Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Transl Res ; 240: 64-86, 2022 02.
Article in English | MEDLINE | ID: mdl-34757194

ABSTRACT

Oncolytic virotherapy is a new and safe therapeutic strategy for cancer treatment. In our previous study, a new type of oncolytic herpes simplex virus type 2 (oHSV2) was constructed. Following the completion of a preclinical study, oHSV2 has now entered into clinical trials for the treatment of melanoma and other solid tumors (NCT03866525). Oncolytic viruses (OVs) are generally able to directly destroy tumor cells and stimulate the immune system to fight tumors. Natural killer (NK) cells are important components of the innate immune system and critical players against tumor cells. But the detailed interactions between oncolytic viruses and NK cells and these interaction effects on the antitumor immune response remain to be elucidated. In particular, the functions of activating surface receptors and checkpoint inhibitors on oHSV2-treated NK cells and tumor cells are still unknown. In this study, we found that UV-oHSV2 potently activates human peripheral blood mononuclear cells, leading to increased antitumor activity in vitro and in vivo. Further investigation indicated that UV-oHSV2-stimulated NK cells release IFN-γ via Toll-like receptor 2 (TLR2)/NF-κB signaling pathway and exert antitumor activity via TLR2. We found for the first time that the expression of a pair of checkpoint molecules, NKG2A (on NK cells) and HLA-E (on tumor cells), is upregulated by UV-oHSV2 stimulation. Anti-NKG2A and anti-HLA-E treatment could further enhance the antitumor effects of UV-oHSV2-stimulated NK92 cells in vitro and in vivo. As our oHSV2 clinical trial is ongoing, we expect that the combination therapy of oncolytic virus oHSV2 and anti-NKG2A/anti-HLA-E antibodies may have synergistic antitumor effects in our future clinical trials.


Subject(s)
Herpesvirus 2, Human/radiation effects , Immune Checkpoint Inhibitors/pharmacology , Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Viruses/radiation effects , Ultraviolet Rays , Virus Inactivation/radiation effects , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cytotoxicity, Immunologic/drug effects , Female , Herpesvirus 2, Human/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Oncolytic Viruses/drug effects , Signal Transduction/drug effects , Toll-Like Receptor 2/metabolism , Virus Inactivation/drug effects , HLA-E Antigens
2.
J Immunother Cancer ; 9(12)2021 12.
Article in English | MEDLINE | ID: mdl-34949694

ABSTRACT

BACKGROUND: In addition to directly lysing tumors, oncolytic viruses also induce antitumor immunity by recruiting and activating immune cells in the local tumor microenvironment. However, the activation of the immune cells induced by oncolytic viruses is always accompanied by high-level expression of immune checkpoints in these cells, which may reduce the efficacy of the oncolytic viruses. The aim of this study is to arm the oncolytic vaccinia virus (VV) with immune checkpoint blockade to enhance its antitumor efficacy. METHODS: Through homologous recombination with the parental VV, an engineered VV-scFv-TIGIT was produced, which encodes a single-chain variable fragment (scFv) targeting T-cell immunoglobulin and ITIM domain (TIGIT). The antitumor efficacy of the VV-scFv-TIGIT was explored in several subcutaneous and ascites tumor models. The antitumor efficacy of VV-scFv-TIGIT combined with programmed cell death 1 (PD-1) or lymphocyte-activation gene 3 (LAG-3) blockade was also investigated. RESULTS: The VV-scFv-TIGIT effectively replicated in tumor cells and lysed them, and prompt the infected tumor cells to secret the functional scFv-TIGIT. Compared with control VV, intratumoral injection of VV-scFv-TIGIT in several mouse subcutaneous tumor models showed superior antitumor efficacy, accompanied by more T cell infiltration and a higher degree of CD8+ T cells activation. Intraperitoneal injection of VV-scFv-TIGIT in a mouse model of malignant ascites also significantly improved T cell infiltration and CD8+ T cell activation, resulting in more than 90% of the tumor-bearing mice being cured. Furthermore, the antitumor immune response induced by VV-scFv-TIGIT was dependent on CD8+ T cells which mediated a long-term immunological memory and a systemic antitumor immunity against the same tumor. Finally, the additional combination of PD-1 or LAG-3 blockade further enhanced the antitumor efficacy of VV-scFv-TIGIT, increasing the complete response rate of tumor-bearing mice. CONCLUSIONS: Oncolytic virotherapy using engineered VV-scFv-TIGIT was an effective strategy for cancer immunotherapy. Administration of VV-scFv-TIGIT caused a profound reshaping of the suppressive tumor microenvironment from 'cold' to 'hot' status. VV-scFv-TIGIT also synergized with PD-1 or LAG-3 blockade to achieve a complete response to tumors with poor response to VV or immune checkpoint blockade monotherapy.


Subject(s)
Antigens, CD/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Oncolytic Viruses/drug effects , Receptors, Immunologic/immunology , Vaccinia virus/drug effects , Animals , Disease Models, Animal , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy/methods , Mice , Lymphocyte Activation Gene 3 Protein
3.
J Immunother Cancer ; 9(7)2021 07.
Article in English | MEDLINE | ID: mdl-34301814

ABSTRACT

BACKGROUND: Oncolytic reovirus therapy for cancer induces a typical antiviral response to this RNA virus, including neutralizing antibodies. Concomitant treatment with cytotoxic chemotherapies has been hypothesized to improve the therapeutic potential of the virus. Chemotherapy side effects can include immunosuppression, which may slow the rate of the antiviral antibody response, as well as potentially make the patient more vulnerable to viral infection. METHOD: Reovirus neutralizing antibody data were aggregated from separate phase I clinical trials of reovirus administered as a single agent or in combination with gemcitabine, docetaxel, carboplatin and paclitaxel doublet or cyclophosphamide. In addition, the kinetics of individual antibody isotypes were profiled in sera collected in these trials. RESULTS: These data demonstrate preserved antiviral antibody responses, with only moderately reduced kinetics with some drugs, most notably gemcitabine. All patients ultimately produced an effective neutralizing antibody response. CONCLUSION: Patients' responses to infection by reovirus are largely unaffected by the concomitant drug treatments tested, providing confidence that RNA viral treatment or infection is compatible with standard of care treatments.


Subject(s)
Antibodies, Viral/therapeutic use , Neoplasms/drug therapy , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/drug effects , Animals , Antibodies, Viral/pharmacology , Humans , Mice , Neoplasms/complications
4.
Cell Death Dis ; 11(12): 1062, 2020 12 12.
Article in English | MEDLINE | ID: mdl-33311488

ABSTRACT

Reversing the highly immunosuppressive tumor microenvironment (TME) is essential to achieve long-term efficacy with cancer immunotherapy. Despite the impressive clinical response to checkpoint blockade in multiple types of cancer, only a minority of patients benefit from this approach. Here, we report that the oncolytic virus M1 induces immunogenic tumor cell death and subsequently restores the ability of dendritic cells to prime antitumor T cells. Intravenous injection of M1 disrupts immune tolerance in the privileged TME, reprogramming immune-silent (cold) tumors into immune-inflamed (hot) tumors. M1 elicits potent CD8+ T cell-dependent therapeutic effects and establishes long-term antitumor immune memory in poorly immunogenic tumor models. Pretreatment with M1 sensitizes refractory tumors to subsequent checkpoint blockade by boosting T-cell recruitment and upregulating the expression of PD-L1. These findings reveal the antitumor immunological mechanism of the M1 virus and indicated that oncolytic viruses are ideal cotreatments for checkpoint blockade immunotherapy.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Oncolytic Viruses/metabolism , T-Lymphocytes/immunology , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Immune Checkpoint Inhibitors/pharmacology , Immunogenic Cell Death/drug effects , Immunologic Memory/drug effects , Inflammation/genetics , Injections, Intravenous , Mice, Inbred C57BL , Oncolytic Viruses/drug effects , T-Lymphocytes/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
5.
Mol Oncol ; 14(12): 3153-3168, 2020 12.
Article in English | MEDLINE | ID: mdl-33037696

ABSTRACT

Oncolytic viruses are potent anticancer agents that replicate within and kill cancer cells rather than normal cells, and their selectivity is largely determined by oncogenic mutations. M1, a novel oncolytic virus strain, has been shown to target cancer cells, but the relationship between its cancer selectivity and oncogenic signaling pathways is poorly understood. Here, we report that RAS mutation promotes the replication and oncolytic effect of M1 in cancer, and we further provide evidence that the inhibition of the RAS/RAF/MEK signaling axis suppresses M1 infection and the subsequent cytopathic effects. Transcriptome analysis revealed that the inhibition of RAS signaling upregulates the type I interferon antiviral response, and further RNA interference screen identified CDKN1A as a key downstream factor that inhibits viral infection. Gain- and loss-of-function experiments confirmed that CDKN1A inhibited the replication and oncolytic effect of M1 virus. Subsequent TCGA data mining and tissue microarray (TMA) analysis revealed that CDKN1A is commonly deficient in human cancers, suggesting extensive clinical application prospects for M1. Our report indicates that virotherapy is feasible for treating undruggable RAS-driven cancers and provides reliable biomarkers for personalized cancer therapy.


Subject(s)
Neoplasms/metabolism , Neoplasms/virology , Oncolytic Viruses/physiology , Signal Transduction , ras Proteins/metabolism , Animals , Antiviral Agents/pharmacology , Biomarkers, Tumor/metabolism , Butadienes/pharmacology , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Gene Expression Regulation, Viral/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , Mutation/genetics , Neoplasms/pathology , Nitriles/pharmacology , Oncolytic Viruses/drug effects , Oncolytic Viruses/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics , Virus Replication/drug effects , Virus Replication/genetics
6.
Cells ; 9(6)2020 06 05.
Article in English | MEDLINE | ID: mdl-32516884

ABSTRACT

Glioblastoma (GBM) is the most aggressive and frequent primary brain tumor in adults with a median overall survival of 15 months. Tumor recurrence and poor prognosis are related to cancer stem cells (CSCs), which drive resistance to therapies. A common characteristic in GBM is CDKN2A gene loss, located close to the cluster of type I IFN genes at Ch9p21. Newcastle disease virus (NDV) is an avian paramyxovirus with oncolytic and immunostimulatory properties that has been proposed for the treatment of GBM. We have analyzed the CDKN2A-IFN I gene cluster in 1018 glioma tumors and evaluated the NDV oncolytic effect in six GBM CSCs ex vivo and in a mouse model. Our results indicate that more than 50% of GBM patients have some IFN deletion. Moreover, GBM susceptibility to NDV is dependent on the loss of the type I IFN. Infection of GBM with an NDV-expressing influenza virus NS1 protein can overcome the resistance to oncolysis by NDV of type I-competent cells. These results highlight the potential of using NDV vectors in antitumor therapies.


Subject(s)
Brain Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Glioma/genetics , Glioma/therapy , Interferon Type I/genetics , Multigene Family , Newcastle disease virus/physiology , Oncolytic Viruses/physiology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Glioma/pathology , Humans , Interferon-beta/pharmacology , Kinetics , Models, Biological , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Newcastle disease virus/pathogenicity , Oncolytic Viruses/drug effects , Recombinant Proteins/pharmacology , Virus Replication/drug effects
7.
Biometals ; 32(3): 545-561, 2019 06.
Article in English | MEDLINE | ID: mdl-31209680

ABSTRACT

Oncolytic viruses rewire the immune system and can lead to long-lasting antitumor defenses against primary and metastatic tumors. However, results from clinical studies have shown heterogeneity in responses suggesting that multiplexed approaches may be necessary to consistently generate positive outcomes in patients. To this end, we explored the combination of oncolytic rhabdovirus VSV∆51 with vanadium(V) dipicolinate derivatives, which have already been explored for their antidiabetic properties in animal models. The combination of vanadium-based dipicolinate compounds with VSV∆51 significantly increased viral replication and cytotoxicity in the human renal cell carcinoma cell line 786-0. The effects of three vanadium(V)-dipicolinate coordination complexes ([VO2dipic]-, [VO2dipic-OH]- and [VO2dipic-Cl]- with -OH or -Cl in the para position) were compared to that of the simple salts using spectroscopy and speciation profiles. Like the vanadate salts and the vanadyl cation, all dioxovanadium(V) dipicolinate complexes tested were found to increase viral infection and cytotoxicity when used in combination with VSV∆51. Viral sensitization is dependent on the vanadium since free dipicolinate ligands exerted no effect on viral infection and viability. The ability of these complexes to interact with interfaces and the stability of the complexes were evaluated under physiological conditions. Results indicate that these complexes undergo hydrolysis in cell culture media thereby generating vanadate. The vanadium dipicolinate derivatives in the context of immunovirotherapy shares similarities with previous studies exploring the antidiabetic properties of the compounds. The synergy between vanadium compounds and the oncolytic virus suggests that these compounds may be valuable in the development of novel and effective pharmaco-viral therapies.


Subject(s)
Antiviral Agents/pharmacology , Coordination Complexes/pharmacology , Oncolytic Virotherapy , Oncolytic Viruses/drug effects , Picolinic Acids/pharmacology , Vanadium Compounds/pharmacology , Virus Diseases/therapy , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Humans , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Molecular Structure , Picolinic Acids/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured , Vanadium Compounds/chemistry , Virus Diseases/drug therapy
8.
Cancer Immunol Res ; 6(12): 1499-1510, 2018 12.
Article in English | MEDLINE | ID: mdl-30352799

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft-tissue sarcomas resistant to most cancer treatments. Surgical resection remains the primary treatment, but this is often incomplete, ultimately resulting in high mortality and morbidity rates. There has been a resurgence of interest in oncolytic virotherapy because of encouraging preclinical and clinical trial results. Oncolytic herpes simplex virus (oHSV) selectively replicates in cancer cells, lysing the cell and inducing antitumor immunity. We previously showed that basal interferon (IFN) signaling increases interferon-stimulated gene (ISG) expression, restricting viral replication in almost 50% of MPNSTs. The FDA-approved drug ruxolitinib (RUX) temporarily resets this constitutively active STAT signaling and renders the tumor cells susceptible to oHSV infection in cell culture. In the studies described here, we translated our in vitro results into a syngeneic MPNST tumor model. Consistent with our previous results, murine MPNSTs exhibit a similar IFN- and ISG-mediated oHSV-resistance mechanism, and virotherapy alone provides no antitumor benefit in vivo However, pretreatment of mice with ruxolitinib reduced ISG expression, making the tumors susceptible to oHSV infection. Ruxolitinib pretreatment improved viral replication and altered the oHSV-induced immune-mediated response. Our results showed that this combination therapy increased CD8+ T-cell activation in the tumor microenvironment and that this population was indispensable for the antitumor benefit that follows from the combination of RUX and oHSV. These data suggest that JAK inhibition prior to oncolytic virus treatment augments both oHSV replication and the immunotherapeutic efficacy of oncolytic herpes virotherapy.


Subject(s)
Herpesvirus 1, Human/drug effects , Nerve Sheath Neoplasms/therapy , Oncolytic Virotherapy/methods , Pyrazoles/pharmacology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Combined Modality Therapy , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation/drug effects , Herpesvirus 1, Human/physiology , Interferons/metabolism , Interferons/pharmacology , Mice, Inbred C57BL , Nerve Sheath Neoplasms/immunology , Nerve Sheath Neoplasms/mortality , Nitriles , Oncolytic Viruses/drug effects , Oncolytic Viruses/physiology , Pyrimidines , Survival Rate , Virus Replication/drug effects
9.
Mar Drugs ; 16(6)2018 Jun 07.
Article in English | MEDLINE | ID: mdl-29880736

ABSTRACT

Lectins play diverse roles in physiological processes as biological recognition molecules. In this report, a gene encoding Tachypleus tridentatus Lectin (TTL) was inserted into an oncolytic vaccinia virus (oncoVV) vector to form oncoVV-TTL, which showed significant antitumor activity in a hepatocellular carcinoma mouse model. Furthermore, TTL enhanced oncoVV replication through suppressing antiviral factors expression such as interferon-inducible protein 16 (IFI16), mitochondrial antiviral signaling protein (MAVS) and interferon-beta (IFN-ß). Further investigations revealed that oncoVV-TTL replication was highly dependent on ERK activity. This study might provide insights into a novel way of the utilization of TTL in oncolytic viral therapies.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cell Proliferation/drug effects , Horseshoe Crabs/metabolism , Lectins/pharmacology , Liver Neoplasms/drug therapy , Oncolytic Viruses/drug effects , Virus Replication/drug effects , Animals , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Carcinoma, Hepatocellular/virology , Cell Line , Cell Line, Tumor , DNA Replication/drug effects , HEK293 Cells , Humans , Interferon-beta/metabolism , Liver Neoplasms/virology , Mice , Mice, Inbred BALB C , Mice, Nude , Oncolytic Virotherapy/methods , Phosphoproteins/metabolism , Vaccinia virus/drug effects
10.
Front Immunol ; 9: 94, 2018.
Article in English | MEDLINE | ID: mdl-29441069

ABSTRACT

Malignancy-induced alterations to cytokine signaling in tumor cells differentially regulate their interactions with the immune system and oncolytic viruses. The abundance of inflammatory cytokines in the tumor microenvironment suggests that such signaling plays key roles in tumor development and therapy efficacy. The JAK-STAT axis transduces signals of interleukin-6 (IL-6) and interferons (IFNs), mediates antiviral responses, and is frequently altered in prostate cancer (PCa) cells. However, how activation of JAK-STAT signaling with different cytokines regulates interactions between oncolytic viruses and PCa cells is not known. Here, we employ LNCaP PCa cells, expressing (or not) JAK1, activated (or not) with IFNs (α or γ) or IL-6, and infected with RNA viruses of different oncolytic potential (EHDV-TAU, hMPV-GFP, or HIV-GFP) to address this matter. We show that in JAK1-expressing cells, IL-6 sensitized PCa cells to viral cell death in the presence or absence of productive infection, with dependence on virus employed. Contrastingly, IFNα induced a cytoprotective antiviral state. Biochemical and genetic (knockout) analyses revealed dependency of antiviral state or cytoprotection on STAT1 or STAT2 activation, respectively. In IL-6-treated cells, STAT3 expression was required for anti-proliferative signaling. Quantitative proteomics (SILAC) revealed a core repertoire of antiviral IFN-stimulated genes, induced by IL-6 or IFNs. Oncolysis in the absence of productive infection, induced by IL-6, correlated with reduction in regulators of cell cycle and metabolism. These results call for matching the viral features of the oncolytic agent, the malignancy-induced genetic-epigenetic alterations to JAK/STAT signaling and the cytokine composition of the tumor microenvironment for efficient oncolytic virotherapy.


Subject(s)
Host-Pathogen Interactions , Interferon-alpha/metabolism , Interleukin-6/metabolism , Janus Kinase 1/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Virus Diseases/metabolism , Virus Diseases/virology , Animals , Antiviral Agents/pharmacology , Biomarkers , Cell Line , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Interferon-alpha/pharmacology , Male , Oncolytic Viruses/drug effects , Oncolytic Viruses/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteome , STAT Transcription Factors/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Virus Diseases/immunology
11.
Sci Transl Med ; 10(425)2018 01 24.
Article in English | MEDLINE | ID: mdl-29367345

ABSTRACT

Resistance to oncolytic virotherapy is frequently associated with failure of tumor cells to get infected by the virus. Dimethyl fumarate (DMF), a common treatment for psoriasis and multiple sclerosis, also has anticancer properties. We show that DMF and various fumaric and maleic acid esters (FMAEs) enhance viral infection of cancer cell lines as well as human tumor biopsies with several oncolytic viruses (OVs), improving therapeutic outcomes in resistant syngeneic and xenograft tumor models. This results in durable responses, even in models otherwise refractory to OV and drug monotherapies. The ability of DMF to enhance viral spread results from its ability to inhibit type I interferon (IFN) production and response, which is associated with its blockade of nuclear translocation of the transcription factor nuclear factor κB (NF-κB). This study demonstrates that unconventional application of U.S. Food and Drug Administration-approved drugs and biological agents can result in improved anticancer therapeutic outcomes.


Subject(s)
Dimethyl Fumarate/pharmacology , NF-kappa B/metabolism , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Animals , Cell Line, Tumor , Cytokines/biosynthesis , Esters/pharmacology , Fumarates/pharmacology , Glutathione/metabolism , Humans , Interferon Type I/pharmacology , Maleates/pharmacology , Mice, Inbred BALB C , Mice, Inbred C57BL , Oncolytic Viruses/drug effects , Xenograft Model Antitumor Assays
12.
Hum Gene Ther ; 29(8): 950-961, 2018 08.
Article in English | MEDLINE | ID: mdl-28750564

ABSTRACT

Oncolytic virotherapy is a novel and intriguing treatment strategy for cancer therapy. However, the clinical potential of oncolytic virus as single agent is limited. M1 virus is a promising oncolytic virus that has been tested in preclinical studies. In this study, we investigated the effect of the combination use of M1 virus and Bcl-2 family inhibitors. A chemical compounds screening including ten Bcl-2 family inhibitors demonstrated that pan-Bcl-2 inhibitors selectively augmented M1 virus oncolysis in cancer cells at very low doses. The mechanism of the enhanced antitumor effect of pan-Bcl-2 inhibitors with M1 virus is mainly due to the inhibition of Bcl-xL, which synergizes with M1-induced upregulation of Bak to trigger apoptosis. In xenograft mouse models and patient-derived tumor tissues, the combination of M1 and pan-Bcl-2 inhibitors significantly inhibited tumor growth and prolonged survival, suggesting the potential therapeutic value of this strategy. These findings offer insights into the synergy between Bcl-xL inhibition and oncolytic virus M1 as a combination anticancer treatment modality.


Subject(s)
Neoplasms/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Apoptosis/drug effects , Cell Line, Tumor , Combined Modality Therapy , Humans , Mice , Mitochondria/drug effects , Neoplasms/drug therapy , Neoplasms/virology , Oncolytic Viruses/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
13.
Cancer Biol Ther ; 19(3): 188-197, 2018 03 04.
Article in English | MEDLINE | ID: mdl-29252087

ABSTRACT

The preclinical evaluation of oncolytic adenoviruses (OAds) has been limited to cancer xenograft mouse models because OAds replicate poorly in murine cancer cells. The alkylating agent temozolomide (TMZ) has been shown to enhance oncolytic virotherapy in human cancer cells; therefore, we investigated whether TMZ could increase OAd replication and oncolysis in murine cancer cells. To test our hypothesis, three murine cancer cells were infected with OAd (E1b-deleted) alone or in combination with TMZ. TMZ increased OAd-mediated oncolysis in all three murine cancer cells tested. This increased oncolysis was, at least in part, due to productive virus replication, apoptosis, and autophagy induction. Most importantly, murine lung non-cancerous cells were not affected by OAd+TMZ. Moreover, TMZ increased Ad transduction efficiency. However, TMZ did not increase coxsackievirus and adenovirus receptor; therefore, other mechanism could be implicated on the transduction efficiency. These results showed, for the first time, that TMZ could render murine tumor cells more susceptible to oncolytic virotherapy. The proposed combination of OAds with TMZ presents an attractive approach towards the evaluation of OAd potency and safety in syngeneic mouse models using these murine cancer cell-lines in vivo.


Subject(s)
Adenoviridae/physiology , Antineoplastic Agents, Alkylating/pharmacology , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Temozolomide/pharmacology , Virus Replication/drug effects , Adenoviridae/drug effects , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Combined Modality Therapy/methods , Drug Evaluation, Preclinical/methods , Genetic Vectors/drug effects , Genetic Vectors/physiology , Mice , Neoplasms/therapy , Oncolytic Viruses/drug effects , Receptors, Virus/metabolism , Transduction, Genetic/methods
14.
Mol Cancer Ther ; 16(11): 2627-2638, 2017 11.
Article in English | MEDLINE | ID: mdl-28847988

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive subtype of non-Hodgkin lymphoma caused by Kaposi's sarcoma-associated herpesvirus (KSHV) infection. Currently, treatment options for patients with PEL are limited. Oncolytic viruses have been engineered as anticancer agents and have recently shown increased therapeutic promise. Similarly, lytic activation of endogenous viruses from latently infected tumor cells can also be applied as a cancer therapy. In theory, such a therapeutic strategy would induce oncolysis by viral replication, while simultaneously stimulating an immune response to viral lytic cycle antigens. We examined the combination of the FDA-approved drug ingenol-3-angelate (PEP005) with epigenetic drugs as a rational therapeutic approach for KSHV-mediated malignancies. JQ1, a bromodomain and extra terminal (BET) protein inhibitor, in combination with PEP005, not only robustly induced KSHV lytic replication, but also inhibited IL6 production from PEL cells. Using the dosages of these agents that were found to be effective in reactivating HIV (as a means to clear latent virus with highly active antiretroviral therapy), we were able to inhibit PEL growth in vitro and delay tumor growth in a PEL xenograft tumor model. KSHV reactivation was mediated by activation of the NF-κB pathway by PEP005, which led to increased occupancy of RNA polymerase II onto the KSHV genome. RNA-sequencing analysis further revealed cellular targets of PEP005, JQ1, and the synergistic effects of both. Thus, combination of PEP005 with a BET inhibitor may be considered as a rational therapeutic approach for the treatment of PEL. Mol Cancer Ther; 16(11); 2627-38. ©2017 AACR.


Subject(s)
Azepines/administration & dosage , Diterpenes/administration & dosage , Lymphoma, Primary Effusion/drug therapy , Sarcoma, Kaposi/therapy , Triazoles/administration & dosage , Animals , Cell Line, Tumor , DNA Replication/drug effects , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology , Mice , NF-kappa B/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Oncolytic Viruses/genetics , Oncolytic Viruses/pathogenicity , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , Virus Replication/drug effects , Xenograft Model Antitumor Assays
15.
Cell Mol Life Sci ; 74(9): 1681-1692, 2017 05.
Article in English | MEDLINE | ID: mdl-28004147

ABSTRACT

Biotherapeutics have revolutionized modern medicine by providing medicines that would not have been possible with small molecules. With respect to cancer therapies, this represents the current sector of the pharmaceutical industry having the largest therapeutic impact, as exemplified by the development of recombinant antibodies and cell-based therapies. In cancer, one of the most common regulatory alterations is the perturbation of translational control. Among these, changes in eukaryotic initiation factor 4F (eIF4F) are associated with tumor initiation, progression, and drug resistance in a number of settings. This, coupled with the fact that systemic suppression of eIF4F appears well tolerated, indicates that therapeutic agents targeting eIF4F hold much therapeutic potential. Here, we discuss opportunities offered by biologicals for this purpose.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Animals , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Eukaryotic Initiation Factor-4F/metabolism , Humans , Oncolytic Viruses/drug effects , Oncolytic Viruses/metabolism , Protein Biosynthesis/drug effects
16.
Sci Rep ; 6: 26786, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27226390

ABSTRACT

The use of engineered viral strains such as gene therapy vectors and oncolytic viruses (OV) to selectively destroy cancer cells is poised to make a major impact in the clinic and revolutionize cancer therapy. In particular, several studies have shown that OV therapy is safe and well tolerated in humans and can infect a broad range of cancers. Yet in clinical studies OV therapy has highly variable response rates. The heterogeneous nature of tumors is widely accepted to be a major obstacle for OV therapeutics and highlights a need for strategies to improve viral replication efficacy. Here, we describe the development of a new class of small molecules for selectively enhancing OV replication in cancer tissue. Medicinal chemistry studies led to the identification of compounds that enhance multiple OVs and gene therapy vectors. Lead compounds increase OV growth up to 2000-fold in vitro and demonstrate remarkable selectivity for cancer cells over normal tissue ex vivo and in vivo. These small molecules also demonstrate enhanced stability with reduced electrophilicity and are highly tolerated in animals. This pharmacoviral approach expands the scope of OVs to include resistant tumors, further potentiating this transformative therapy. It is easily foreseeable that this approach can be applied to therapeutically enhance other attenuated viral vectors.


Subject(s)
Furans/pharmacology , Herpesvirus 1, Human/drug effects , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Vesicular stomatitis Indiana virus/drug effects , Virus Replication/drug effects , Adenocarcinoma/therapy , Animals , Cell Line, Tumor , Colonic Neoplasms/therapy , Drug Evaluation, Preclinical , Drug Stability , Female , Glutathione/analysis , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Immediate-Early Proteins/deficiency , Immediate-Early Proteins/genetics , Mice , Mice, Inbred BALB C , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Serum , Stimulation, Chemical , Structure-Activity Relationship , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/physiology , Viral Matrix Proteins/deficiency , Viral Matrix Proteins/genetics
17.
Cancer Lett ; 376(2): 240-8, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27063964

ABSTRACT

The consistent latent presence of Epstein-Barr virus (EBV) in tumor cells offers potential for virus-targeted therapies. The switch from the latent form of EBV to the lytic form in tumor cells can lead to tumor cell lysis. In this study, we report that a natural small molecule compound, cordycepin, can induce lytic EBV infection in tumor cells. Subsequently, we demonstrate that cordycepin can enhance EBV reactivating capacity and EBV-positive tumor cell killing ability of low dose doxorubicin. The combination of cordycepin and doxorubicin phosphorylates CCAAT/enhancer binding protein ß (C/EBPß) through protein kinase C (PKC)-p38 mitogen activated protein kinases (p38 MAPK) signaling pathway, and C/EBPß is required for the activation of lytic EBV infection. Most importantly, an in vivo experiment demonstrates that the combination of cordycepin and doxorubicin is more effective in inhibiting tumor growth in SCID mice than is doxorubicin alone. Our findings establish that cordycepin can enhance the efficacy of conventional chemotherapy for treatment of EBV-positive tumors.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxyadenosines/pharmacology , Doxorubicin/pharmacology , Herpesvirus 4, Human/drug effects , Oncolytic Virotherapy , Oncolytic Viruses/drug effects , Stomach Neoplasms/therapy , Virus Activation/drug effects , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Herpesvirus 4, Human/pathogenicity , Humans , Mice, SCID , Oncolytic Viruses/pathogenicity , Phosphorylation , Protein Kinase C/metabolism , RNA Interference , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/virology , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Biomaterials ; 84: 76-85, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26826297

ABSTRACT

Interactions between malignant and stromal cells and the 3D spatial architecture of the tumor both substantially modify tumor behavior, including the responses to small molecule drugs and biological therapies. Conventional 2D culture systems cannot replicate this complexity. To overcome these limitations and more accurately model solid tumors, we developed a highly versatile 3D PEG-fibrin hydrogel model of human lung adenocarcinoma. Our model relevantly recapitulates the effect of oncolytic adenovirus; tumor responses in this setting nearly reproduce those observed in vivo. We have also validated the use of this model for complex, long-term, 3D cultures of cancer cells and their stroma (fibroblasts and endothelial cells). Both tumor proliferation and invasiveness were enhanced in the presence of stromal components. These results validate our 3D hydrogel model as a relevant platform to study cancer biology and tumor responses to biological treatments.


Subject(s)
Adenoviridae/physiology , Fibrin/pharmacology , Models, Biological , Neoplasms/pathology , Oncolytic Viruses/physiology , Polyethylene Glycols/pharmacology , Tumor Microenvironment/drug effects , Adenoviridae/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate , Mice, SCID , Neoplasm Invasiveness , Oncolytic Viruses/drug effects , Organoids , Phenotype , Stromal Cells/drug effects , Stromal Cells/pathology
19.
Viruses ; 8(1)2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26751469

ABSTRACT

Oncolytic viruses (OVs) target and destroy cancer cells while sparing their normal counterparts. These viruses have been evaluated in numerous studies at both pre-clinical and clinical levels and the recent Food and Drug Administration (FDA) approval of an oncolytic herpesvirus-based treatment raises optimism that OVs will become a therapeutic option for cancer patients. However, to improve clinical outcome, there is a need to increase OV efficacy. In addition to killing cancer cells directly through lysis, OVs can stimulate the induction of anti-tumour immune responses. The host immune system thus represents a "double-edged sword" for oncolytic virotherapy: on the one hand, a robust anti-viral response will limit OV replication and spread; on the other hand, the immune-mediated component of OV therapy may be its most important anti-cancer mechanism. Although the relative contribution of direct viral oncolysis and indirect, immune-mediated oncosuppression to overall OV efficacy is unclear, it is likely that an initial period of vigorous OV multiplication and lytic activity will most optimally set the stage for subsequent adaptive anti-tumour immunity. In this review, we consider the use of histone deacetylase (HDAC) inhibitors as a means of boosting virus replication and lessening the negative impact of innate immunity on the direct oncolytic effect. We also discuss an alternative approach, aimed at potentiating OV-elicited anti-tumour immunity through the blockade of immune checkpoints. We conclude by proposing a two-phase combinatorial strategy in which initial OV replication and spread is maximised through transient HDAC inhibition, with anti-tumour immune responses subsequently enhanced by immune checkpoint blockade.


Subject(s)
Histone Deacetylase Inhibitors/administration & dosage , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Animals , Humans , Neoplasms/drug therapy , Oncolytic Viruses/drug effects , Oncolytic Viruses/genetics
20.
Virology ; 485: 340-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26331681

ABSTRACT

Vesicular stomatitis virus (VSV) is an effective oncolytic virus against most human pancreatic ductal adenocarcinoma (PDAC) cell lines. However, some PDAC cell lines are highly resistant to oncolytic VSV-ΔM51 infection. To better understand the mechanism of resistance, we tested a panel of 16 small molecule inhibitors of different cellular signaling pathways, and identified TPCA-1 (IKK-ß inhibitor) and ruxolitinib (JAK1/2 inhibitor), as strong enhancers of VSV-ΔM51 replication and virus-mediated oncolysis in all VSV-resistant PDAC cell lines. Both TPCA-1 and ruxolitinib similarly inhibited STAT1 and STAT2 phosphorylation and decreased expression of antiviral genes MxA and OAS. Moreover, an in situ kinase assay provided biochemical evidence that TPCA-1 directly inhibits JAK1 kinase activity. Together, our data demonstrate that TPCA-1 is a unique dual inhibitor of IKK-ß and JAK1 kinase, and provide a new evidence that upregulated type I interferon signaling plays a major role in resistance of pancreatic cancer cells to oncolytic viruses.


Subject(s)
Amides/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/virology , Protein Kinase Inhibitors/pharmacology , Thiophenes/pharmacology , Vesicular stomatitis Indiana virus/physiology , Cell Line, Tumor , Cytopathogenic Effect, Viral/drug effects , Humans , Interferon Type I/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Myxovirus Resistance Proteins/genetics , Nitriles , Oncolytic Viruses/drug effects , Oncolytic Viruses/physiology , Pyrazoles/pharmacology , Pyrimidines , STAT Transcription Factors/metabolism , Sendai virus/drug effects , Sendai virus/physiology , Signal Transduction/drug effects , Vesicular stomatitis Indiana virus/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...