , Xenograft Model Antitumor Assays , Female , Humans , Animals , Ovarian Neoplasms/therapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Mice , Adenoviridae/genetics , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Interferon-beta/genetics , Interferon-beta/metabolism , Oncolytic Virotherapy/methods , Tumor Microenvironment/immunology , CD40 Ligand/genetics , CD40 Ligand/metabolism , Cell Line, Tumor , Cystadenocarcinoma, Serous/therapy , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Disease Models, Animal , Neoplasm Grading , Immunotherapy/methods
17.
Mol Biomed ; 5(1): 39, 2024 09 22.
Article in English | MEDLINE | ID: mdl-39306655

ABSTRACT

Colorectal cancer (CRC) is the second common cause of cancer mortality worldwide, and it still lacks effective approaches for relapsed and metastatic CRC. Recently, oncolytic virus has been emerged as a promising immune therapeutic strategy. In this study, we develop a novel oncolytic adenovirus, rAd.mDCN.mCD40L, which drive oncolytic activity by telomerase reverse transcriptase promoter (TERTp). rAd.mDCN.mCD40L expressed both mouse genes of decorin (mDCN) and CD40 ligand (mCD40L), and produced effective cytotoxicity in both human and mouse CRC cells. Moreover, oncolytic adenovirus mediated mDCN over-expression inhibited Met expression in vitro. In CT26 subcutaneous tumor model, intratumorally delivery of oncolytic adenoviruses could inhibit tumor growth and liver metastasis, while mDCN and/or mCD40L armed oncolytic adenoviruses produced much more impressive responses. No obvious toxicity was detected in lung, liver and spleen. Moreover, mDCN and/or mCD40L armed oncolytic adenoviruses altered the immune state to activate anti-tumor responses, including increasing CD8+ T effector cells and CD4+ memory T cells, reducing MDSCs and Tregs in peripheral blood. Furthermore, mDCN and/or mCD40L armed oncolytic adenoviruses mediated mDCN and/or mCD40L expression in tumors, and up-regulated Th1 cytokines and reduced Th2 cytokines in tumors, which will be benefit for remodeling tumor microenvironment. Importantly, rAd.mDCN.mCD40L and rAd.mCD40L prevented tumor liver metastasis much more effectively than rAd.Null and rAd.mDCN. Therefore, rAd.mDCN.mCD40L and rAd.mCD40L are promising approaches for CRC therapy.


Subject(s)
Adenoviridae , CD40 Ligand , Colorectal Neoplasms , Decorin , Liver Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Colorectal Neoplasms/therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/genetics , Decorin/genetics , Decorin/metabolism , Adenoviridae/genetics , Humans , Mice , Liver Neoplasms/therapy , Liver Neoplasms/secondary , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/genetics , CD40 Ligand/genetics , CD40 Ligand/metabolism , CD40 Ligand/immunology , Oncolytic Virotherapy/methods , Cell Line, Tumor , Oncolytic Viruses/genetics , Mice, Inbred BALB C , Disease Models, Animal
18.
Cells ; 13(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39329778

ABSTRACT

Lung cancer remains a formidable health challenge due to its high mortality and morbidity rates. Non-small cell lung cancer (NSCLC) constitutes approximately 85% of all lung cancer cases, with small cell lung cancer (SCLC) accounting for the remainder. Both NSCLC and SCLC cells express receptor tyrosine kinases, which may be overexpressed or mutated in lung cancer, leading to increased activation. The c-Met receptor tyrosine kinase, crucial for cell transformation and tumor growth, invasion, and metastasis, became the focus of our study. We used an E1B55KD-deleted, replication-selective oncolytic adenovirus (Ad.What), driven by the c-Met promoter, targeting lung cancer cells with c-Met overexpression, thus sparing normal cells. Previous studies have shown the enhanced antitumor efficacy of oncolytic adenoviruses when combined with chemotherapeutic agents. We explored combining rapamycin, a selective mTOR inhibitor with promising clinical trial outcomes for various cancers, with Ad.What. This combination increased infectivity by augmenting the expression of coxsackievirus and adenovirus receptors and αV integrin on cancer cells and induced autophagy. Our findings suggest that combining a c-Met promoter-driven oncolytic adenovirus with rapamycin could be an effective lung cancer treatment strategy, offering a targeted approach to exploit lung cancer cells' vulnerabilities, potentially marking a significant advancement in managing this deadly disease.


Subject(s)
Adenoviridae , Lung Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Promoter Regions, Genetic , Proto-Oncogene Proteins c-met , Sirolimus , Humans , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-met/genetics , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Lung Neoplasms/pathology , Promoter Regions, Genetic/genetics , Adenoviridae/genetics , Cell Line, Tumor , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Sirolimus/pharmacology , Sirolimus/therapeutic use , Animals , Mice , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/pathology
19.
J Control Release ; 374: 89-102, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39122217

ABSTRACT

Small extracellular vesicles (SEV) have attracted much attention both as mediators of intercellular communication and as drug delivery systems. In addition, recent studies have shown that SEV containing virus components and virus particles are released from virus-infected cells. Oncolytic viruses, which efficiently kill tumor cells by tumor cell-specific replication, have been actively studied as novel anticancer agents in clinical and preclinical studies. However, it remains to be fully elucidated whether SEV released from oncolytic virus-infected cells are involved in the antitumor effects of oncolytic viruses. In this study, we examined the tumor cell killing efficiencies and innate immune responses following treatment with SEV released from oncolytic reovirus-infected tumor cells in vitro and in vivo. Reovirus-infected B16 cells secreted SEV associated with or containing reovirus particles (Reo-SEV) with a diameter of approximately 130 nm and a zeta potential of -17 mV, although death of reovirus-infected B16 cells was not observed. The secreted Reo-SEV also contained interferon (IFN)-ß, tumor antigens, and damage-associated molecular patterns (DAMPs), including heat shock proteins (HSPs). Reo-SEV were secreted from the tumor tissues of reovirus-injected mice. Inhibition of the SEV secretion pathway using GW4869, which is a neutral sphingomyelinase inhibitor, resulted in significant reduction in the infectious titers of reovirus in the culture supernatants, suggesting that the cells released progeny virus via the SEV secretion pathway. Reo-SEV more efficiently killed mouse tumor cells and induced innate immune responses in mouse bone marrow-derived dendritic cells than reovirus. Reovirus and Reo-SEV mediated efficient and comparable levels of growth suppression of B16 subcutaneous tumors and induction of tumor infiltration of CD8+ T cells following intravenous administration. These results indicate that Reo-SEV are a promising oncolytic agent and that SEV are an effective delivery vehicle for oncolytic virus.


Subject(s)
Antigens, Neoplasm , Extracellular Vesicles , Interferon-beta , Mice, Inbred C57BL , Reoviridae , Animals , Cell Line, Tumor , Antigens, Neoplasm/immunology , Mice , Oncolytic Virotherapy/methods , Oncolytic Viruses , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Aniline Compounds/pharmacology , Aniline Compounds/administration & dosage , Immunity, Innate , Female , Benzylidene Compounds/pharmacology , Humans
20.
Exp Dermatol ; 33(9): e15169, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39207089

ABSTRACT

Despite rising melanoma incidence in recent decades, there is a trend towards overall decreased mortality, reflecting multiple factors including improved treatment options for metastatic disease. While local treatments are the mainstay for early-stage melanoma, metastatic disease necessitates systemic treatment, with oncolytic virotherapy emerging as a promising option. For this review, articles were retrieved from PubMed from 1964 through 2024. We conducted title, abstract and full-text screening in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines to identify articles describing the use of coxsackievirus A21 (V937), either as monotherapy or as part of combination therapy for malignant melanoma. Fifteen articles met inclusion criteria, offering preclinical and clinical data on V937's efficacy in reducing tumour burden. In addition to reporting manageable safety profiles, clinical trial data examining intratumoral V937 combination therapy with pembrolizumab and ipilimumab also endorsed favourable objective response rates compared to immune checkpoint inhibitor monotherapy (47% vs. 38% and 21% vs. 10%, respectively). In contrast, intravenous V937 monotherapy failed to yield additional benefit in a cohort of patients with Stage IIIC/IV melanoma (n = 3) despite achieving detectable levels in tumour tissue (1 × 109 TCID50). Although small subsets of patients experienced severe adverse effects and study design limitations imposed constraints on collected data, evidence for the efficacy of V937 remains encouraging. With few clinical trials evaluating V937 in melanoma, additional data is required before routine usage in standard treatment for metastatic lesions.


Subject(s)
Antibodies, Monoclonal, Humanized , Melanoma , Oncolytic Virotherapy , Skin Neoplasms , Humans , Melanoma/therapy , Skin Neoplasms/therapy , Skin Neoplasms/pathology , Antibodies, Monoclonal, Humanized/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Enterovirus , Ipilimumab/therapeutic use , Combined Modality Therapy , Oncolytic Viruses
SELECTION OF CITATIONS
SEARCH DETAIL