1
|
Stergiopoulos GM, Iankov I, Galanis E. Personalizing Oncolytic Immunovirotherapy Approaches. Mol Diagn Ther 2024; 28:153-168. [PMID: 38150172 DOI: 10.1007/s40291-023-00689-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2023] [Indexed: 12/28/2023]
Abstract
Development of successful cancer therapeutics requires exploration of the differences in genetics, metabolism, and interactions with the immune system among malignant and normal cells. The clinical observation of spontaneous tumor regression following natural infection with microorganism has created the premise of their use as cancer therapeutics. Oncolytic viruses (OVs) originate from viruses with attenuated virulence in humans, well-characterized vaccine strains of known human pathogens, or engineered replication-deficient viral vectors. Their selectivity is based on receptor expression level and post entry restriction factors that favor replication in the tumor, while keeping the normal cells unharmed. Clinical trials have demonstrated a wide range of patient responses to virotherapy, with subgroups of patients significantly benefiting from OV administration. Tumor-specific gene signatures, including antiviral interferon-stimulated gene (ISG) expression profile, have demonstrated a strong correlation with tumor permissiveness to infection. Furthermore, the combination of OVs with immunotherapeutics, including anticancer vaccines and immune checkpoint inhibitors [ICIs, such as anti-PD-1/PD-L1 or anti-CTLA-4 and chimeric antigen receptor (CAR)-T or CAR-NK cells], could synergistically improve the therapeutic outcome. Creating response prediction algorithms represents an important step for the transition to individualized immunovirotherapy approaches in the clinic. Integrative predictors could include tumor mutational burden (TMB), inflammatory gene signature, phenotype of tumor-infiltrating lymphocytes, tumor microenvironment (TME), and immune checkpoint receptor expression on both immune and target cells. Additionally, the gut microbiota has recently been recognized as a systemic immunomodulatory factor and could further be used in the optimization of individualized immunovirotherapy algorithms.
Collapse
Affiliation(s)
| | - Ianko Iankov
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Evanthia Galanis
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Wu A, Li Z, Wang Y, Chen Y, Peng J, Zhu M, Li Y, Song H, Zhou D, Zhang C, Lv Y, Zhao Z. Recombinant measles virus vaccine rMV-Hu191 exerts an oncolytic effect on esophageal squamous cell carcinoma via caspase-3/GSDME-mediated pyroptosis. Cell Death Discov 2023; 9:171. [PMID: 37202386 DOI: 10.1038/s41420-023-01466-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/20/2023] Open
Abstract
Oncolytic viruses have recently been proven to be an effective and promising cancer therapeutic strategy, but there is rare data about oncolytic therapy in esophageal squamous cell carcinoma (ESCC), especially oncolytic measles virotherapy. Therefore, this study aimed to explore whether the recombinant measles virus vaccine strain rMV-Hu191 has an oncolytic effect against ESCC cells in vitro and in vivo and elucidate the underlying mechanisms. Our results showed that rMV-Hu191 could efficiently replicate in and kill ESCC cells through caspase-3/GSDME-mediated pyroptosis. Mechanistically, rMV-Hu191 triggers mitochondrial dysfunction to induce pyroptosis, which is mediated by BAK (BCL2 antagonist/killer 1) or BAX (BCL2 associated X). Further analysis revealed that rMV-Hu191 activates inflammatory signaling in ESCC cells, which may enhance the oncolytic efficiency. Moreover, intratumoral injection of rMV-Hu191 induced dramatic tumor regression in an ESCC xenograft model. Collectively, these findings imply that rMV-Hu191 exhibits an antitumor effect through BAK/BAX-dependent caspase-3/GSDME-mediated pyroptosis and provides a potentially promising new therapy for ESCC treatment.
Collapse
Affiliation(s)
- Ailing Wu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Zhongyue Li
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yilong Wang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yi Chen
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Jinkai Peng
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Mengying Zhu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yueyue Li
- Zhejiang Biosan Biotechnology Co., Ltd., Hangzhou, China
| | - Hai Song
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Thoracic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dongming Zhou
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chudi Zhang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Lv
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhengyan Zhao
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
3
|
Sakamoto A, Inoue H, Miyamoto S, Ito S, Soda Y, Tani K. Coxsackievirus A11 is an immunostimulatory oncolytic virus that induces complete tumor regression in a human non-small cell lung cancer. Sci Rep 2023; 13:5924. [PMID: 37046036 PMCID: PMC10097657 DOI: 10.1038/s41598-023-33126-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 04/07/2023] [Indexed: 04/14/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality worldwide. Innovative treatment is required to improve overall survival rates for advanced NSCLC. Oncolytic virotherapy using enteroviruses has emerged as a promising anticancer strategy. To identify a novel, potent virotherapy with an improved safety profile, we assessed the oncolytic activity of 28 enteroviral strains and focused on coxsackievirus A11 (CVA11). CVA11 infection caused extensive oncolytic activity in all three of the examined human NSCLC cell lines, with high intercellular adhesion molecule-1 (ICAM-1) expression associated with greater CVA11-induced cytotoxicity. In vitro inhibition analysis using a pan-caspase inhibitor and western blot detection of cleaved poly (ADP-ribose) polymerase (PARP) indicated that apoptosis partly contributed to CVA11-driven cytotoxicity. CVA11 infection-induced immunogenic cell death in vitro was strongly suggested by substantial calreticulin expression and release of high mobility group box-1 protein (HMGB1). Moreover, in vivo treatment of human NSCLC xenografts with intratumoral CVA11 injection caused complete tumor regression in all treated mice, without significant weight loss. Our findings indicate that novel oncolytic virotherapy utilizing CVA11 may be less toxic and more effective than current treatments for human NSCLC, thus warranting further investigation in clinical trial settings, especially in combination with immunotherapy.
Collapse
Affiliation(s)
- Akira Sakamoto
- Laboratory of ALA Advanced Medical Research, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Inoue
- Department of Respiratory Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan.
| | - Shohei Miyamoto
- Laboratory of ALA Advanced Medical Research, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Division of Oncology, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Shun Ito
- Laboratory of ALA Advanced Medical Research, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Yasushi Soda
- Laboratory of ALA Advanced Medical Research, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Kenzaburo Tani
- Laboratory of ALA Advanced Medical Research, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
4
|
Okamura K, Inoue H, Tanaka K, Ikematsu Y, Furukawa R, Ota K, Yoneshima Y, Iwama E, Okamoto I. Immunostimulatory oncolytic activity of coxsackievirus A11 in human malignant pleural mesothelioma. Cancer Sci 2023; 114:1095-1107. [PMID: 36369966 PMCID: PMC9986072 DOI: 10.1111/cas.15645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 11/14/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive solid cancer with a poor prognosis, whereas coxsackievirus A11 (CVA11) is a potential oncolytic virus for cancer treatment. We here investigated the oncolytic activity of CVA11 with human MPM cell lines. CVA11 infection was cytotoxic in all six MPM cell lines examined and showed no or minimal cytotoxicity toward normal human normal cell lines. MPM cells with a higher surface level of intercellular adhesion molecule-1 (ICAM-1) expression tended to be more susceptible to CVA11-induced cytotoxicity, and a neutralizing antibody to ICAM-1 attenuated such cytotoxicity. CVA11 infection activated signaling by Akt and extracellular signal-regulated kinase (ERK) pathways, and inhibitors of such signaling also abrogated CVA11-mediated cytotoxicity. Furthermore, CVA11 infection-triggered multiple modes of tumor cell death including apoptosis, pyroptosis, and necroptosis, and such death was accompanied by the release or exposure of the proinflammatory cytokine interleukin-1β and damage-associated molecular patterns such as calreticulin, high-mobility group box-1, annexin A1, and heat shock protein 70, which are hallmarks of immunogenic cell death. Notably, in vivo treatment of human MPM xenografts with intratumoral CVA11 injection resulted in significant suppression of tumor growth in SCID mice, and all mice infected with CVA11 showed no significant change in body weight. Our findings collectively suggest that the oncolytic activity of CVA11 for MPM is dependent on ICAM-1 as a virus receptor, as well as on Akt and ERK signaling, and that oncolytic virotherapy with CVA11 is a promising treatment modality with immunostimulatory activity for human MPM.
Collapse
Affiliation(s)
- Koji Okamura
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Inoue
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Respiratory Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Kentaro Tanaka
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuki Ikematsu
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Respiratory Medicine, National Hospital Organization Omuta Hospital, Fukuoka, Japan
| | - Rie Furukawa
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Respiratory Medicine, Hamanomachi Hospital, Fukuoka, Japan
| | - Keiichi Ota
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Yasuto Yoneshima
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Iwama
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
5
|
Chen XT, Dai SY, Zhan Y, Yang R, Chen DQ, Li Y, Zhou EQ, Dong R. Progress of oncolytic virotherapy for neuroblastoma. Front Pediatr 2022; 10:1055729. [PMID: 36467495 PMCID: PMC9716318 DOI: 10.3389/fped.2022.1055729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022] Open
Abstract
As a neuroendocrine tumor derived from the neural crest, neuroblastoma (NB) is the most common extracranial solid tumor in children. The prognosis in patients with low- and intermediate-risk NB is favorable while that in high-risk patients is often detrimental. However, the management of the considerably large proportion of high-risk patients remains challenging in clinical practice. Among various new approaches, oncolytic virus (OV) therapy offers great advantages in tumor treatment, especially for high-risk NB. Genetic modified OVs can target NB specifically without affecting normal tissue and avoid the widespread drug resistance issue in anticancer monotherapy. Meanwhile, its safety profile provides great potential in combination therapy with chemo-, radio-, and immunotherapy. The therapeutic efficacy of OV for NB is impressive from bench to bedside. The effectiveness and safety of OVs have been demonstrated and reported in studies on children with NB. Furthermore, clinical trials on some OVs (Celyvir, Pexa-Vec (JX-594) and Seneca Valley Virus (NTX-010)) have reported great results. This review summarizes the latest evidence in the therapeutic application of OVs in NB, including those generated in cell lines, animal models and clinical trials.
Collapse
Affiliation(s)
- Xiao-Tong Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Shu-Yang Dai
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Yong Zhan
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Ran Yang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - De-Qian Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Yi Li
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - En-Qing Zhou
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| |
Collapse
|
6
|
Zhang CD, Wang YL, Zhou DM, Zhu MY, Lv Y, Hao XQ, Qu CF, Chen Y, Gu WZ, Wu BQ, Chen PC, Zhao ZY. A recombinant Chinese measles virus vaccine strain rMV-Hu191 inhibits human colorectal cancer growth through inducing autophagy and apoptosis regulating by PI3K/AKT pathway. Transl Oncol 2021; 14:101091. [PMID: 33848808 PMCID: PMC8063909 DOI: 10.1016/j.tranon.2021.101091] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/15/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
The recombinant Chinese measles virus vaccine strain rMV-Hu191 induced efficient infection and oncolytic effects in human CRC both in vitro and in vivo. rMV-Hu191 induced the caspase-dependent apoptosis and complete autophagy in CRC cells. Autophagy served as a protective role in human CRC cells’ apoptosis induced by rMV-Hu191. rMV-Hu191-induced autophagy and apoptosis were regulated by the PI3K/AKT signaling pathway in human CRC.
The potential therapeutic effects of oncolytic measles virotherapy have been verified against plenty of malignancies. However, the oncolytic effects and underlying mechanisms of the recombinant Chinese measles virus vaccine strain Hu191 (rMV-Hu191) against human colorectal cancer (CRC) remain elusive. In this study, the antitumor effects of rMV-Hu191 were evaluated in CRC both in vitro and in vivo. From our data, rMV-Hu191 induced remarkably caspase-dependent apoptosis and complete autophagy in vitro. In mice bearing CRC xenografts, tumor volume was remarkably suppressed and median survival was prolonged significantly with intratumoral treatment of rMV-Hu191. To gain further insight into the relationship of rMV-Hu191-induced apoptosis and autophagy, we utilized Rapa and shATG7 to regulate autophagy. Our data suggested that autophagy was served as a protective role in rMV-Hu191-induced apoptosis in CRC. PI3K/AKT signaling pathway as one of the common upstream pathways of apoptosis and autophagy was activated in CRC after treatment with rMV-Hu191. And inhibition of PI3K/AKT pathway using LY294002 was accompanied by enhanced apoptosis and decreased autophagy which suggested that PI3K/AKT pathway promoted rMV-Hu191-induced autophagy and inhibited rMV-Hu191-induced apoptosis. This is the first study to demonstrate that rMV-Hu191 could be used as a potentially effective therapeutic agent in CRC treatment. As part of the underlying cellular mechanisms, apoptosis and autophagy were involved in the oncolytic effects generated by rMV-Hu191. And the cross-talk between these two processes and the PI3K/AKT signaling pathway was well identified.
Collapse
Affiliation(s)
- Chu-di Zhang
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China; Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang. China.
| | - Yi-Long Wang
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China.
| | - Dong-Ming Zhou
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China.
| | - Meng-Ying Zhu
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China; Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang. China.
| | - Yao Lv
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China.
| | - Xiao-Qiang Hao
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China.
| | - Chu-Fan Qu
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China; Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang. China.
| | - Yi Chen
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China; Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang. China.
| | - Wei-Zhong Gu
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China.
| | - Ben-Qing Wu
- University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen 518000, China.
| | - Pei-Chun Chen
- University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen 518000, China.
| | - Zheng-Yan Zhao
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang. China; Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang. China.
| |
Collapse
|
7
|
Measles Virus as an Oncolytic Immunotherapy. Cancers (Basel) 2021; 13:cancers13030544. [PMID: 33535479 PMCID: PMC7867054 DOI: 10.3390/cancers13030544] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Measles virus (MeV) preferentially replicates in malignant cells, leading to tumor lysis and priming of antitumor immunity. Live attenuated MeV vaccine strains are therefore under investigation as cancer therapeutics. The versatile MeV reverse genetics systems allows for engineering of advanced targeted, armed, and shielded oncolytic viral vectors. Therapeutic efficacy can further be enhanced by combination treatments. An emerging focus in this regard is combination immunotherapy, especially with immune checkpoint blockade. Despite challenges arising from antiviral immunity, availability of preclinical models, and GMP production, early clinical trials have demonstrated safety of oncolytic MeV and yielded promising efficacy data. Future clinical trials with engineered viruses, rational combination regimens, and comprehensive translational research programs will realize the potential of oncolytic immunotherapy.
Collapse
|
8
|
Mühlebach MD. Measles virus in cancer therapy. Curr Opin Virol 2020; 41:85-97. [PMID: 32861945 DOI: 10.1016/j.coviro.2020.07.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Over the last years, the development of viruses to treat cancer patients has re-gained considerable attention. A genetically modified herpesvirus, Talimogene laherparepvec, has already been authorized for the treatment of melanoma patients. Also recombinant measles virus (MeV) is developed as an oncolytic virus. Because of its high genetic flexibility, a number of different MeV strains have been the basis for the generation of targeted, armed, or shielded viruses that are highly specific for a given tumor target, more effective, or protected against serum neutralization. Such MeV have been extensively tested in vitro and in vivo, whereby remarkable oncolytic potency is accompanied by safety also in non-human primates. Therefore, MeV has been introduced into 19 different clinical trials and has reached phase II against two different tumor entities, multiple myeloma and ovarian carcinoma. Remarkably, one patient with advanced stage myeloma experienced long-term remission after treatment, visualizing the potency of this approach.
Collapse
Affiliation(s)
- Michael D Mühlebach
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, D-63225 Langen, Germany.
| |
Collapse
|
9
|
Abstract
With the recognition of oncolytic virotherapy as an immunotherapy, the distinct interactions between oncolytic agents and the immune system have come into focus. The role of the immune system in oncolytic virotherapy is somewhat ambiguous: While preexisting or arising immunity directed against viral antigens may preclude efficient viral replication and spread, immunity directed against tumor antigens is considered essential for long-term treatment success. Aside from the antiviral and antitumor immune status of the patient, the specific immunological microenvironment in a given tumor adds an additional layer of complexity.In this review we focus on the case of measles virus, which has long been known for its multifaceted interplay with the immune system. The high prevalence of measles-neutralizing antibodies in the general population may pose additional challenges. The oncolytic measles virus vector platform offers manifold opportunities for tumor-targeted immunomodulation. This review provides a survey of immunomodulation in the context of measles virotherapy including strategies to suppress or circumvent antiviral immunity as well as enhance antitumor immunity that have been pursued in preclinical and clinical studies. Understanding and selective manipulation of the intricate balance between antiviral and antitumor immunity will be crucial to develop the full potential of oncolytic virotherapy.
Collapse
|
10
|
Extremely Low Organ Toxicity and Strong Antitumor Activity of miR-34-Regulated Oncolytic Coxsackievirus B3. MOLECULAR THERAPY-ONCOLYTICS 2019; 12:246-258. [PMID: 30891489 PMCID: PMC6406029 DOI: 10.1016/j.omto.2019.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/17/2019] [Indexed: 12/19/2022]
Abstract
Oncolytic virotherapies have emerged as new modalities for cancer treatment. We previously reported that coxsackievirus B3 (CVB3) is a novel oncolytic virus (OV) with a strong ability to lyse human non-small cell lung cancer cells; however, its non-specific toxicity against normal cells remains to be resolved. To improve its safety profile, microRNA target sequences complementary to miR-34a/c, which is expressed preferentially in normal cells, were inserted into the 5′ UTR or 3′ UTR of the CVB3 genome. In the presence of miR-34a/c, the gene-modified CVB3 could not replicate in normal cells. We also found that the pathogenicity of CVB3 was reduced to a greater extent by targeting miR-34a than miR-34c; in addition, it was more effective to insert the target sequences into the 3′ UTR rather than the 5′ UTR of the viral genome. Ultimately, we developed a double-miR-34a targeting virus (53a-CVB) by inserting miR-34a targets in both the 5′ UTR and 3′ UTR of the virus. 53a-CVB was minimally toxic to cells in normal tissue, but maintained nearly its full oncolytic activity in mice xenografted with human lung cancer. 53a-CVB is the first miR-34-regulated OV and represents a promising platform for the development of safe and effective anti-cancer therapies.
Collapse
|
11
|
Qi Y, Xing K, Zhang L, Zhao F, Yao M, Hu A, Wu X. Protective immunity elicited by measles vaccine exerts anti-tumor effects on measles virus hemagglutinin gene-modified cancer cells in a mouse model. J Cancer Res Clin Oncol 2018; 144:1945-1957. [PMID: 30090962 DOI: 10.1007/s00432-018-2720-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/21/2018] [Indexed: 12/28/2022]
Abstract
PURPOSE Measles vaccine is widely used in China to prevent the measles virus (MV) infection. People immunized with measles vaccine can obtain long-term protective immunity. Measles virus surface glycoprotein hemagglutinin (H) can also induce MV-specific immune responses. However, little is known about whether the existence of the protective immune system against MV in the host can exert anti-tumor effects and whether the MV-H gene can serve as a therapeutic gene. METHODS We first vaccinated mice with measles vaccine, then inoculated them with MV-H protein-expressing tumor cells and observed the rate of tumor formation. We also treated mice with H protein-expressing tumor cells with measles vaccine and assessed tumor size and overall survival. RESULTS Active vaccination using measles vaccine not only protected mice from developing tumors, but also eradicated established tumors. Measles vaccine elicited H-specific IFN-γ, TNF-α and granzyme B-producing CD8+ T cells and increased cytotoxic T lymphocyte (CTL) activity specific for H antigen, which provided a strong therapeutic benefit against H protein-expressing tumors. In addition, measles vaccine decreased the population of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). CONCLUSIONS Our study demonstrated that tumor cells expressing H protein could activate the immune memory response against MV, which exerted specific anti-tumor effects, and indicated that the MV-H gene can be used as a potential therapeutic gene for cancer gene therapy.
Collapse
Affiliation(s)
- Yuan Qi
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Kailin Xing
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lanlin Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Fangyu Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln2200 Xietu Road, Shanghai, 200032, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln2200 Xietu Road, Shanghai, 200032, China
| | - Aiqun Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln2200 Xietu Road, Shanghai, 200032, China.
| | - Xianghua Wu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Abstract
INTRODUCTION Oncolytic viruses represent a novel treatment modality that is unencumbered by the standard resistance mechanisms limiting the therapeutic efficacy of conventional antineoplastic agents. Attenuated engineered measles virus strains derived from the Edmonston vaccine lineage have undergone extensive preclinical evaluation with significant antitumor activity observed in a broad range of preclinical tumoral models. These have laid the foundation for several clinical trials in both solid and hematologic malignancies, which have demonstrated safety, biologic activity and the ability to elicit antitumor immune responses. Areas covered: This review examines the published preclinical data which supported the clinical translation of this therapeutic platform, reviews the available clinical trial data and expands on ongoing phase II testing. It also looks at approaches to optimize clinical applicability and offers future perspectives. Expert opinion: Reverse genetic engineering has allowed the generation of oncolytic MV strains retargeted to increase viral tumor specificity, or armed with therapeutic and immunomodulatory genes in order to enhance anti-tumor efficacy. Continuous efforts focusing on exploring methods to overcome resistance pathways and determining optimal combinatorial strategies will facilitate further development of this encouraging antitumor strategy.
Collapse
Affiliation(s)
- Steven Robinson
- a Division of Medical Oncology , Mayo Clinic , Rochester , MN , USA
| | - Evanthia Galanis
- a Division of Medical Oncology , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
13
|
A novel, polymer-coated oncolytic measles virus overcomes immune suppression and induces robust antitumor activity. MOLECULAR THERAPY-ONCOLYTICS 2016; 3:16022. [PMID: 27847861 PMCID: PMC5091787 DOI: 10.1038/mto.2016.22] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/06/2016] [Accepted: 06/06/2016] [Indexed: 12/16/2022]
Abstract
Although various therapies are available to treat cancers, including surgery, chemotherapy, and radiotherapy, cancer has been the leading cause of death in Japan for the last 30 years, and new therapeutic modalities are urgently needed. As a new modality, there has recently been great interest in oncolytic virotherapy, with measles virus being a candidate virus expected to show strong antitumor effects. The efficacy of virotherapy, however, was strongly limited by the host immune response in previous clinical trials. To enhance and prolong the antitumor activity of virotherapy, we combined the use of two newly developed tools: the genetically engineered measles virus (MV-NPL) and the multilayer virus-coating method of layer-by-layer deposition of ionic polymers. We compared the oncolytic effects of this polymer-coated MV-NPL with the naked MV-NPL, both in vitro and in vivo. In the presence of anti-MV neutralizing antibodies, the polymer-coated virus showed more enhanced oncolytic activity than did the naked MV-NPL in vitro. We also examined antitumor activities in virus-treated mice. Complement-dependent cytotoxicity and antitumor activities were higher in mice treated with polymer-coated MV-NPL than in mice treated with the naked virus. This novel, polymer-coated MV-NPL is promising for clinical cancer therapy in the future.
Collapse
|
14
|
Wang B, Yan X, Guo Q, Li Y, Zhang H, Xie JS, Meng X. Deficiency of caspase 3 in tumor xenograft impairs therapeutic effect of measles virus Edmoston strain. Oncotarget 2016; 6:16019-30. [PMID: 25909216 PMCID: PMC4599253 DOI: 10.18632/oncotarget.3496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/07/2015] [Indexed: 11/25/2022] Open
Abstract
The oncolytic measles virus Edmonston (MV-Edm) strain shows considerable oncolytic activity against a variety of human tumors. In this study, we report MV-Edm is able to trigger apoptosis pathways in infected tumor cells and elucidate the roles of cellular apoptosis in the whole oncolytic process. We also show that activated caspase 3, a key executioner of apoptosis, plays key roles in the oncolytic virotherapy. Activated caspase 3 can accelerate viral replication in cervical cancer cells and enhance the killing effects of the virus. Deficiency of caspase 3 either in tumor cells or in tumor xenograft significantly desensitized tumor to oncolysis with MV-Edm. In the infected cells, caspase 3 regulates interferon α release, which can inhibit viral replication in neighboring tumor cells. We propose that caspase-3 activation enhances the oncolytic effects of MV-Edm, thus inhibiting tumor growth in mice.
Collapse
Affiliation(s)
- Biao Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University Shenyang, P.R. China
| | - Xu Yan
- Department of Prosthodontics, School of Stomatology, China Medical University, Shenyang, P.R. China
| | - Qingguo Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University Shenyang, P.R. China
| | - Yan Li
- Department of Oncology, Tumour Angiogenesis and Microenvironment Laboratory (TAML), First Affiliated Hospital, Liaoning Medical College, Jinzhou, P.R. China
| | - Haiyan Zhang
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang, P.R. China
| | - Ji Sheng Xie
- Department of Ecsomatics, Youjiang Medical College for Nationalities, Baise City, P.R. China
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University Shenyang, P.R. China
| |
Collapse
|
15
|
Tsun A, Miao XN, Wang CM, Yu DC. Oncolytic Immunotherapy for Treatment of Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 909:241-83. [PMID: 27240460 DOI: 10.1007/978-94-017-7555-7_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Immunotherapy entails the treatment of disease by modulation of the immune system. As detailed in the previous chapters, the different modes of achieving immune modulation are many, including the use of small/large molecules, cellular therapy, and radiation. Oncolytic viruses that can specifically attack, replicate within, and destroy tumors represent one of the most promising classes of agents for cancer immunotherapy (recently termed as oncolytic immunotherapy). The notion of oncolytic immunotherapy is considered as the way in which virus-induced tumor cell death (known as immunogenic cancer cell death (ICD)) allows the immune system to recognize tumor cells and provide long-lasting antitumor immunity. Both immune responses toward the virus and ICD together contribute toward successful antitumor efficacy. What is now becoming increasingly clear is that monotherapies, through any of the modalities detailed in this book, are neither sufficient in eradicating tumors nor in providing long-lasting antitumor immune responses and that combination therapies may deliver enhanced efficacy. After the rise of the genetic engineering era, it has been possible to engineer viruses to harbor combination-like characteristics to enhance their potency in cancer immunotherapy. This chapter provides a historical background on oncolytic virotherapy and its future application in cancer immunotherapy, especially as a combination therapy with other treatment modalities.
Collapse
Affiliation(s)
- A Tsun
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - X N Miao
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - C M Wang
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - D C Yu
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China.
| |
Collapse
|
16
|
Chung HK, Jacobs CL, Huo Y, Yang J, Krumm SA, Plemper RK, Tsien RY, Lin MZ. Tunable and reversible drug control of protein production via a self-excising degron. Nat Chem Biol 2015. [PMID: 26214256 PMCID: PMC4543534 DOI: 10.1038/nchembio.1869] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
An effective method for direct chemical control over the production of specific proteins would be widely useful. We describe Small Molecule-Assisted Shutoff (SMASh), a technique in which proteins are fused to a degron that removes itself in the absence of drug, leaving untagged protein. Clinically tested HCV protease inhibitors can then block degron removal, inducing rapid degradation of subsequently synthesized protein copies. SMASh allows reversible and dose-dependent shutoff of various proteins in multiple mammalian cell types and in yeast. We also used SMASh to confer drug responsiveness onto a RNA virus for which no licensed inhibitors exist. As SMASh does not require permanent fusion of a large domain, it should be useful when control over protein production with minimal structural modification is desired. Furthermore, as SMASh only involves a single genetic modification and does not rely on modulating protein-protein interactions, it should be easy to generalize to multiple biological contexts.
Collapse
Affiliation(s)
- Hokyung K Chung
- Department of Biology, Stanford University, Stanford, California, USA
| | - Conor L Jacobs
- Department of Biology, Stanford University, Stanford, California, USA
| | - Yunwen Huo
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Jin Yang
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Stefanie A Krumm
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Richard K Plemper
- 1] Department of Pediatrics, Emory University, Atlanta, Georgia, USA. [2] Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Roger Y Tsien
- 1] Department of Pharmacology, University of California, San Diego, La Jolla, California, USA. [2] Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA. [3] Howard Hughes Medical Institute, University of California, San Diego, La Jolla, California, USA
| | - Michael Z Lin
- 1] Department of Pediatrics, Stanford University, Stanford, California, USA. [2] Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
17
|
Tumor Restrictions to Oncolytic Virus. Biomedicines 2014; 2:163-194. [PMID: 28548066 PMCID: PMC5423468 DOI: 10.3390/biomedicines2020163] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/17/2014] [Accepted: 03/28/2014] [Indexed: 02/06/2023] Open
Abstract
Oncolytic virotherapy has advanced since the days of its conception but therapeutic efficacy in the clinics does not seem to reach the same level as in animal models. One reason is premature oncolytic virus clearance in humans, which is a reasonable assumption considering the immune-stimulating nature of the oncolytic agents. However, several studies are beginning to reveal layers of restriction to oncolytic virotherapy that are present before an adaptive neutralizing immune response. Some of these barriers are present constitutively halting infection before it even begins, whereas others are raised by minute cues triggered by virus infection. Indeed, we and others have noticed that delivering viruses to tumors may not be the biggest obstacle to successful therapy, but instead the physical make-up of the tumor and its capacity to mount antiviral defenses seem to be the most important efficacy determinants. In this review, we summarize the constitutive and innate barriers to oncolytic virotherapy and discuss strategies to overcome them.
Collapse
|
18
|
Inoue H, Tani K. Multimodal immunogenic cancer cell death as a consequence of anticancer cytotoxic treatments. Cell Death Differ 2013; 21:39-49. [PMID: 23832118 DOI: 10.1038/cdd.2013.84] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 05/06/2013] [Accepted: 05/14/2013] [Indexed: 02/06/2023] Open
Abstract
Apoptotic cell death generally characterized by a morphologically homogenous entity has been considered to be essentially non-immunogenic. However, apoptotic cancer cell death, also known as type 1 programmed cell death (PCD), was recently found to be immunogenic after treatment with several chemotherapeutic agents and oncolytic viruses through the emission of various danger-associated molecular patterns (DAMPs). Extensive studies have revealed that two different types of immunogenic cell death (ICD) inducers, recently classified by their distinct actions in endoplasmic reticulum (ER) stress, can reinitiate immune responses suppressed by the tumor microenvironment. Indeed, recent clinical studies have shown that several immunotherapeutic modalities including therapeutic cancer vaccines and oncolytic viruses, but not conventional chemotherapies, culminate in beneficial outcomes, probably because of their different mechanisms of ICD induction. Furthermore, interests in PCD of cancer cells have shifted from its classical form to novel forms involving autophagic cell death (ACD), programmed necrotic cell death (necroptosis), and pyroptosis, some of which entail immunogenicity after anticancer treatments. In this review, we provide a brief outline of the well-characterized DAMPs such as calreticulin (CRT) exposure, high-mobility group protein B1 (HMGB1), and adenosine triphosphate (ATP) release, which are induced by the morphologically distinct types of cell death. In the latter part, our review focuses on how emerging oncolytic viruses induce different forms of cell death and the combinations of oncolytic virotherapies with further immunomodulation by cyclophosphamide and other immunotherapeutic modalities foster dendritic cell (DC)-mediated induction of antitumor immunity. Accordingly, it is increasingly important to fully understand how and which ICD inducers cause multimodal ICD, which should aid the design of reasonably multifaceted anticancer modalities to maximize ICD-triggered antitumor immunity and eliminate residual or metastasized tumors while sparing autoimmune diseases.
Collapse
Affiliation(s)
- H Inoue
- 1] Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan [2] Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan [3] Department of Advanced Molecular and Cell Therapy, Kyushu University Hospital,Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
19
|
Antitumor Virotherapy by Attenuated Measles Virus (MV). BIOLOGY 2013; 2:587-602. [PMID: 24832799 PMCID: PMC3960896 DOI: 10.3390/biology2020587] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 02/28/2013] [Accepted: 03/05/2013] [Indexed: 12/21/2022]
Abstract
Antitumor virotherapy consists of the use of replication-competent viruses to infect and kill tumor cells preferentially, without damaging healthy cells. Vaccine-attenuated strains of measles virus (MV) are good candidates for this approach. Attenuated MV uses the CD46 molecule as a major entry receptor into cells. This molecule negatively regulates the complement system and is frequently overexpressed by cancer cells to escape lysis by the complement system. MV exhibits oncolytic properties in many cancer types in vitro, and in mouse models. Phase I clinical trials using MV are currently underway. Here, we review the state of this therapeutic approach, with a focus on the effects of MV on the antitumor immune response.
Collapse
|
20
|
Msaouel P, Iankov ID, Dispenzieri A, Galanis E. Attenuated oncolytic measles virus strains as cancer therapeutics. Curr Pharm Biotechnol 2013; 13:1732-41. [PMID: 21740361 DOI: 10.2174/138920112800958896] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 09/18/2010] [Indexed: 12/18/2022]
Abstract
Attenuated measles virus vaccine strains have emerged as a promising oncolytic vector platform, having shown significant anti-tumor activity against a broad range of malignant neoplasms. Measles virus strains derived from the attenuated Edmonston-B (MV-Edm) vaccine lineage have been shown to selectively infect, replicate in and lyse cancer cells while causing minimal cytopathic effect on normal tissues. This review summarizes the preclinical data that led to the rapid clinical translation of oncolytic measles vaccine strains and provides an overview of early clinical data using this oncolytic platform. Furthermore, novel approaches currently under development to further enhance the oncolytic efficacy of MV-Edm strains, including strategies to circumvent immunity or modulate immune system responses, combinatorial approaches with standard treatment modalities, virus retargeting as well as strategies for in vivo monitoring of viral replication are discussed.
Collapse
Affiliation(s)
- P Msaouel
- Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
21
|
Msaouel P, Opyrchal M, Domingo Musibay E, Galanis E. Oncolytic measles virus strains as novel anticancer agents. Expert Opin Biol Ther 2013; 13:483-502. [PMID: 23289598 DOI: 10.1517/14712598.2013.749851] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Replication-competent oncolytic measles virus (MV) strains preferentially infect and destroy a wide variety of cancer tissues. Clinical translation of engineered attenuated MV vaccine derivatives is demonstrating the therapeutic potential and negligible pathogenicity of these strains in humans. AREAS COVERED The present review summarizes the mechanisms of MV tumor selectivity and cytopathic activity as well as the current data on the oncolytic efficacy and preclinical testing of MV strains. Investigational strategies to reprogram MV selectivity, escape antiviral immunity and modulate the immune system to enhance viral delivery and tumor oncolysis are also discussed. EXPERT OPINION Clinical viral kinetic data derived from noninvasive monitoring of reporter transgene expression will guide future protocols to enhance oncolytic MV efficacy. Anti-measles immunity is a major challenge of measles-based therapeutics and various strategies are being investigated to modulate immunity. These include the combination of MV therapy with immunosuppressive drugs, such as cyclophosphamide, the use of cell carriers and the introduction of immunomodulatory transgenes and wild-type virulence genes. Available MV retargeting technologies can address safety considerations that may arise as more potent oncolytic MV vectors are being developed.
Collapse
Affiliation(s)
- Pavlos Msaouel
- Albert Einstein College of Medicine, Jacobi Medical Center, Department of Internal Medicine, Bronx, NY, USA
| | | | | | | |
Collapse
|
22
|
Zhang SC, Cai WS, Zhang Y, Jiang KL, Zhang KR, Wang WL. Engineered measles virus Edmonston strain used as a novel oncolytic viral system against human neuroblastoma through a CD46 and nectin 4-independent pathway. Cancer Lett 2012; 325:227-37. [PMID: 22796607 DOI: 10.1016/j.canlet.2012.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 07/04/2012] [Accepted: 07/04/2012] [Indexed: 10/28/2022]
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in children. In this study, we investigated the potential antitumor capability of the engineered Edmonston strain of the carcinoembryonic antigen-expressing measles virus (MV-CEA) against human NB. The infection of a variety of NB cell lines, including SK-N-SH, SMS-KCNR, and primary NB cells, resulted in significant cytopathic effects. None of the NB cell lines showed an overexpression of the measles virus receptor CD46 and nectin 4, but the cell lines did support robust viral replication. The efficacy of this approach was examined in murine SK-N-SH xenograft models. Flow cytometry and TUNEL assays indicated an apoptotic mechanism of cell death. In summary, MV-CEA has potent therapeutic efficacy against NB mediated by a CD46- and nectin 4-independent pathway.
Collapse
Affiliation(s)
- Shu-Cheng Zhang
- Department of Pediatric Surgery, Major Laboratory of the Chinese Health Ministry for Congenital Malformations, Shengjing Hospital of China Medical University, Heping District, Shenyang, PR China
| | | | | | | | | | | |
Collapse
|
23
|
Zhang SC, Wang WL, Cai WS, Jiang KL, Yuan ZW. Engineered measles virus Edmonston strain used as a novel oncolytic viral system against human hepatoblastoma. BMC Cancer 2012; 12:427. [PMID: 23009685 PMCID: PMC3488522 DOI: 10.1186/1471-2407-12-427] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 09/23/2012] [Indexed: 12/26/2022] Open
Abstract
Background Hepatoblastoma (HB) is the most common primary, malignant pediatric liver tumor in children. The treatment results for affected children have markedly improved in recent decades. However, the prognosis for high-risk patients who have extrahepatic extensions, invasion of the large hepatic veins, distant metastases and very high alpha-fetoprotein (AFP) serum levels remains poor. There is an urgent need for the development of novel therapeutic approaches. Methods An attenuated strain of measles virus, derived from the Edmonston vaccine lineage, was genetically engineered to produce carcinoembryonic antigen (CEA). We investigated the antitumor potential of this novel viral agent against human HB both in vitro and in vivo. Results Infection of the Hep2G and HUH6 HB cell lines, at multiplicities of infection (MOIs) ranging from 0.01 to 1, resulted in a significant cytopathic effect consisting of extensive syncytia formation and massive cell death at 72–96 h after infection. Both of the HB lines overexpressed the measles virus receptor CD46 and supported robust viral replication, which correlated with CEA production. The efficacy of this approach in vivo was examined in murine Hep2G xenograft models. Flow cytometry assays indicated an apoptotic mechanism of cell death. Intratumoral administration of MV-CEA resulted in statistically significant delay of tumor growth and prolongation of survival. Conclusions The engineered measles virus Edmonston strain MV-CEA has potent therapeutic efficacy against HB cell lines and xenografts. Trackable measles virus derivatives merit further exploration in HB treatment.
Collapse
Affiliation(s)
- Shu-Cheng Zhang
- Department of Pediatric Surgery, Major Laboratory of Chinese Health Ministry for Congenital Malformations, Shengjing Hospital of China Medical University, 36 Sanhao Street Heping District, Shenyang 110004, P.R. China.
| | | | | | | | | |
Collapse
|
24
|
Hiss DC, Fielding BC. Optimization and preclinical design of genetically engineered viruses for human oncolytic therapy. Expert Opin Biol Ther 2012; 12:1427-47. [PMID: 22788715 DOI: 10.1517/14712598.2012.707183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Oncolytic viruses (OVs) occupy a strategic niche in the dynamic era of biological and gene therapy of human cancers. However, the use of OVs is the subject of close scrutiny due to impediments such as the insufficiency of patient generalizations posed by heterogeneous tumor responses to treatment, inherent or potentially lethal viral pathogenicities, unanticipated host- or immune-related adverse effects, and the emergence of virus-resistant cancer cells. These challenges can be overcome by the design and development of more definitive (optimized, targeted, and individualized) cancer virotherapeutics. AREAS COVERED The translation of current knowledge and recent innovations into rational treatment prospects hinges on an iterative loop of variables pertaining to genetically engineered viral oncolytic efficacy and safety profiles, mechanism-of-action data, potencies of synergistic oncolytic viral combinations with conventional tumor, immuno-, chemo-, and radiation treatment modalities, optimization of the probabilities of treatment successes in heterogeneous (virus-sensitive and -resistant) tumor cell populations by mathematical modeling, and lessons learned from preclinical studies and human clinical trials. EXPERT OPINION In recent years, it has become increasingly clear that proof-of-principle is critical for the preclinical optimization of oncolytic viruses to target heterogeneous forms of cancer and to prioritize current concerns related to the efficacy and safety of oncolytic virotherapy.
Collapse
Affiliation(s)
- Donavon C Hiss
- University of the Western Cape, Department of Medical Biosciences, Molecular Oncology Research Laboratory, Bellville, 7535, South Africa.
| | | |
Collapse
|
25
|
Lawson KA, Morris DG. Oncolytic virotherapy for renal cell carcinoma: a novel treatment paradigm? Expert Opin Biol Ther 2012; 12:891-903. [DOI: 10.1517/14712598.2012.685713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
26
|
Miyamoto S, Inoue H, Nakamura T, Yamada M, Sakamoto C, Urata Y, Okazaki T, Marumoto T, Takahashi A, Takayama K, Nakanishi Y, Shimizu H, Tani K. Coxsackievirus B3 Is an Oncolytic Virus with Immunostimulatory Properties That Is Active against Lung Adenocarcinoma. Cancer Res 2012; 72:2609-21. [DOI: 10.1158/0008-5472.can-11-3185] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Abstract
The cytolytic properties of viruses can be used to treat cancer. Replication of certain viruses is favoured in cancer cells, whereas others can be modified to obtain tumour specificity. This approach has evolved to become a new discipline called virotherapy. In addition, these replication-competent (oncolytic) viruses can be adapted as vectors for cancer gene therapy. The "armed" viruses show a double mechanism of action: direct destruction of cancer cells as a consequence of the lytic viral cycle, in combination with the effect of the therapeutic gene incorporated in the viral genome. Current trends in the field include strategies to increase the oncolytic potency of existing viruses; the evaluation of new candidates; the search for synergistic effects between different viruses and conventional therapies; and a rational approach to take advantage of the interplay between the viruses and the host immune system. This review summarises the most relevant achievements in recent years.
Collapse
|
28
|
Luo R, Cannon L, Hernandez J, Piovoso MJ, Zurakowski R. Controlling the Evolution of Resistance. JOURNAL OF PROCESS CONTROL 2011; 21:367-378. [PMID: 21516198 PMCID: PMC3079266 DOI: 10.1016/j.jprocont.2010.11.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Evolution has long been understood as the driving force for many problems of medical interest. The evolution of drug resistance in HIV and bacterial infections is recognized as one of the most significant emerging problems in medicine. In cancer therapy, the evolution of resistance to chemotherapeutic agents is often the differentiating factor between effective therapy and disease progression or death. Interventions to manage the evolution of resistance have, up to this point, been based on steady-state analysis of mutation and selection models. In this paper, we review the mathematical methods applied to studying evolution of resistance in disease. We present a broad review of several classical applications of mathematical modeling of evolution, and review in depth two recent problems which demonstrate the potential for interventions which exploit the dynamic behavior of resistance evolution models. The first problem addresses the problem of sequential treatment failures in HIV; we present a review of our recent publications addressing this problem. The second problem addresses a novel approach to gene therapy for pancreatic cancer treatment, where selection is used to encourage optimal spread of susceptibility genes through a target tumor, which is then eradicated during a second treatment phase. We review the recent in Vitro laboratory work on this topic, present a new mathematical model to describe the treatment process, and show why model-based approaches will be necessary to successfully implement this novel and promising approach.
Collapse
Affiliation(s)
- Rutao Luo
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE 19716, USA
| | - LaMont Cannon
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE 19716, USA
| | - Jason Hernandez
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE 19716, USA
| | - Michael J. Piovoso
- Department of Electrical Engineering, Penn State Great Valley, 30 E. Swedesford Road, Malvern, PA 19355
| | - Ryan Zurakowski
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE 19716, USA
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711
| |
Collapse
|
29
|
Lech PJ, Russell SJ. Use of attenuated paramyxoviruses for cancer therapy. Expert Rev Vaccines 2011; 9:1275-302. [PMID: 21087107 DOI: 10.1586/erv.10.124] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Paramyxoviruses, measles virus (MV), mumps virus (MuV) and Newcastle disease virus (NDV), are well known for causing measles and mumps in humans and Newcastle disease in birds. These viruses have been tamed (attenuated) and successfully used as vaccines to immunize their hosts. Remarkably, pathogenic MuV and vaccine strains of MuV, MV and NDV efficiently infect and kill cancer cells and are consequently being investigated as novel cancer therapies (oncolytic virotherapy). Phase I/II clinical trials have shown promise but treatment efficacy needs to be enhanced. Technologies being developed to increase treatment efficacy include: virotherapy in combination with immunosuppressive drugs (cyclophosphamide); retargeting of viruses to specific tumor types or tumor vasculature; using infected cell carriers to protect and deliver the virus to tumors; and genetic manipulation of the virus to increase viral spread and/or express transgenes during viral replication. Transgenes have enabled noninvasive imaging or tracking of viral gene expression and enhancement of tumor destruction.
Collapse
Affiliation(s)
- Patrycja J Lech
- Mayo Clinic, Department of Molecular Medicine, 200 1st Street SW, Rochester, MN 55905, USA.
| | | |
Collapse
|