1
|
Chekaoui A, Garofalo M, Gad B, Staniszewska M, Chiaro J, Pancer K, Gryciuk A, Cerullo V, Salmaso S, Caliceti P, Masny A, Wieczorek M, Pesonen S, Kuryk L. Cancer vaccines: an update on recent achievements and prospects for cancer therapy. Clin Exp Med 2024; 25:24. [PMID: 39720956 DOI: 10.1007/s10238-024-01541-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024]
Abstract
Decades of basic and translational research have led to a momentum shift in dissecting the relationship between immune cells and cancer. This culminated in the emergence of breakthrough immunotherapies that paved the way for oncologists to manage certain hard-to-treat cancers. The application of high-throughput techniques of genomics, transcriptomics, and proteomics was conclusive in making and expediting the manufacturing process of cancer vaccines. Using the latest research technologies has also enabled scientists to interpret complex and multiomics data of the tumour mutanome, thus identifying new tumour-specific antigens to design new generations of cancer vaccines with high specificity and long-term efficacy. Furthermore, combinatorial regimens of cancer vaccines with immune checkpoint inhibitors have offered new therapeutic approaches and demonstrated impressive efficacy in cancer patients over the last few years. In the present review, we summarize the current state of cancer vaccines, including their potential therapeutic effects and the limitations that hinder their effectiveness. We highlight the current efforts to mitigate these limitations and highlight ongoing clinical trials. Finally, a special focus will be given to the latest milestones expected to transform the landscape of cancer therapy and nurture hope among cancer patients.
Collapse
Affiliation(s)
- Arezki Chekaoui
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy.
| | - Beata Gad
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Monika Staniszewska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Jacopo Chiaro
- Drug Research Program (DRP), ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE) University of Helsinki, Helsinki, Finland
- Translational Immunology Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, Helsinki, Finland
| | - Katarzyna Pancer
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Aleksander Gryciuk
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Vincenzo Cerullo
- Drug Research Program (DRP), ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE) University of Helsinki, Helsinki, Finland
- Translational Immunology Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Medical Biotechnology and CEINGE, University Federico II of Naples, Naples, Italy
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Aleksander Masny
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | | | - Lukasz Kuryk
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland.
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy.
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland.
- Valo Therapeutics Oy, Helsinki, Finland.
| |
Collapse
|
2
|
Lecoultre M, Walker PR, El Helali A. Oncolytic virus and tumor-associated macrophage interactions in cancer immunotherapy. Clin Exp Med 2024; 24:202. [PMID: 39196415 PMCID: PMC11358230 DOI: 10.1007/s10238-024-01443-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024]
Abstract
Oncolytic viruses (OV) are a promising strategy in cancer immunotherapy. Their capacity to promote anti-tumoral immunity locally raises hope that cancers unresponsive to current immunotherapy approaches could be tackled more efficiently. In this context, tumor-associated macrophages (TAM) must be considered because of their pivotal role in cancer immunity. Even though TAM tend to inhibit anti-tumoral responses, their ability to secrete pro-inflammatory cytokines and phagocytose cancer cells can be harnessed to promote therapeutic cancer immunity. OVs have the potential to promote TAM pro-inflammatory functions that favor anti-tumoral immunity. But in parallel, TAM pro-inflammatory functions induce OV clearance in the tumor, thereby limiting OV efficacy and highlighting that the interaction between OV and TAM is a double edge sword. Moreover, engineered OVs were recently developed to modulate specific TAM functions such as phagocytic activity. The potential of circulating monocytes to deliver OV into the tumor after intravenous administration is also emerging. In this review, we will present the interaction between OV and TAM, the potential of engineered OV to modulate specific TAM functions, and the promising role of circulating monocytes in OV delivery to the tumor.
Collapse
Affiliation(s)
- Marc Lecoultre
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, China
- Division of General Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Paul R Walker
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Immunobiology of Brain Tumours Laboratory, Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland
| | - Aya El Helali
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, China.
| |
Collapse
|
3
|
Welsh OL, Roth AN, Sutherland DM, Dermody TS. Sequence polymorphisms in the reovirus σ1 attachment protein modulate encapsidation efficiency and replication in mice. J Virol 2024; 98:e0030524. [PMID: 38771042 PMCID: PMC11237452 DOI: 10.1128/jvi.00305-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
Many functions of viral attachment proteins are established, but less is known about the biological importance of viral attachment protein encapsidation efficiency. The mammalian orthoreovirus (reovirus) σ1 attachment protein forms filamentous trimers that incorporate into pentamers of the λ2 capsid protein. Reovirus strains vary in the efficiency of σ1 encapsidation onto progeny virions, which influences viral stability during entry into cells and the efficacy of tumor cell lysis. While the role of σ1 encapsidation has been evaluated in studies using cultured cells, the contribution of attachment protein encapsidation efficiency to viral infection in animals is less clear. Polymorphisms in reovirus σ1 at residues 22 and 249 have been implicated in viral dissemination in mice and susceptibility to proteolysis in the murine intestine, respectively. To determine whether these residues contribute to σ1 encapsidation efficiency, we engineered σ1 mutant viruses with single- and double-residue substitutions at sites 22 and 249. We found that substitutions at these sites alter the encapsidation of σ1 and that reoviruses encapsidating higher amounts of σ1 bind cells more avidly and have a modest replication advantage in a cell-type-specific manner relative to low σ1-encapsidating reoviruses. Furthermore, we found that a high σ1-encapsidating reovirus replicates and disseminates more efficiently in mice relative to a low σ1-encapsidating reovirus. These findings provide evidence of a relationship between viral attachment protein encapsidation efficiency and viral replication in cell culture and animal hosts. IMPORTANCE Viral attachment proteins can serve multiple functions during viral replication, including attachment to host cells, cell entry and disassembly, and modulation of host immune responses. The relationship between viral attachment protein encapsidation efficiency and viral replication in cells and animals is poorly understood. We engineered and characterized a panel of reoviruses that differ in the capacity to encapsidate the σ1 attachment protein. We found that strains encapsidating σ1 with higher efficiency bind cells more avidly and replicate and spread more efficiently in mice relative to those encapsidating σ1 with lower efficiency. These results highlight a function for σ1 attachment protein capsid abundance in viral replication in cells and animals, which may inform future use of reovirus as an oncolytic therapeutic.
Collapse
Affiliation(s)
- Olivia L. Welsh
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alexa N. Roth
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Danica M. Sutherland
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Terence S. Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Cristi F, Walters M, Narayan N, Agopsowicz K, Hitt MM, Shmulevitz M. Improved oncolytic activity of a reovirus mutant that displays enhanced virus spread due to reduced cell attachment. Mol Ther Oncolytics 2023; 31:100743. [PMID: 38033400 PMCID: PMC10685048 DOI: 10.1016/j.omto.2023.100743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Wild-type reovirus serotype 3 Dearing (T3wt), a non-pathogenic intestinal virus, has shown promise as a cancer therapy in clinical trials, but it would benefit from an increased potency. Given that T3wt is naturally adapted to the intestinal environment (rather than tumors), we genetically modified reovirus to improve its infectivity in cancer cells. Various reovirus mutants were created, and their oncolytic potency was evaluated in vitro using plaque size as a measure of virus fitness in cancer cells. Notably, Super Virus 5 (SV5), carrying five oncolytic mutations, displayed the largest plaques in breast cancer cells among the mutants tested, indicating the potential for enhancing oncolytic potency through the combination of mutations. Furthermore, in a HER2+ murine breast cancer model, mice treated with SV5 exhibited superior tumor reduction and increased survival compared with those treated with PBS or T3wt. Intriguingly, SV5 did not replicate faster than T3wt in cultured cells but demonstrated a farther spread relative to T3wt, attributed to its reduced attachment to cancer cells. These findings highlight the significance of increased virus spread as a crucial mechanism for improving oncolytic virus activity. Thus, genetic modifications of reovirus hold the potential for augmenting its efficacy in cancer therapy.
Collapse
Affiliation(s)
- Francisca Cristi
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton AB T6G 2E1, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton AB T6G 2E1, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton AB T6G 2E1, Canada
| | - Maiah Walters
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton AB T6G 2E1, Canada
| | - Nashae Narayan
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton AB T6G 2E1, Canada
| | - Kate Agopsowicz
- Department of Oncology, University of Alberta, Edmonton AB T6G 1Z2, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton AB T6G 2E1, Canada
| | - Mary M. Hitt
- Department of Oncology, University of Alberta, Edmonton AB T6G 1Z2, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton AB T6G 2E1, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton AB T6G 2E1, Canada
| | - Maya Shmulevitz
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton AB T6G 2E1, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton AB T6G 2E1, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton AB T6G 2E1, Canada
| |
Collapse
|
5
|
Letafati A, Ardekani OS, Naderisemiromi M, Fazeli MM, Jemezghani NA, Yavarian J. Oncolytic viruses against cancer, promising or delusion? Med Oncol 2023; 40:246. [PMID: 37458862 DOI: 10.1007/s12032-023-02106-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 06/23/2023] [Indexed: 07/20/2023]
Abstract
Cancer treatment is one of the most challenging topics in medical sciences. Different methods such as chemotherapy, tumor surgery, and immune checkpoint inhibitors therapy (ICIs) are potential approaches to treating cancer and killing tumor cells, but clinical studies have shown that they have been successful for a limited group of patients. Using viruses as a treatment can be considered as an effective treatment in the field of medicine. This is considered as a potential treatment, especially in comparison to chemotherapy, which has severe side effects related to the immune system. Most oncolytic viruses (OVs) have the potential to multiply in cancer cells, which are more than normal cells in malignant tissue and can induce immune responses. Therefore, tons of efforts and research have been started on the utilization of OVs as a treatment for cancer and have shown promising in treating cancers with less side effects. In this article, we have gathered studies about oncolytic viruses and their effectiveness in cancer treatment.Please confirm if the author names are presented accurately and in the correct sequence (given name, middle name/initial, family name). Author 1 Given name: [Omid Salahi] Last name [Ardekani], Author 2 Given name: [Mohammad Mehdi] Last name [Fazeli], Author 3 Given name: [Nillofar Asadi] Last name [Jemezghani]. Also, kindly confirm the details in the metadata are correct.Confirmed.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Salahi Ardekani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Mina Naderisemiromi
- Department of Immunology, Faculty of Medicine and Health, The University of Manchester, Manchester, UK
| | - Mohammad Mehdi Fazeli
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | | | - Jila Yavarian
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Sutherland DM, Strebl M, Koehler M, Welsh OL, Yu X, Hu L, dos Santos Natividade R, Knowlton JJ, Taylor GM, Moreno RA, Wörz P, Lonergan ZR, Aravamudhan P, Guzman-Cardozo C, Kour S, Pandey UB, Alsteens D, Wang Z, Prasad BVV, Stehle T, Dermody TS. NgR1 binding to reovirus reveals an unusual bivalent interaction and a new viral attachment protein. Proc Natl Acad Sci U S A 2023; 120:e2219404120. [PMID: 37276413 PMCID: PMC10268256 DOI: 10.1073/pnas.2219404120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/19/2023] [Indexed: 06/07/2023] Open
Abstract
Nogo-66 receptor 1 (NgR1) binds a variety of structurally dissimilar ligands in the adult central nervous system to inhibit axon extension. Disruption of ligand binding to NgR1 and subsequent signaling can improve neuron outgrowth, making NgR1 an important therapeutic target for diverse neurological conditions such as spinal crush injuries and Alzheimer's disease. Human NgR1 serves as a receptor for mammalian orthoreovirus (reovirus), but the mechanism of virus-receptor engagement is unknown. To elucidate how NgR1 mediates cell binding and entry of reovirus, we defined the affinity of interaction between virus and receptor, determined the structure of the virus-receptor complex, and identified residues in the receptor required for virus binding and infection. These studies revealed that central NgR1 surfaces form a bridge between two copies of viral capsid protein σ3, establishing that σ3 serves as a receptor ligand for reovirus. This unusual binding interface produces high-avidity interactions between virus and receptor to prime early entry steps. These studies refine models of reovirus cell-attachment and highlight the evolution of viruses to engage multiple receptors using distinct capsid components.
Collapse
Affiliation(s)
- Danica M. Sutherland
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Michael Strebl
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076Tübingen, Germany
| | - Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Olivia L. Welsh
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Xinzhe Yu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Rita dos Santos Natividade
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Jonathan J. Knowlton
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Cryo-Electron Microscopy and Tomography Core, Baylor College of Medicine, Houston, TX77030
| | - Gwen M. Taylor
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Rodolfo A. Moreno
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Patrick Wörz
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076Tübingen, Germany
| | - Zachery R. Lonergan
- Cryo-Electron Microscopy and Tomography Core, Baylor College of Medicine, Houston, TX77030
| | - Pavithra Aravamudhan
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Camila Guzman-Cardozo
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Sukhleen Kour
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
| | - Udai Bhan Pandey
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN37232
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA15261
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
- Children’s Neuroscience Institute, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Zhao Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
- Walloon Excellence in Life Sciences and Biotechnology, 1300Wavre, Belgium
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| | - B. V. Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX77030
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076Tübingen, Germany
| | - Terence S. Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA15219
| |
Collapse
|
7
|
Wang M, Wang X, Jin X, Zhou J, Zhang Y, Yang Y, Liu Y, Zhang J. Cell-based and cell-free immunotherapies for glioblastoma: current status and future directions. Front Immunol 2023; 14:1175118. [PMID: 37304305 PMCID: PMC10248152 DOI: 10.3389/fimmu.2023.1175118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
Abstract
Glioblastoma (GBM) is among the most fatal and recurring malignant solid tumors. It arises from the GBM stem cell population. Conventional neurosurgical resection, temozolomide (TMZ)-dependent chemotherapy and radiotherapy have rendered the prognosis of patients unsatisfactory. Radiotherapy and chemotherapy can frequently induce non-specific damage to healthy brain and other tissues, which can be extremely hazardous. There is therefore a pressing need for a more effective treatment strategy for GBM to complement or replace existing treatment options. Cell-based and cell-free immunotherapies are currently being investigated to develop new treatment modalities against cancer. These treatments have the potential to be both selective and successful in minimizing off-target collateral harm in the normal brain. In this review, several aspects of cell-based and cell-free immunotherapies related to GBM will be discussed.
Collapse
Affiliation(s)
- Mingming Wang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Xiaojie Wang
- Basic Medical School, Shenyang Medical College, Shenyang, Liaoning, China
| | - Xiaoyan Jin
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Jingjing Zhou
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yufu Zhang
- Department of Hepatobiliary Surgery, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Yiyuan Yang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yusi Liu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Jing Zhang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| |
Collapse
|
8
|
dos Santos Natividade R, Koehler M, Gomes PSFC, Simpson JD, Smith SC, Gomes DEB, de Lhoneux J, Yang J, Ray A, Dermody TS, Bernardi RC, Ogden KM, Alsteens D. Deciphering molecular mechanisms stabilizing the reovirus-binding complex. Proc Natl Acad Sci U S A 2023; 120:e2220741120. [PMID: 37186838 PMCID: PMC10214207 DOI: 10.1073/pnas.2220741120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Mammalian orthoreoviruses (reoviruses) serve as potential triggers of celiac disease and have oncolytic properties, making these viruses potential cancer therapeutics. Primary attachment of reovirus to host cells is mainly mediated by the trimeric viral protein, σ1, which engages cell-surface glycans, followed by high-affinity binding to junctional adhesion molecule-A (JAM-A). This multistep process is thought to be accompanied by major conformational changes in σ1, but direct evidence is lacking. By combining biophysical, molecular, and simulation approaches, we define how viral capsid protein mechanics influence virus-binding capacity and infectivity. Single-virus force spectroscopy experiments corroborated by in silico simulations show that GM2 increases the affinity of σ1 for JAM-A by providing a more stable contact interface. We demonstrate that conformational changes in σ1 that lead to an extended rigid conformation also significantly increase avidity for JAM-A. Although its associated lower flexibility impairs multivalent cell attachment, our findings suggest that diminished σ1 flexibility enhances infectivity, indicating that fine-tuning of σ1 conformational changes is required to successfully initiate infection. Understanding properties underlying the nanomechanics of viral attachment proteins offers perspectives in the development of antiviral drugs and improved oncolytic vectors.
Collapse
Affiliation(s)
- Rita dos Santos Natividade
- Louvain Institute of Biomolecular Science and Technology, NanoBiophysics lab, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology, NanoBiophysics lab, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, 85354Freising, Germany
| | | | - Joshua D. Simpson
- Louvain Institute of Biomolecular Science and Technology, NanoBiophysics lab, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Sydni Caet Smith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 37232Nashville, TN
| | | | - Juliette de Lhoneux
- Louvain Institute of Biomolecular Science and Technology, NanoBiophysics lab, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Jinsung Yang
- Louvain Institute of Biomolecular Science and Technology, NanoBiophysics lab, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Ankita Ray
- Louvain Institute of Biomolecular Science and Technology, NanoBiophysics lab, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Terence S. Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15213
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA15213
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center, Children’s Hospital of Pittsburgh, Pittsburgh, PA15213
| | | | - Kristen M. Ogden
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 37232Nashville, TN
- Department of Pediatrics, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN37232
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, NanoBiophysics lab, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
- Walloon Excellence in Life sciences and Biotechnology, Walloon Excellence Research Institute, 1300Wavre, Belgium
| |
Collapse
|
9
|
Jandick NA, Kirner N, Miller CL. Mammalian orthoreovirus infection in human epidermal growth factor receptor 2 positive (HER2+) breast cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540250. [PMID: 37214868 PMCID: PMC10197616 DOI: 10.1101/2023.05.10.540250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Mammalian orthoreovirus (MRV) is a clinically benign oncolytic virus which has been investigated for use in multiple cancer types, including breast cancer (BC). In human clinical trials, MRV has been shown to be safe, and multiple BC patients have shown partial responses to intratumoral and intravenous virus delivery. Combination therapies inclusive of MRV and current FDA approved BC chemotherapies are being investigated to target metastatic, early BC, and triple negative BC. Though MRV is being tested clinically, we still do not fully understand the highly variable patient responses to MRV therapy. One of the most aggressive BC subtypes is HER2+ BC, in which human epidermal growth factor receptor 2 (HER2) is dysregulated, resulting in increased growth, survival, and metastasis of cancer cells. FDA approved therapies, trastuzumab and pertuzumab, target HER2 to prevent signaling of the phosphoinositide 3-kinase (PI3K) pathway. However, recent findings show that accumulation of hypoxia inducible factor-1 alpha (HIF-1α) in HER2+ BC cells contributes to trastuzumab resistance. In this work, we provide evidence that MRV infects, replicates in, and kills HER2 overexpressing cells. MRV infection is also found to have variable effects on signaling pathways that activate or are activated by HER2 expression. Finally, we show that MRV reduces HIF-1α accumulation in all the cell lines tested, including a HER2+ BC cell line. These studies provide further evidence that MRV holds promise for use in conjunction with trastuzumab to treat HER2+ BC patients.
Collapse
|
10
|
van den Wollenberg DJM, Kemp V, Rabelink MJWE, Hoeben RC. Reovirus Type 3 Dearing Variants Do Not Induce Necroptosis in RIPK3-Expressing Human Tumor Cell Lines. Int J Mol Sci 2023; 24:ijms24032320. [PMID: 36768641 PMCID: PMC9916669 DOI: 10.3390/ijms24032320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Reoviruses are used as oncolytic viruses to destroy tumor cells. The concomitant induction of anti-tumor immune responses enhances the efficacy of therapy in tumors with low amounts of immune infiltrates before treatment. The reoviruses should provoke immunogenic cell death (ICD) to stimulate a tumor cell-directed immune response. Necroptosis is considered a major form of ICD, and involves receptor-interacting protein kinase 1 (RIPK1), RIPK3 and phosphorylation of mixed-lineage kinase domain-like protein (MLKL). This leads to cell membrane disintegration and the release of damage-associated molecular patterns that can activate immune responses. Reovirus Type 3 Dearing (T3D) can induce necroptosis in mouse L929 fibroblast cells and mouse embryonic fibroblasts. Most human tumor cell lines have a defect in RIPK3 expression and consequently fail to induce necroptosis as measured by MLKL phosphorylation. We used the human colorectal adenocarcinoma HT29 cell line as a model to study necroptosis in human cells since this cell line has frequently been described in necroptosis-related studies. To stimulate MLKL phosphorylation and induce necroptosis, HT29 cells were treated with a cocktail consisting of TNFα, the SMAC mimetic BV6, and the caspase inhibitor Z-VAD-FMK. While this treatment induced necroptosis, three different reovirus T3D variants, i.e., the plasmid-based reverse genetics generated virus (T3DK), the wild-type reovirus T3D isolate R124, and the junction adhesion molecule-A-independent reovirus mutant (jin-1) failed to induce necroptosis in HT29 cells. In contrast, these viruses induced MLKL phosphorylation in murine L929 cells, albeit with varying efficiencies. Our study shows that while reoviruses efficiently induce necroptosis in L929 cells, this is not a common phenotype in human cell lines. This study emphasizes the difficulties of translating the results of ICD studies from murine cells to human cells.
Collapse
|
11
|
Cristi F, Gutiérrez T, Hitt MM, Shmulevitz M. Genetic Modifications That Expand Oncolytic Virus Potency. Front Mol Biosci 2022; 9:831091. [PMID: 35155581 PMCID: PMC8826539 DOI: 10.3389/fmolb.2022.831091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/06/2022] [Indexed: 12/20/2022] Open
Abstract
Oncolytic viruses (OVs) are a promising type of cancer therapy since they selectively replicate in tumor cells without damaging healthy cells. Many oncolytic viruses have progressed to human clinical trials, however, their performance as monotherapy has not been as successful as expected. Importantly, recent literature suggests that the oncolytic potential of these viruses can be further increased by genetically modifying the viruses. In this review, we describe genetic modifications to OVs that improve their ability to kill tumor cells directly, to dismantle the tumor microenvironment, or to alter tumor cell signaling and enhance anti-tumor immunity. These advances are particularly important to increase virus spread and reduce metastasis, as demonstrated in animal models. Since metastasis is the principal cause of mortality in cancer patients, having OVs designed to target metastases could transform cancer therapy. The genetic alterations reported to date are only the beginning of all possible improvements to OVs. Modifications described here could be combined together, targeting multiple processes, or with other non-viral therapies with potential to provide a strong and lasting anti-tumor response in cancer patients.
Collapse
Affiliation(s)
- Francisca Cristi
- Shmulevitz Laboratory, Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Tomás Gutiérrez
- Goping Laboratory, Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mary M. Hitt
- Hitt Laboratory, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Mary M. Hitt, ; Maya Shmulevitz,
| | - Maya Shmulevitz
- Shmulevitz Laboratory, Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Mary M. Hitt, ; Maya Shmulevitz,
| |
Collapse
|
12
|
Schuelke MR, Gundelach JH, Coffey M, West E, Scott K, Johnson DR, Samson A, Melcher A, Vile RG, Bram RJ. Phase I trial of sargramostim/pelareorep therapy in pediatric patients with recurrent or refractory high-grade brain tumors. Neurooncol Adv 2022; 4:vdac085. [PMID: 35821679 PMCID: PMC9268737 DOI: 10.1093/noajnl/vdac085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Brain tumors are the leading cause of cancer death for pediatric patients. Pelareorep, an immunomodulatory oncolytic reovirus, has intravenous efficacy in preclinical glioma models when preconditioned with GM-CSF (sargramostim). We report a phase I trial with the primary goal of evaluating the safety of sargramostim/pelareorep in pediatric patients with recurrent or refractory high-grade brain tumors and a secondary goal of characterizing immunologic responses. Methods The trial was open to pediatric patients with recurrent or refractory high-grade brain tumors (3 + 3 cohort design). Each cycle included 3 days of subcutaneous sargramostim followed by 2 days of intravenous pelareorep. Laboratory studies and imaging were acquired upon recruitment and periodically thereafter. Results Six patients participated, including three glioblastoma, two diffuse intrinsic pontine glioma, and one medulloblastoma. Two pelareorep dose levels of 3 × 108 and 5 × 108 tissue culture infectious dose 50 (TCID50) were assessed. One patient experienced a dose limiting toxicity of persistent hyponatremia. Common low-grade (1 or 2) adverse events included transient fatigue, hypocalcemia, fever, flu-like symptoms, thrombocytopenia, and leukopenia. High-grade (3 or 4) adverse events included neutropenia, lymphopenia, leukopenia, hypophosphatemia, depressed level of consciousness, and confusion. All patients progressed on therapy after a median of 32.5 days and died a median of 108 days after recruitment. Imaging at progression did not show evidence of pseudoprogression or inflammation. Correlative assays revealed transient but consistent changes in immune cells across patients. Conclusions Sargramostim/pelareorep was administered to pediatric patients with recurrent or refractory high-grade brain tumors. Hyponatremia was the only dose limiting toxicity (DLT), though maximum tolerated dose (MTD) was not determined.
Collapse
Affiliation(s)
- Matthew R Schuelke
- Medical Scientist Training Program, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Matt Coffey
- Oncolytics Biotech, Calgary, Alberta, Canada
| | - Emma West
- Faculty of Medicine and Health, Leeds Institute of Medical Research, University of Leeds, St James' University Hospital, Leeds, UK
| | - Karen Scott
- Faculty of Medicine and Health, Leeds Institute of Medical Research, University of Leeds, St James' University Hospital, Leeds, UK
| | - Derek R Johnson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Adel Samson
- Faculty of Medicine and Health, Leeds Institute of Medical Research, University of Leeds, St James' University Hospital, Leeds, UK
| | - Alan Melcher
- The Institute of Cancer Research/Royal Marsden, National Institute for Health Research Biomedical Research Centre, London, UK
| | - Richard G Vile
- Faculty of Medicine and Health, Leeds Institute of Medical Research, University of Leeds, St James' University Hospital, Leeds, UK
| | - Richard J Bram
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
13
|
Kim SY, Maitra R, Goel S. Multimodal immune activation abilities and characteristics of reovirus. Am J Transl Res 2021; 13:14176-14185. [PMID: 35035763 PMCID: PMC8748157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/28/2021] [Indexed: 06/14/2023]
Abstract
Reovirus is a ubiquitous, non-pathogenic, double stranded RNA virus with anti-tumor properties. The virus's replicative potential is regulated by phosphorylation of protein kinase receptor (PKR). In cancers with RAS pathway activation which leads to dysregulation of PKR, the virus maintains its protein translational potential and induces oncolysis. Systemic chemotherapy remains the standard of care for metastatic colorectal cancer with the addition of biologic agents in KRAS wildtype subtypes. In KRAS mutant colorectal cancers, there has been no added benefit to biologic agents. The therapeutic potential of reovirus (Reolysin®, pelareorep, Oncolytic Inc., Calgary, Canada), which induces its oncolysis with RAS activation through multimodal immune mechanisms, has been demonstrated in preclinical and clinical studies. In this review, we outline the specific immune mechanisms of reovirus induced oncolysis and provide both preclinical and clinical data on its applications in metastatic colorectal cancer patients.
Collapse
Affiliation(s)
- So Yeon Kim
- Montefiore Medical Center1695 Eastchester Road, Bronx, New York 10461, USA
| | - Radhashree Maitra
- Department of Biology, Yeshiva University500 West 185th Street, New York 10033, USA
| | - Sanjay Goel
- Montefiore Medical Center1695 Eastchester Road, Bronx, New York 10461, USA
- Albert Einstein College of Medicine1300 Morris Park Ave, Bronx, New York 10461, USA
| |
Collapse
|
14
|
The reovirus μ2 C-terminal loop inversely regulates NTPase and transcription functions versus binding to factory-forming μNS and promotes replication in tumorigenic cells. J Virol 2021; 95:JVI.02006-20. [PMID: 33658345 PMCID: PMC8139653 DOI: 10.1128/jvi.02006-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Wild type reovirus serotype 3 'Dearing PL strain' (T3wt) is being heavily evaluated as an oncolytic and immunotherapeutic treatment for cancers. Mutations that promote reovirus entry into tumor cells were previously reported to enhance oncolysis; herein we aimed to discover mutations that enhance the post-entry steps of reovirus infection in tumor cells. Using directed evolution, we identified that reovirus variant T3v10M1 exhibited enhanced replication relative to T3wt on a panel of cancer cells. T3v10M1 contains an alanine-to-valine substitution (A612V) in the core-associated μ2, which was previously found to have NTPase activities in virions and to facilitate virus factory formation by association with μNS. Paradoxically, the A612V mutation in μ2 from T3v10M1 was discovered to impair NTPase activities and RNA synthesis, leading to five-fold higher probability of abortive infection for T3v10M1 relative to T3wt. The A612V mutation resides in a previously uncharacterized C-terminal region that juxtaposes the template entry site of the polymerase μ2; our findings thus support an important role for this domain during virus transcription. Despite crippled onset of infection, T3v10M1 exhibited greater accumulation of viral proteins and progeny during replication, leading to increased overall virus burst size. Both Far-Western and co-immunoprecipitation approaches corroborated that the A612V mutation in μ2 increased association with the non-structural virus protein μNS and enhances burst size. Altogether the data supports that mutations in the C-terminal loop domain of μ2 inversely regulate NTPase and RNA synthesis versus interactions with μNS, but with a net gain of replication in tumorigenic cells.SIGNIFICANCEReovirus is a model system for understanding virus replication but also a clinically relevant virus for cancer therapy. We identified the first mutation that increases reovirus infection in tumorigenic cells by enhancing post-entry stages of reovirus replication. The mutation is in a previously uncharacterized c-terminal region of the M1-derived μ2 protein, which we demonstrated affects multiple functions of μ2; NTPase, RNA synthesis, inhibition of antiviral immune response and association with the virus replication factory-forming μNS protein. These findings promote a mechanistic understanding of viral protein functions. In the future, the benefits of μ2 mutations may be useful for enhancing reovirus potency in tumors.
Collapse
|
15
|
Oncolytic Virotherapy for Cancer: Clinical Experience. Biomedicines 2021; 9:biomedicines9040419. [PMID: 33924556 PMCID: PMC8069290 DOI: 10.3390/biomedicines9040419] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/22/2022] Open
Abstract
Oncolytic viruses are a new class of therapeutics which are largely in the experimental stage, with just one virus approved by the FDA thus far. While the concept of oncolytic virotherapy is not new, advancements in the fields of molecular biology and virology have renewed the interest in using viruses as oncolytic agents. Backed by robust preclinical data, many oncolytic viruses have entered clinical trials. Oncolytic viruses that have completed some levels of clinical trials or are currently undergoing clinical trials are mostly genetically engineered viruses, with the exception of some RNA viruses. Reolysin, an unmodified RNA virus is clinically the most advanced oncolytic RNA virus that has completed different phases of clinical trials. Other oncolytic viruses that have been studied in clinical trials are mostly DNA viruses that belong to one of the three families: herpesviridae, poxviridae or adenoviridae. In this review work we discuss recent clinical studies with oncolytic viruses, especially herpesvirus, poxvirus, adenovirus and reovirus. In summary, the oncolytic viruses tested so far are well tolerated, even in immune-suppressed patients. For most oncolytic viruses, mild and acceptable toxicities are seen at the currently defined highest feasible doses. However, anti-tumor efficacies of oncolytic viruses have been modest, especially when used as monotherapy. Therefore, the potency of oncolytic viruses needs to be enhanced for more oncolytic viruses to hit the clinic. Aiming to achieve higher therapeutic benefits, oncolytic viruses are currently being studied in combination with other therapies. Here we discuss the currently available clinical data on oncolytic viruses, either as monotherapy or in combination with other treatments.
Collapse
|
16
|
Hromic-Jahjefendic A, Lundstrom K. Viral Vector-Based Melanoma Gene Therapy. Biomedicines 2020; 8:E60. [PMID: 32187995 PMCID: PMC7148454 DOI: 10.3390/biomedicines8030060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023] Open
Abstract
Gene therapy applications of oncolytic viruses represent an attractive alternative for cancer treatment. A broad range of oncolytic viruses, including adenoviruses, adeno-associated viruses, alphaviruses, herpes simplex viruses, retroviruses, lentiviruses, rhabdoviruses, reoviruses, measles virus, Newcastle disease virus, picornaviruses and poxviruses, have been used in diverse preclinical and clinical studies for the treatment of various diseases, including colon, head-and-neck, prostate and breast cancer as well as squamous cell carcinoma and glioma. The majority of studies have focused on immunotherapy and several drugs based on viral vectors have been approved. However, gene therapy for malignant melanoma based on viral vectors has not been utilized to its full potential yet. This review represents a summary of the achievements of preclinical and clinical studies using viral vectors, with the focus on malignant melanoma.
Collapse
Affiliation(s)
- Altijana Hromic-Jahjefendic
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | | |
Collapse
|
17
|
Giacomantonio MA, Sterea AM, Kim Y, Paulo JA, Clements DR, Kennedy BE, Bydoun MJ, Shi G, Waisman DM, Gygi SP, Giacomantonio CA, Murphy JP, Gujar S. Quantitative Proteome Responses to Oncolytic Reovirus in GM-CSF- and M-CSF-Differentiated Bone Marrow-Derived Cells. J Proteome Res 2020; 19:708-718. [PMID: 31884793 PMCID: PMC7294930 DOI: 10.1021/acs.jproteome.9b00583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The efficacy of oncolytic viruses (OVs), such as reovirus, is dictated by host immune responses, including those mediated by the pro- versus anti-inflammatory macrophages. As such, a detailed understanding of the interaction between reovirus and different macrophage types is critical for therapeutic efficacy. To explore reovirus-macrophage interactions, we performed tandem mass tag (TMT)-based quantitative temporal proteomics on mouse bone marrow-derived macrophages (BMMs) generated with two cytokines, macrophage colony stimulating factor (M-CSF) and granulocytic-macrophage colony stimulating factor (GM-CSF), representing anti- and proinflammatory macrophages, respectively. We quantified 6863 proteins across five time points in duplicate, comparing M-CSF (M-BMM) and GM-CSF (GM-BMM) in response to OV. We find that GM-BMMs have lower expression of key intrinsic proteins that facilitate an antiviral immune response, express higher levels of reovirus receptor protein JAM-A, and are more susceptible to oncolytic reovirus infection compared to M-BMMs. Interestingly, although M-BMMs are less susceptible to reovirus infection and subsequent cell death, they initiate an antireovirus adaptive T cell immune response comparable to that of GM-BMMs. Taken together, these data describe distinct proteome differences between these two macrophage populations in terms of their ability to mount antiviral immune responses.
Collapse
Affiliation(s)
| | - Andra M Sterea
- Department of Microbiology and Immunology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
| | - Youra Kim
- Department of Pathology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
| | - Joao A Paulo
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115-5730 , United States
| | - Derek R Clements
- Department of Pathology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
| | - Barry E Kennedy
- Department of Pathology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
| | - Moamen J Bydoun
- Department of Pathology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
| | - Ge Shi
- Department of Pathology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
| | - David M Waisman
- Department of Pathology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
- Department of Biochemistry & Molecular Biology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
| | - Steven P Gygi
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115-5730 , United States
| | - Carman A Giacomantonio
- Department of Pathology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
- Department of Surgery , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
| | - J Patrick Murphy
- Department of Pathology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
- Department of Biology , University of Prince Edward Island , Room 443, Duffy Science Centre, 550 University Avenue , Charlottetown , Prince Edward Island C1A 4P3 , Canada
| | - Shashi Gujar
- Department of Pathology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
- Department of Microbiology and Immunology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
- Beatrice Hunter Cancer Research Institute , Halifax , Nova Scotia B3H 4R2 , Canada
- Department of Biology , Dalhousie University , Halifax , Nova Scotia B3H 4R2 , Canada
| |
Collapse
|
18
|
Igase M, Shibutani S, Kurogouchi Y, Fujiki N, Hwang CC, Coffey M, Noguchi S, Nemoto Y, Mizuno T. Combination Therapy with Reovirus and ATM Inhibitor Enhances Cell Death and Virus Replication in Canine Melanoma. Mol Ther Oncolytics 2019; 15:49-59. [PMID: 31650025 PMCID: PMC6804779 DOI: 10.1016/j.omto.2019.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 08/16/2019] [Indexed: 12/13/2022] Open
Abstract
Oncolytic virotherapy using reovirus is a promising new anti-cancer treatment with potential for use in humans and dogs. Because reovirus monotherapy shows limited efficacy in human and canine cancer patients, the clinical development of a combination therapy is necessary. To identify candidate components of such a combination, we screened a 285-compound drug library for those that enhanced reovirus cytotoxicity in a canine melanoma cell line. Here, we show that exposure to an inhibitor of the ataxia telangiectasia mutated protein (ATM) enhances the oncolytic potential of reovirus in five of six tested canine melanoma cell lines. Specifically, the ATM inhibitor potentiated reovirus replication in cancer cells along with promoting the lysosomal activity, resulting in an increased proportion of caspase-dependent apoptosis and cell cycle arrest at G2/M compared to those observed with reovirus alone. Overall, our study suggests that the combination of reovirus and the ATM inhibitor may be an attractive option in cancer therapy.
Collapse
Affiliation(s)
- Masaya Igase
- Laboratory of Molecular Diagnostics and Therapeutics, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Shusaku Shibutani
- Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yosuke Kurogouchi
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Noriyuki Fujiki
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Chung Chew Hwang
- Laboratory of Molecular Diagnostics and Therapeutics, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Matt Coffey
- Oncolytics Biotech Inc., Calgary, AB, Canada
| | - Shunsuke Noguchi
- Laboratory of Veterinary Radiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Yuki Nemoto
- Laboratory of Molecular Diagnostics and Therapeutics, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Takuya Mizuno
- Laboratory of Molecular Diagnostics and Therapeutics, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
19
|
Breast Tumor-Associated Metalloproteases Restrict Reovirus Oncolysis by Cleaving the σ1 Cell Attachment Protein and Can Be Overcome by Mutation of σ1. J Virol 2019; 93:JVI.01380-19. [PMID: 31462562 DOI: 10.1128/jvi.01380-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 01/01/2023] Open
Abstract
Reovirus is undergoing clinical testing as an oncolytic therapy for breast cancer. Given that reovirus naturally evolved to thrive in enteric environments, we sought to better understand how breast tumor microenvironments impinge on reovirus infection. Reovirus was treated with extracellular extracts generated from polyomavirus middle T-antigen-derived mouse breast tumors. Unexpectedly, these breast tumor extracellular extracts inactivated reovirus, reducing infectivity of reovirus particles by 100-fold. Mechanistically, inactivation was attributed to proteolytic cleavage of the viral cell attachment protein σ1, which diminished virus binding to sialic acid (SA)-low tumor cells. Among various specific protease class inhibitors and metal ions, EDTA and ZnCl2 effectively modulated σ1 cleavage, indicating that breast tumor-associated zinc-dependent metalloproteases are responsible for reovirus inactivation. Moreover, media from MCF7, MB468, MD-MB-231, and HS578T breast cancer cell lines recapitulated σ1 cleavage and reovirus inactivation, suggesting that inactivation of reovirus is shared among mouse and human breast cancers and that breast cancer cells by themselves can be a source of reovirus-inactivating proteases. Binding assays and quantification of SA levels on a panel of cancer cells showed that truncated σ1 reduced virus binding to cells with low surface SA. To overcome this restriction, we generated a reovirus mutant with a mutation (T249I) in σ1 that prevents σ1 cleavage and inactivation by breast tumor-associated proteases. The mutant reovirus showed similar replication kinetics in tumorigenic cells, toxicity equivalent to that of wild-type reovirus in a severely compromised mouse model, and increased tumor titers. Overall, the data show that tumor microenvironments have the potential to reduce infectivity of reovirus.IMPORTANCE We demonstrate that metalloproteases in breast tumor microenvironments can inactivate reovirus. Our findings expose that tumor microenvironment proteases could have a negative impact on proteinaceous cancer therapies, such as reovirus, and that modification of such therapies to circumvent inactivation by tumor metalloproteases merits consideration.
Collapse
|
20
|
CSV2018: The 2nd Symposium of the Canadian Society for Virology. Viruses 2019; 11:v11010079. [PMID: 30669273 PMCID: PMC6356965 DOI: 10.3390/v11010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 11/16/2022] Open
Abstract
The 2nd Symposium of the Canadian Society for Virology (CSV2018) was held in June 2018 in Halifax, Nova Scotia, Canada, as a featured event marking the 200th anniversary of Dalhousie University. CSV2018 attracted 175 attendees from across Canada and around the world, more than double the number that attended the first CSV symposium two years earlier. CSV2018 provided a forum to discuss a wide range of topics in virology including human, veterinary, plant, and microbial pathogens. Invited keynote speakers included David Kelvin (Dalhousie University and Shantou University Medical College) who provided a historical perspective on influenza on the 100th anniversary of the 1918 pandemic; Sylvain Moineau (Université Laval) who described CRISPR-Cas systems and anti-CRISPR proteins in warfare between bacteriophages and their host microbes; and Kate O’Brien (then from Johns Hopkins University, now relocated to the World Health Organization where she is Director of Immunization, Vaccines and Biologicals), who discussed the underlying viral etiology for pneumonia in the developing world, and the evidence for respiratory syncytial virus (RSV) as a primary cause. Reflecting a strong commitment of Canadian virologists to science communication, CSV2018 featured the launch of Halifax’s first annual Soapbox Science event to enable public engagement with female scientists, and the live-taping of the 499th episode of the This Week in Virology (TWIV) podcast, hosted by Vincent Racaniello (Columbia University) and science writer Alan Dove. TWIV featured interviews of CSV co-founders Nathalie Grandvaux (Université de Montréal) and Craig McCormick (Dalhousie University), who discussed the origins and objectives of the new society; Ryan Noyce (University of Alberta), who discussed technical and ethical considerations of synthetic virology; and Kate O’Brien, who discussed vaccines and global health. Finally, because CSV seeks to provide a better future for the next generation of Canadian virologists, the symposium featured a large number of oral and poster presentations from trainees and closed with the awarding of presentation prizes to trainees, followed by a tour of the Halifax Citadel National Historic Site and an evening of entertainment at the historic Alexander Keith’s Brewery.
Collapse
|
21
|
Systemically Administered Reovirus-Induced Downregulation of Hypoxia Inducible Factor-1α in Subcutaneous Tumors. MOLECULAR THERAPY-ONCOLYTICS 2018; 12:162-172. [PMID: 30788427 PMCID: PMC6369106 DOI: 10.1016/j.omto.2018.12.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/26/2018] [Indexed: 12/21/2022]
Abstract
Reovirus, which possesses a 10-segmented double-stranded RNA genome, mediates superior antitumor effects via not only virus replication in a tumor cell-specific manner but also other mechanisms distinct from virus replication. Several groups, including ours, reported the reovirus-mediated downregulation of hypoxia inducible factor-1α (HIF-1α) following infection in cultured tumor cells; however, it remained to be clarified whether reovirus downregulates the expression of HIF-1α and its target genes in tumor-bearing hosts. We found that reovirus induced significant downregulation of protein levels of HIF-1α and its target genes in the subcutaneous tumors at 120 h post-systemic administration. Expression of reovirus capsid protein σ3 was found in the pimonidazole-positive hypoxic area in the tumor. Significant levels of tumor cell apoptosis were not found in the tumors of reovirus-treated mice at this time point, suggesting that reovirus-mediated tumor cell killing did not largely contribute to the downregulation of HIF-1α protein levels in the tumors. UV-inactivated reovirus did not induce downregulation of HIF-1α expression in the tumors, indicating that virus replication was indispensable for downregulation of HIF-1α expression in the subcutaneous tumors. This study provides important information for the development of reovirus-mediated virotherapy against various types of tumors.
Collapse
|
22
|
Reovirus Neurotropism and Virulence Are Dictated by Sequences in the Head Domain of the Viral Attachment Protein. J Virol 2018; 92:JVI.00974-18. [PMID: 30209169 DOI: 10.1128/jvi.00974-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/04/2018] [Indexed: 11/20/2022] Open
Abstract
Viral capsid components that bind cellular receptors mediate critical functions in viral tropism and disease pathogenesis. Mammalian orthoreoviruses (reoviruses) spread systemically in newborn mice to cause serotype-specific disease in the central nervous system (CNS). Serotype 1 (T1) reovirus infects ependymal cells to cause nonlethal hydrocephalus, whereas serotype 3 (T3) reovirus infects neurons to cause fulminant and lethal encephalitis. This serotype-dependent difference in tropism and concomitant disease is attributed to the σ1 viral attachment protein, which is composed of head, body, and tail domains. To identify σ1 sequences that contribute to tropism for specific cell types in the CNS, we engineered a panel of viruses expressing chimeric σ1 proteins in which discrete σ1 domains have been reciprocally exchanged. Parental and chimeric σ1 viruses were compared for replication, tropism, and disease induction following intracranial inoculation of newborn mice. Viruses expressing T1 σ1 head sequences infect the ependyma, produce relatively lower titers in the brain, and do not cause significant disease. In contrast, viruses expressing T3 σ1 head sequences efficiently infect neurons, replicate to relatively higher titers in the brain, and cause a lethal encephalitis. Additionally, T3 σ1 head-expressing viruses display enhanced infectivity of cultured primary cortical neurons compared with T1 σ1 head-expressing viruses. These results indicate that T3 σ1 head domain sequences promote infection of neurons, likely by interaction with a neuron-specific receptor, and dictate tropism in the CNS and induction of encephalitis.IMPORTANCE Viral encephalitis is a serious and often life-threatening inflammation of the brain. Mammalian orthoreoviruses are promising oncolytic therapeutics for humans but establish virulent, serotype-dependent disease in the central nervous system (CNS) of many young mammals. Serotype 1 reoviruses infect ependymal cells and produce hydrocephalus, whereas serotype 3 reoviruses infect neurons and cause encephalitis. Reovirus neurotropism is hypothesized to be dictated by the filamentous σ1 viral attachment protein. However, it is not apparent how this protein mediates disease. We discovered that sequences forming the most virion-distal domain of T1 and T3 σ1 coordinate infection of either ependyma or neurons, respectively, leading to mutually exclusive patterns of tropism and disease in the CNS. These studies contribute new knowledge about how reoviruses target cells for infection in the brain and inform the rational design of improved oncolytic therapies to mitigate difficult-to-treat tumors of the CNS.
Collapse
|
23
|
Thadi A, Khalili M, Morano WF, Richard SD, Katz SC, Bowne WB. Early Investigations and Recent Advances in Intraperitoneal Immunotherapy for Peritoneal Metastasis. Vaccines (Basel) 2018; 6:E54. [PMID: 30103457 PMCID: PMC6160982 DOI: 10.3390/vaccines6030054] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 12/23/2022] Open
Abstract
Peritoneal metastasis (PM) is an advanced stage malignancy largely refractory to modern therapy. Intraperitoneal (IP) immunotherapy offers a novel approach for the control of regional disease of the peritoneal cavity by breaking immune tolerance. These strategies include heightening T-cell response and vaccine induction of anti-cancer memory against tumor-associated antigens. Early investigations with chimeric antigen receptor T cells (CAR-T cells), vaccine-based therapies, dendritic cells (DCs) in combination with pro-inflammatory cytokines and natural killer cells (NKs), adoptive cell transfer, and immune checkpoint inhibitors represent significant advances in the treatment of PM. IP delivery of CAR-T cells has shown demonstrable suppression of tumors expressing carcinoembryonic antigen. This response was enhanced when IP injected CAR-T cells were combined with anti-PD-L1 or anti-Gr1. Similarly, CAR-T cells against folate receptor α expressing tumors improved T-cell tumor localization and survival when combined with CD137 co-stimulatory signaling. Moreover, IP immunotherapy with catumaxomab, a trifunctional antibody approved in Europe, targets epithelial cell adhesion molecule (EpCAM) and has shown considerable promise with control of malignant ascites. Herein, we discuss immunologic approaches under investigation for treatment of PM.
Collapse
Affiliation(s)
- Anusha Thadi
- Department of Surgery, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| | - Marian Khalili
- Department of Surgery, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| | - William F Morano
- Department of Surgery, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| | - Scott D Richard
- Department of Obstetrics and Gynecology, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA.
| | - Steven C Katz
- Department of Surgery, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Wilbur B Bowne
- Department of Surgery, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| |
Collapse
|
24
|
Mammalian Orthoreovirus Factories Modulate Stress Granule Protein Localization by Interaction with G3BP1. J Virol 2017; 91:JVI.01298-17. [PMID: 28794026 DOI: 10.1128/jvi.01298-17] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 08/03/2017] [Indexed: 01/28/2023] Open
Abstract
Mammalian orthoreovirus (MRV) infection induces phosphorylation of translation initiation factor eIF2α, which promotes the formation of discrete cytoplasmic inclusions, termed stress granules (SGs). SGs are emerging as a component of the innate immune response to virus infection, and modulation of SG assembly is a common mechanism employed by viruses to counter this antiviral response. We previously showed that MRV infection induces SGs early and then interferes with SG formation as infection proceeds. In this work, we found that SG-associated proteins localized to the periphery of virus-encoded cytoplasmic structures, termed virus factories (VFs), where viral transcription, translation, and replication occur. The localization of SG proteins to VFs was dependent on polysome dissociation and occurred via association of the SG effector protein, Ras-GAP SH3-binding protein 1 (G3BP1), with the MRV nonstructural protein σNS, which localizes to VFs via association with VF nucleating protein, μNS. Deletion analysis of the σNS RNA binding domain and G3BP1 RNA (RRM) and ribosomal (RGG) binding domains showed that σNS association and VF localization phenotypes of G3BP1 do not occur solely through RNA or ribosomal binding but require both the RRM and RGG domains of G3BP1 for maximal viral-factory-like structure (VFL) localization and σNS association. Coexpression of σNS and μNS resulted in disruption of normal SG puncta, and in cells lacking G3BP1, MRV replication was enhanced in a manner correlating with strain-dependent induction of host translation shutoff. These results suggest that σNS association with G3BP1 and relocalization of G3BP1 to the VF periphery play roles in SG disruption to facilitate MRV replication in the host translational shutoff environment.IMPORTANCE SGs and SG effector proteins have emerged as important, yet poorly understood, players in the host's innate immune response to virus infection. MRV infection induces SGs early during infection that are dispersed and/or prevented from forming during late stages of infection despite continued activation of the eIF2α signaling pathway. Cellular and viral components involved in disruption of SGs during late stages of MRV infection remain to be elucidated. This work provides evidence that MRV disruption of SGs may be facilitated by association of the MRV nonstructural protein σNS with the major SG effector protein G3BP1 and subsequent localization of G3BP1 and other SG-associated proteins around the peripheries of virus-encoded factories, interrupting the normal formation of SGs. Our findings also reveal the importance of G3BP1 as an inhibitor of MRV replication during infection for the first time.
Collapse
|
25
|
Zhao X, Ouyang W, Chester C, Long S, Wang N, He Z. Cytokine-induced killer cell delivery enhances the antitumor activity of oncolytic reovirus. PLoS One 2017; 12:e0184816. [PMID: 28922411 PMCID: PMC5602626 DOI: 10.1371/journal.pone.0184816] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 08/31/2017] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OV) have recently emerged as a promising therapeutic modality in cancer treatment. OV selectively infect and kill tumor cells, while sparing untransformed cells. The direct cytotoxic effects combined with the capacity to trigger an immune response make OV an appealing combination partner in the burgeoning field of cancer immunotherapy. One of the leading OV therapeutic candidates is the double-stranded RNA virus reovirus. In order to improve the oncolytic activity of reovirus and allow for systemic administration despite the prevalence of neutralizing antibodies, cytokine-induced killer (CIK) cells were explored as cell carriers for reovirus delivery. In this study, CIK cells were successfully loaded with reovirus ex vivo, and viral replication was limited in CIK cells. Confocal microscopy and flow cytometry demonstrated that CIK cells retained reovirus on the surface. Moreover, CIK cells could promote reovirus infection of tumor cells in the presence of neutralizing antibodies; meanwhile, cytotoxicity of CIK cells was increased after loading with reovirus. These findings support further investigation of reovirus and CIK combination for antitumor therapy.
Collapse
Affiliation(s)
- Xing Zhao
- Stem Cell and Tissue Engineering Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Weiwei Ouyang
- Department of Thoracic Oncology, Affiliated Hospital of Guizhou Medical University, and Guizhou Cancer Hospital, Guiyang, Guizhou, China
| | - Cariad Chester
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, United States of America
| | - Shiqi Long
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Nianxue Wang
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhixu He
- Stem Cell and Tissue Engineering Research Center, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
26
|
Clements DR, Murphy JP, Sterea A, Kennedy BE, Kim Y, Helson E, Almasi S, Holay N, Konda P, Paulo JA, Sharif T, Lee PW, Weekes MP, Gygi SP, Gujar S. Quantitative Temporal in Vivo Proteomics Deciphers the Transition of Virus-Driven Myeloid Cells into M2 Macrophages. J Proteome Res 2017; 16:3391-3406. [PMID: 28768414 PMCID: PMC5648240 DOI: 10.1021/acs.jproteome.7b00425] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
![]()
Myeloid
cells play a central role in the context of viral eradication,
yet precisely how these cells differentiate throughout the course
of acute infections is poorly understood. In this study, we have developed
a novel quantitative temporal in vivo proteomics (QTiPs) platform
to capture proteomic signatures of temporally transitioning virus-driven
myeloid cells directly in situ, thus taking into consideration host–virus
interactions throughout the course of an infection. QTiPs, in combination
with phenotypic, functional, and metabolic analyses, elucidated a
pivotal role for inflammatory CD11b+, Ly6G–, Ly6Chigh-low cells in antiviral immune response and
viral clearance. Most importantly, the time-resolved QTiPs data set
showed the transition of CD11b+, Ly6G–, Ly6Chigh-low cells into M2-like macrophages, which displayed
increased antigen-presentation capacities and bioenergetic demands
late in infection. We elucidated the pivotal role of myeloid cells
in virus clearance and show how these cells phenotypically, functionally,
and metabolically undergo a timely transition from inflammatory to
M2-like macrophages in vivo. With respect to the growing appreciation
for in vivo examination of viral–host interactions and for
the role of myeloid cells, this study elucidates the use of quantitative
proteomics to reveal the role and response of distinct immune cell
populations throughout the course of virus infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Erin Helson
- Faculty of Medicine, University of Toronto , Toronto, Ontario M5S 1A8, Canada
| | | | | | | | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | | | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge , Cambridge CB2 0XY, United Kingdom
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Shashi Gujar
- Centre for Innovative and Collaborative Health Systems Research, IWK Health Centre , Halifax, Nova Scotia B3K 6R8, Canada
| |
Collapse
|
27
|
Orzechowska BU, Jędryka M, Zwolińska K, Matkowski R. VSV based virotherapy in ovarian cancer: the past, the present and …future? J Cancer 2017; 8:2369-2383. [PMID: 28819441 PMCID: PMC5560156 DOI: 10.7150/jca.19473] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
The standard approach to treating patients with advanced epithelial ovarian cancer (EOC) after primary debulking surgery remains taxane and platinum-based chemotherapy. Despite treatment with this strategy, the vast majority of patients relapse and develop drug-resistant metastatic disease that may be driven by cancer stem cells (CSCs) or cancer initiating cells (CICs). Oncolytic viruses circumvent typical drug-resistance mechanisms, therefore they may provide a safe and effective alternative treatment for chemotherapy-resistant CSCs/CICs. Among oncolytic viruses vesicular stomatitis virus (VSV) has demonstrated oncolysis and preferential replication in cancer cells. In this review, we summarize the recent findings regarding existing knowledge on biology of the ovarian cancer and the role of ovarian CSCs (OCSCs) in tumor dissemination and chemoresistance. In addition we also present an overview of recent advances in ovarian cancer therapies with oncolytic viruses (OV). We focus particularly on key genetic or immune response pathways involved in tumorigenesis in ovarian cancer which facilitate oncolytic activity of vesicular stomatitis virus (VSV). We highlight the prospects of targeting OCSCs with VSV. The importance of testing an emerging ovarian cancer animal models and ovarian cancer cell culture conditions influencing oncolytic efficacy of VSV is also addressed.
Collapse
Affiliation(s)
- Beata Urszula Orzechowska
- Laboratory of Virology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Marcin Jędryka
- Division of Surgical Oncology, Gynaecological Oncology, Chemotherapy and Department of Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wrocław, Poland
- Lower Silesian Oncology Centre, Wroclaw, Plac Hirszfelda 12, 53-413 Wrocław, Poland
| | - Katarzyna Zwolińska
- Laboratory of Virology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Rafał Matkowski
- Division of Surgical Oncology, Gynaecological Oncology, Chemotherapy and Department of Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wrocław, Poland
- Lower Silesian Oncology Centre, Wroclaw, Plac Hirszfelda 12, 53-413 Wrocław, Poland
| |
Collapse
|
28
|
Kanai Y, Kobayashi T. [A plasmid-based reverse genetics system for rotaviruses]. Uirusu 2017; 67:99-110. [PMID: 30369541 DOI: 10.2222/jsv.67.99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Rotavirus (RV), a non-enveloped icosahedral virus containing eleven gene segments of double-stranded RNA, is the leading cause of severe, acute diarrhea among infants and young children worldwide. Safe and effective rotavirus vaccines have been available since 2006, and have markedly reduced the number of deaths by severe gastroenteritis. However, rotaviruses are still responsible for approximately 200,000 deaths annually worldwide. Reverse genetics systems for the manipulation of viral genomes are a powerful approach for studying viral replication and pathogenesis, and for developing vaccines and viral vectors. The understanding of the molecular mechanisms underlying RV pathogenesis, or development of next generation vaccines, has been hampered by the lack of a complete reverse genetics system. Recently, we developed a novel reverse genetics system which enabled recovery of recombinant RVs entirely from cloned cDNAs. This new strategy requires co-expression of a small transmembrane protein that accelerates cell-to-cell fusion and vaccinia virus capping enzyme. In this review, the strategies and history of the development of reverse genetics systems for the family Reoviridae are described.
Collapse
Affiliation(s)
- Yuta Kanai
- Department of Virology, Research Institute for Microbial Diseases, Osaka University
| | - Takeshi Kobayashi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University
| |
Collapse
|
29
|
Boudreault S, Martenon-Brodeur C, Caron M, Garant JM, Tremblay MP, Armero VES, Durand M, Lapointe E, Thibault P, Tremblay-Létourneau M, Perreault JP, Scott MS, Lemay G, Bisaillon M. Global Profiling of the Cellular Alternative RNA Splicing Landscape during Virus-Host Interactions. PLoS One 2016; 11:e0161914. [PMID: 27598998 PMCID: PMC5012649 DOI: 10.1371/journal.pone.0161914] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/15/2016] [Indexed: 12/13/2022] Open
Abstract
Alternative splicing (AS) is a central mechanism of genetic regulation which modifies the sequence of RNA transcripts in higher eukaryotes. AS has been shown to increase both the variability and diversity of the cellular proteome by changing the composition of resulting proteins through differential choice of exons to be included in mature mRNAs. In the present study, alterations to the global RNA splicing landscape of cellular genes upon viral infection were investigated using mammalian reovirus as a model. Our study provides the first comprehensive portrait of global changes in the RNA splicing signatures that occur in eukaryotic cells following infection with a human virus. We identify 240 modified alternative splicing events upon infection which belong to transcripts frequently involved in the regulation of gene expression and RNA metabolism. Using mass spectrometry, we also confirm modifications to transcript-specific peptides resulting from AS in virus-infected cells. These findings provide additional insights into the complexity of virus-host interactions as these splice variants expand proteome diversity and function during viral infection.
Collapse
Affiliation(s)
- Simon Boudreault
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Camille Martenon-Brodeur
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Marie Caron
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Jean-Michel Garant
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Marie-Pier Tremblay
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Victoria E. S. Armero
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Mathieu Durand
- Laboratoire de Génomique Fonctionnelle, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Elvy Lapointe
- Laboratoire de Génomique Fonctionnelle, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Philippe Thibault
- Laboratoire de Génomique Fonctionnelle, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Maude Tremblay-Létourneau
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Jean-Pierre Perreault
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Michelle S. Scott
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
| | - Guy Lemay
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montreal, Quebec, H3C 3J7, Canada
| | - Martin Bisaillon
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, J1E 4K8, Canada
- * E-mail:
| |
Collapse
|
30
|
Kemp V, Hoeben RC, van den Wollenberg DJM. Exploring Reovirus Plasticity for Improving Its Use as Oncolytic Virus. Viruses 2015; 8:E4. [PMID: 26712782 PMCID: PMC4728564 DOI: 10.3390/v8010004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/04/2015] [Accepted: 12/15/2015] [Indexed: 12/13/2022] Open
Abstract
Reoviruses are non-enveloped viruses with a segmented double stranded RNA genome. In humans, they are not associated with serious disease. Human reoviruses exhibit an inherent preference to replicate in tumor cells, which makes them ideally suited for use in oncolytic virotherapies. Their use as anti-cancer agent has been evaluated in several clinical trials, which revealed that intra-tumoral and systemic delivery of reoviruses are well tolerated. Despite evidence of anti-tumor effects, the efficacy of reovirus in anti-cancer monotherapy needs to be further enhanced. The opportunity to treat both the primary tumor as well as metastases makes systemic delivery a preferred administration route. Several pre-clinical studies have been conducted to address the various hurdles connected to systemic delivery of reoviruses. The majority of those studies have been done in tumor-bearing immune-deficient murine models. This thwarts studies on the impact of the contribution of the immune system to the tumor cell eradication. This review focuses on key aspects of the reovirus/host-cell interactions and the methods that are available to modify the virus to alter these interactions. These aspects are discussed with a focus on improving the reovirus' antitumor efficacy.
Collapse
Affiliation(s)
- Vera Kemp
- Department of Molecular Cell Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| | - Rob C Hoeben
- Department of Molecular Cell Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| | - Diana J M van den Wollenberg
- Department of Molecular Cell Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| |
Collapse
|
31
|
Binz E, Lauer UM. Chemovirotherapy: combining chemotherapeutic treatment with oncolytic virotherapy. Oncolytic Virother 2015; 4:39-48. [PMID: 27512669 PMCID: PMC4918378 DOI: 10.2147/ov.s54780] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Oncolytic virotherapy has made significant progress in recent years, however, widespread approval of virotherapeutics is still limited. Primarily, this is due to the fact that currently available virotherapeutics are mostly tested in monotherapeutic clinical trials exclusively (ie, not in combination with other therapies) and so far have achieved only small and often clinically insignificant responses. Given that the predominantly immunotherapeutic mechanism of virotherapeutics is somewhat time-dependent and rapidly growing tumors therefore exhibit only minor chances of being captured in time, scenarios with combination partners are postulated to be more effective. Combinatory settings would help to achieve a rapid stabilization or even reduction of onset tumor masses while providing enough time (numerous months) for achieving immuno(viro)therapeutic success. For this reason, combination strategies of virotherapy with highly genotoxic regimens, such as chemotherapy, are of major interest. A number of clinical trials bringing the concepts of chemotherapy and virotherapy together have previously been undertaken, but optimal scheduling of chemovirotherapy (maximizing the anti-tumor effect while minimizing the risk of overlapping toxicity) still constitutes a major challenge. Therefore, an overview of published as well as ongoing Phase I–III trials should improve our understanding of current challenges and future developments in this field.
Collapse
Affiliation(s)
- Eike Binz
- Department of Internal Medicine, Division of Hepatology, Gastroenterology, Infectiology, Medical University Hospital, Tuebingen, Germany
| | - Ulrich M Lauer
- Department of Internal Medicine, Division of Hepatology, Gastroenterology, Infectiology, Medical University Hospital, Tuebingen, Germany
| |
Collapse
|