1
|
Soroush A, Shahhosseini R, Ghavamikia N, Hjazi A, Roudaki S, KhalatbariLimaki M, Mirbolouk M, Pakmehr S, Karimi P. Improvement of current immunotherapies with engineered oncolytic viruses that target cancer stem cells. Cell Biochem Funct 2024; 42:e4055. [PMID: 38856033 DOI: 10.1002/cbf.4055] [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: 02/26/2024] [Revised: 05/04/2024] [Accepted: 05/12/2024] [Indexed: 06/11/2024]
Abstract
The heterogeneity of the solid tumor microenvironment (TME) impairs the therapeutic efficacy of standard therapies and also reduces the infiltration of antitumor immune cells, all of which lead to tumor progression and invasion. In addition, self-renewing cancer stem cells (CSCs) support tumor dormancy, drug resistance, and recurrence, all of which might pose challenges to the eradication of malignant tumor masses with current therapies. Natural forms of oncolytic viruses (OVs) or engineered OVs are known for their potential to directly target and kill tumor cells or indirectly eradicate tumor cells by involving antitumor immune responses, including enhancement of infiltrating antitumor immune cells, induction of immunogenic cell death, and reprogramming of cold TME to an immune-sensitive hot state. More importantly, OVs can target stemness factors that promote tumor progression, which subsequently enhances the efficacy of immunotherapies targeting solid tumors, particularly the CSC subpopulation. Herein, we describe the role of CSCs in tumor heterogeneity and resistance and then highlight the potential and remaining challenges of immunotherapies targeting CSCs. We then review the potential of OVs to improve tumor immunogenicity and target CSCs and finally summarize the challenges within the therapeutic application of OVs in preclinical and clinical trials.
Collapse
Affiliation(s)
| | | | - Nima Ghavamikia
- Cardiovascular Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin AbdulAziz University, Al-Kharj, Saudi Arabia
| | - Shahrzad Roudaki
- School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mahtab Mirbolouk
- School of Pharmacy, Cyprus International University, Nicosia, North Cyprus
| | | | - Parvin Karimi
- Fars Population-Based Cancer Registry, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Clinical activity of single-dose systemic oncolytic VSV virotherapy in patients with relapsed refractory T-cell lymphoma. Blood Adv 2022; 6:3268-3279. [PMID: 35175355 PMCID: PMC9198941 DOI: 10.1182/bloodadvances.2021006631] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/25/2022] [Indexed: 12/14/2022] Open
Abstract
Clinical success with intravenous (IV) oncolytic virotherapy (OV) has to-date been anecdotal. We conducted a phase 1 clinical trial of systemic OV and investigated the mechanisms of action in responding patients. A single IV dose of vesicular stomatitis virus (VSV) interferon-β (IFN-β) with sodium iodide symporter (NIS) was administered to patients with relapsed/refractory hematologic malignancies to determine safety and efficacy across 4 dose levels (DLs). Correlative studies were undertaken to evaluate viremia, virus shedding, virus replication, and immune responses. Fifteen patients received VSV-IFNβ-NIS. Three patients were treated at DL1 through DL3 (0.05, 0.17, and 0.5 × 1011 TCID50), and 6 were treated at DL4 (1.7 × 1011 TCID50) with no dose-limiting toxicities. Three of 7 patients with T-cell lymphoma (TCL) had responses: a 3-month partial response (PR) at DL2, a 6-month PR, and a complete response (CR) ongoing at 20 months at DL4. Viremia peaked at the end of infusion, g was detected. Plasma IFN-β, a biomarker of VSV-IFNβ-NIS replication, peaked between 4 hours and 48 hours after infusion. The patient with CR had robust viral replication with increased plasma cell-free DNA, high peak IFN-β of 18 213 pg/mL, a strong anti-VSV neutralizing antibody response, and increased numbers of tumor reactive T-cells. VSV-IFNβ-NIS as a single agent was effective in patients with TCL, resulting in durable disease remissions in heavily pretreated patients. Correlative analyses suggest that responses may be due to a combination of direct oncolytic tumor destruction and immune-mediated tumor control. This trial is registered at www.clinicaltrials.gov as #NCT03017820.
Collapse
|
3
|
Ikeda-Imafuku M, Wang LLW, Rodrigues D, Shaha S, Zhao Z, Mitragotri S. Strategies to improve the EPR effect: A mechanistic perspective and clinical translation. J Control Release 2022; 345:512-536. [PMID: 35337939 DOI: 10.1016/j.jconrel.2022.03.043] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
Many efforts have been made to achieve targeted delivery of anticancer drugs to enhance their efficacy and to reduce their adverse effects. These efforts include the development of nanomedicines as they can selectively penetrate through tumor blood vessels through the enhanced permeability and retention (EPR) effect. The EPR effect was first proposed by Maeda and co-workers in 1986, and since then various types of nanoparticles have been developed to take advantage of the phenomenon with regards to drug delivery. However, the EPR effect has been found to be highly variable and thus unreliable due to the complex tumor microenvironment. Various physical and pharmacological strategies have been explored to overcome this challenge. Here, we review key advances and emerging concepts of such EPR-enhancing strategies. Furthermore, we analyze 723 clinical trials of nanoparticles with EPR enhancers and discuss their clinical translation.
Collapse
Affiliation(s)
- Mayumi Ikeda-Imafuku
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Danika Rodrigues
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Suyog Shaha
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA; Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612, USA.
| | - Samir Mitragotri
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA.
| |
Collapse
|
4
|
Hong B, Sahu U, Mullarkey MP, Kaur B. Replication and Spread of Oncolytic Herpes Simplex Virus in Solid Tumors. Viruses 2022; 14:v14010118. [PMID: 35062322 PMCID: PMC8778098 DOI: 10.3390/v14010118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 12/11/2022] Open
Abstract
Oncolytic herpes simplex virus (oHSV) is a highly promising treatment for solid tumors. Intense research and development efforts have led to first-in-class approval for an oHSV for melanoma, but barriers to this promising therapy still exist that limit efficacy. The process of infection, replication and transmission of oHSV in solid tumors is key to obtaining a good lytic destruction of infected cancer cells to kill tumor cells and release tumor antigens that can prime anti-tumor efficacy. Intracellular tumor cell signaling and tumor stromal cells present multiple barriers that resist oHSV activity. Here, we provide a review focused on oncolytic HSV and the essential viral genes that allow for virus replication and spread in order to gain insight into how manipulation of these pathways can be exploited to potentiate oHSV infection and replication among tumor cells.
Collapse
|
5
|
Batalla-Covello J, Ngai HW, Flores L, McDonald M, Hyde C, Gonzaga J, Hammad M, Gutova M, Portnow J, Synold T, Curiel DT, Lesniak MS, Aboody KS, Mooney R. Multiple Treatment Cycles of Neural Stem Cell Delivered Oncolytic Adenovirus for the Treatment of Glioblastoma. Cancers (Basel) 2021; 13:6320. [PMID: 34944938 PMCID: PMC8699772 DOI: 10.3390/cancers13246320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/09/2021] [Indexed: 11/16/2022] Open
Abstract
Tumor tropic neural stem cells (NSCs) can improve the anti-tumor efficacy of oncovirotherapy agents by protecting them from rapid clearance by the immune system and delivering them to multiple distant tumor sites. We recently completed a first-in-human trial assessing the safety of a single intracerebral dose of NSC-delivered CRAd-Survivin-pk7 (NSC.CRAd-S-pk7) combined with radiation and chemotherapy in newly diagnosed high-grade glioma patients. The maximum feasible dose was determined to be 150 million NSC.CRAd-Sp-k7 (1.875 × 1011 viral particles). Higher doses were not assessed due to volume limitations for intracerebral administration and the inability to further concentrate the study agent. It is possible that therapeutic efficacy could be maximized by administering even higher doses. Here, we report IND-enabling studies in which an improvement in treatment efficacy is achieved in immunocompetent mice by administering multiple treatment cycles intracerebrally. The results imply that pre-existing immunity does not preclude therapeutic benefits attainable by administering multiple rounds of an oncolytic adenovirus directly into the brain.
Collapse
Affiliation(s)
- Jennifer Batalla-Covello
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Hoi Wa Ngai
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Linda Flores
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Marisa McDonald
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Caitlyn Hyde
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Joanna Gonzaga
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Mohamed Hammad
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Margarita Gutova
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Jana Portnow
- Department of Medical Oncology, City of Hope, Duarte, CA 91010, USA;
| | - Tim Synold
- Department of Cancer Biology, City of Hope, Duarte, CA 91010, USA;
| | - David T. Curiel
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA;
| | - Karen S. Aboody
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Rachael Mooney
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| |
Collapse
|
6
|
Cook J, Acosta-Medina AA, Peng KW, Lacy M, Russell S. Oncolytic virotherapy - Forging its place in the immunomodulatory paradigm for Multiple Myeloma. Cancer Treat Res Commun 2021; 29:100473. [PMID: 34673439 DOI: 10.1016/j.ctarc.2021.100473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/25/2021] [Indexed: 12/23/2022]
Abstract
The treatment focus for multiple myeloma (MM) has recently pivoted towards immune modulating strategies, with T-cell redirection therapies currently at the forefront of drug development. Yet, despite this revolution in treatment, MM remains without a sustainable cure. At the same time, tremendous advancement has been made in recombinant and gene editing techniques for oncolytic viruses (OV), which have increased their tumor specificity, improved safety, and enhanced the oncolytic and immunostimulatory potential. These breakthrough developments in oncolytic virotherapy have opened new avenues for OVs to be used in combination with other immune-based therapies such as checkpoint inhibitors, chimeric antigen receptor T-cells (CAR-T) and bispecific T-cell engagers. In this review, the authors place the spotlight on systemic oncolytic virotherapy as an adaptable immunotherapeutic for MM, highlight the unique mechanism of OVs in activating the immune-suppressive marrow microenvironment, and lastly showcase the OV platforms and the promising combination strategies in the pipeline for MM.
Collapse
Affiliation(s)
- Joselle Cook
- Division of Hematology, Mayo Clinic, Rochester MN, United States.
| | | | - Kah Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester MN , United States
| | - Martha Lacy
- Division of Hematology, Mayo Clinic, Rochester MN, United States
| | - Stephen Russell
- Division of Hematology, Mayo Clinic, Rochester MN, United States; Department of Molecular Medicine, Mayo Clinic, Rochester MN , United States
| |
Collapse
|
7
|
ABO blood group antigen therapy: a potential new strategy against solid tumors. Sci Rep 2021; 11:16241. [PMID: 34376742 PMCID: PMC8355358 DOI: 10.1038/s41598-021-95794-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 07/29/2021] [Indexed: 11/17/2022] Open
Abstract
The economic burden of tumors is increasing, so there is an urgent need to develop new therapies for their treatment. Killing tumors by activating complement is an effective strategy for the treatment. We used the ABO blood group system and the corresponding antibodies to activate the killer cell capacity of the complement system. After the construction of a mouse model containing blood group A antibodies and inoculating colorectal cancer and breast cancer cells into the axillae of the mice, intratumoural injection using a lentivirus carrying a blood group antigen as a drug significantly reduced the tumor volume of the mice. Compared with the control group, the content of the C5b-9 complement membrane attack complex in the tumors of mice treated with the blood group A antigen was significantly increased, and the proportion of NK cells was also significantly increased. In vitro cell-based experiments proved that tumor cells expressing blood group A antigens showed significantly inhibited cell proliferation when added to serum containing blood group A antibodies. These results all prove that the ABO blood group antigen may become a powerful tool for the treatment of tumors in patients.
Collapse
|
8
|
Gomes-Santos IL, Amoozgar Z, Kumar AS, Ho WW, Roh K, Talele NP, Curtis H, Kawaguchi K, Jain RK, Fukumura D. Exercise Training Improves Tumor Control by Increasing CD8 + T-cell Infiltration via CXCR3 Signaling and Sensitizes Breast Cancer to Immune Checkpoint Blockade. Cancer Immunol Res 2021; 9:765-778. [PMID: 33839688 PMCID: PMC8295193 DOI: 10.1158/2326-6066.cir-20-0499] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/23/2020] [Accepted: 03/31/2021] [Indexed: 11/16/2022]
Abstract
The mechanisms behind the antitumor effects of exercise training (ExTr) are not fully understood. Using mouse models of established breast cancer, we examined here the causal role of CD8+ T cells in the benefit acquired from ExTr in tumor control, as well as the ability of ExTr to improve immunotherapy responses. We implanted E0771, EMT6, MMTV-PyMT, and MCa-M3C breast cancer cells orthotopically in wild-type or Cxcr3-/- female mice and initiated intensity-controlled ExTr sessions when tumors reached approximately 100 mm3 We characterized the tumor microenvironment (TME) using flow cytometry, transcriptome analysis, proteome array, ELISA, and immunohistochemistry. We used antibodies against CD8+ T cells for cell depletion. Treatment with immune checkpoint blockade (ICB) consisted of anti-PD-1 alone or in combination with anti-CTLA-4. ExTr delayed tumor growth and induced vessel normalization, demonstrated by increased pericyte coverage and perfusion and by decreased hypoxia. ExTr boosted CD8+ T-cell infiltration, with enhanced effector function. CD8+ T-cell depletion prevented the antitumor effect of ExTr. The recruitment of CD8+ T cells and the antitumor effects of ExTr were abrogated in Cxcr3-/- mice, supporting the causal role of the CXCL9/CXCL11-CXCR3 pathway. ExTr also sensitized ICB-refractory breast cancers to treatment. Our results indicate that ExTr can normalize the tumor vasculature, reprogram the immune TME, and enhance the antitumor activity mediated by CD8+ T cells via CXCR3, boosting ICB responses. Our findings and mechanistic insights provide a rationale for the clinical translation of ExTr to improve immunotherapy of breast cancer.
Collapse
Affiliation(s)
- Igor L Gomes-Santos
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ashwin S Kumar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - William W Ho
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kangsan Roh
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nilesh P Talele
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hannah Curtis
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kosuke Kawaguchi
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
9
|
Kwan A, Winder N, Muthana M. Oncolytic Virotherapy Treatment of Breast Cancer: Barriers and Recent Advances. Viruses 2021; 13:1128. [PMID: 34208264 PMCID: PMC8230950 DOI: 10.3390/v13061128] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Oncolytic virotherapy (OV) is an emerging class of immunotherapeutic drugs. Their mechanism of action is two-fold: direct cell lysis and unmasking of the cancer through immunogenic cell death, which allows the immune system to recognize and eradicate tumours. Breast cancer is the most common cancer in women and is challenging to treat with immunotherapy modalities because it is classically an immunogenically "cold" tumour type. This provides an attractive niche for OV, given viruses have been shown to turn "cold" tumours "hot," thereby opening a plethora of treatment opportunities. There has been a number of pre-clinical attempts to explore the use of OV in breast cancer; however, these have not led to any meaningful clinical trials. This review considers both the potential and the barriers to OV in breast cancer, namely, the limitations of monotherapy and the scope for combination therapy, improving viral delivery and challenges specific to the breast cancer population (e.g., tumour subtype, menopausal status, age).
Collapse
Affiliation(s)
| | | | - Munitta Muthana
- Department of Oncology and Metabolism, University of Sheffield Medical School, Beech Hill Road, Sheffield S10 2RX, UK; (A.K.); (N.W.)
| |
Collapse
|
10
|
Farrera-Sal M, Moya-Borrego L, Bazan-Peregrino M, Alemany R. Evolving Status of Clinical Immunotherapy with Oncolytic Adenovirus. Clin Cancer Res 2021; 27:2979-2988. [PMID: 33526422 DOI: 10.1158/1078-0432.ccr-20-1565] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/02/2020] [Accepted: 01/15/2021] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapy targeting immune checkpoint inhibitors shows efficacy in several human cancers, but "cold tumors" that lack immune cells are typically unresponsive. Among the potential therapeutic approaches that could "heat" or promote lymphocyte infiltration of cold tumors, oncolytic viruses have attracted interest for their lytic and immunogenic mechanisms of action. In this article, we review the use of oncolytic adenoviruses in cancer immunotherapy, with a particular focus on preclinical and clinical data of oncolytic adenovirus-triggered immune responses against tumor antigens. We also discuss parameters to consider in clinical trial design and the combination of oncolytic adenoviruses with conventional treatments or other immunotherapies.
Collapse
Affiliation(s)
- Martí Farrera-Sal
- ProCure Program, IDIBELL-Institut Català d'Oncologia, Barcelona, Spain.,VCN Biosciences SL, Barcelona, Spain
| | | | | | - Ramon Alemany
- ProCure Program, IDIBELL-Institut Català d'Oncologia, Barcelona, Spain.
| |
Collapse
|
11
|
Zhou Z, Tian J, Zhang W, Xiang W, Ming Y, Chen L, Zhou J. Multiple strategies to improve the therapeutic efficacy of oncolytic herpes simplex virus in the treatment of glioblastoma. Oncol Lett 2021; 22:510. [PMID: 33986870 DOI: 10.3892/ol.2021.12771] [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: 10/22/2020] [Accepted: 03/29/2021] [Indexed: 11/06/2022] Open
Abstract
Oncolytic viruses have attracted widespread attention as biological anticancer agents that can selectively kill tumor cells without affecting normal cells. Although progress has been made in therapeutic strategies, the prognosis of patients with glioblastoma (GBM) remains poor and no ideal treatment approach has been developed. Recently, oncolytic herpes simplex virus (oHSV) has been considered a promising novel treatment approach for GBM. However, the therapeutic efficacy of oHSV in GBM, with its intricate pathophysiology, remains unsatisfactory due to several obstacles, such as limited replication and attenuated potency of oHSV owing to deletions or mutations in virulence genes, and ineffective delivery of the therapeutic virus. Multiple strategies have attempted to identify the optimal strategy for the successful clinical application of oHSV. Several preclinical trials have demonstrated that engineering novel oHSVs, developing combination therapies and improving methods for delivering oHSV to tumor cells seem to hold promise for improving the efficacy of this virotherapy.
Collapse
Affiliation(s)
- Zhengjun Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Junjie Tian
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Wenyan Zhang
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Wei Xiang
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Yang Ming
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Ligang Chen
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan 646000, P.R. China.,Neurological Diseases and Brain Function Laboratory, Luzhou, Sichuan 646000, P.R. China
| | - Jie Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan 646000, P.R. China.,Neurological Diseases and Brain Function Laboratory, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
12
|
Yang C, Hua N, Xie S, Wu Y, Zhu L, Wang S, Tong X. Oncolytic viruses as a promising therapeutic strategy for hematological malignancies. Biomed Pharmacother 2021; 139:111573. [PMID: 33894623 DOI: 10.1016/j.biopha.2021.111573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/23/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
Abstract
The incidence of hematological malignancies such as multiple myeloma, leukemia, and lymphoma has increased over time. Although bone marrow transplantation, immunotherapy and chemotherapy have led to significant improvements in efficacy, poor prognosis in elderly patients, recurrence and high mortality among hematological malignancies remain major challenges, and innovative therapeutic strategies should be explored. Besides directly lyse tumor cells, oncolytic viruses can activate immune responses or be engineered to express therapeutic factors to increase antitumor efficacy, and have gradually been recognized as an appealing approach for fighting cancers. An increasing number of studies have applied oncolytic viruses in hematological malignancies and made progress. In particular, strategies combining immunotherapy and oncolytic virotherapy are emerging. Various phase I clinical trials of oncolytic reovirus with lenalidomide or programmed death 1(PD-1) immune checkpoint inhibitors in multiple myeloma are ongoing. Moreover, preclinical studies of combinations with chimeric antigen receptor T (CAR-T) cells are underway. Thus, oncolytic virotherapy is expected to be a promising approach to cure hematological malignancies. This review summarizes progress in oncolytic virus research in hematological malignancies. After briefly reviewing the development and oncolytic mechanism of oncolytic viruses, we focus on delivery methods of oncolytic viruses, especially systemic delivery that is suitable for hematological tumors. We then discuss the main types of oncolytic viruses applied for hematological malignancies and related clinical trials. In addition, we present several ways to improve the antitumor efficacy of oncolytic viruses. Finally, we discuss current challenges and provide suggestions for future studies.
Collapse
Affiliation(s)
- Chen Yang
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; Department of Clinical Medicine, Qingdao University, Qingdao, PR China
| | - Nanni Hua
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Shufang Xie
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Yi Wu
- Phase I clinical research center, Zhejiang Provincial People's Hospital,Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China
| | - Lifeng Zhu
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China
| | - Shibing Wang
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital ,Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China.
| | - Xiangmin Tong
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital ,Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China.
| |
Collapse
|
13
|
Grundy M, Bau L, Hill C, Paverd C, Mannaris C, Kwan J, Crake C, Coviello C, Coussios C, Carlisle R. Improved therapeutic antibody delivery to xenograft tumors using cavitation nucleated by gas-entrapping nanoparticles. Nanomedicine (Lond) 2021; 16:37-50. [PMID: 33426913 DOI: 10.2217/nnm-2020-0263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aims: Testing ultrasound-mediated cavitation for enhanced delivery of the therapeutic antibody cetuximab to tumors in a mouse model. Methods: Tumors with strong EGF receptor expression were grown bilaterally. Cetuximab was coadministered intravenously with cavitation nuclei, consisting of either the ultrasound contrast agent Sonovue or gas-stabilizing nanoscale SonoTran Particles. One of the two tumors was exposed to focused ultrasound. Passive acoustic mapping localized and monitored cavitation activity. Both tumors were then excised and cetuximab concentration was quantified. Results: Cavitation increased tumoral cetuximab concentration. When nucleated by Sonovue, a 2.1-fold increase (95% CI 1.3- to 3.4-fold) was measured, whereas SonoTran Particles gave a 3.6-fold increase (95% CI 2.3- to 5.8-fold). Conclusions: Ultrasound-mediated cavitation, especially when nucleated by nanoscale gas-entrapping particles, can noninvasively increase site-specific delivery of therapeutic antibodies to solid tumors.
Collapse
Affiliation(s)
- Megan Grundy
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Luca Bau
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Claudia Hill
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Catherine Paverd
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Christophoros Mannaris
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - James Kwan
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK
| | - Calum Crake
- OxSonics Therapeutics, Oxford Science Park, Oxford OX4 4GA, UK
| | | | - Constantin Coussios
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Robert Carlisle
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
14
|
Tselikas L, Champiat S, Sheth RA, Yevich S, Ammari S, Deschamps F, Farhane S, Roux C, Susini S, Mouraud S, Delpla A, Raoult T, Robert C, Massard C, Barlesi F, Soria JC, Marabelle A, de Baere T. Interventional Radiology for Local Immunotherapy in Oncology. Clin Cancer Res 2021; 27:2698-2705. [PMID: 33419781 DOI: 10.1158/1078-0432.ccr-19-4073] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/24/2020] [Accepted: 01/04/2021] [Indexed: 11/16/2022]
Abstract
Human intratumoral immunotherapy (HIT-IT) is under rapid development, with promising preliminary results and high expectations for current phase III trials. While outcomes remain paramount for patients and the referring oncologists, the technical aspects of drug injection are critical to the interventional radiologist to ensure optimal and reproducible outcomes. The technical considerations for HIT-IT affect the safety, efficacy, and further development of this treatment option. Image-guided access to the tumor allows the therapeutic index of a treatment to be enhanced by increasing the intratumoral drug concentration while minimizing its systemic exposure and associated on-target off-tumor adverse events. Direct access to the tumor also enables the acquisition of cancer tissue for sequential sampling to better understand the pharmacodynamics of the injected immunotherapy and its efficacy through correlation of immune responses, pathologic responses, and imaging tumor response. The aim of this article is to share the technical insights of HIT-IT, with particular consideration for patient selection, lesion assessment, image guidance, and technical injection options. In addition, the organization of a standard patient workflow is discussed, so as to optimize HIT-IT outcome and the patient experience.
Collapse
Affiliation(s)
- Lambros Tselikas
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France. .,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Stephane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Rahul A Sheth
- Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steve Yevich
- Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samy Ammari
- Radiology Department, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Frederic Deschamps
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France
| | - Siham Farhane
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Charles Roux
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France
| | - Sandrine Susini
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Severine Mouraud
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Alexandre Delpla
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France
| | - Thibault Raoult
- Service de Promotion des Etudes Cliniques (SPEC), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Caroline Robert
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.,Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Christophe Massard
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Fabrice Barlesi
- Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Jean-Charles Soria
- Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Aurélien Marabelle
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France.,Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thierry de Baere
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France.,Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| |
Collapse
|
15
|
Schumacher O, Galvão DA, Taaffe DR, Chee R, Spry N, Newton RU. Exercise modulation of tumour perfusion and hypoxia to improve radiotherapy response in prostate cancer. Prostate Cancer Prostatic Dis 2020; 24:1-14. [PMID: 32632128 PMCID: PMC8012204 DOI: 10.1038/s41391-020-0245-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/25/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022]
Abstract
Background An increasing number of studies indicate that exercise plays an important role in the overall care of prostate cancer (PCa) patients before, during and after treatment. Historically, research has focused on exercise as a modulator of physical function, psychosocial well-being as well as a countermeasure to cancer- and treatment-related adverse effects. However, recent studies reveal that exercise may also directly influence tumour physiology that could beneficially affect the response to radiotherapy. Methods In this narrative review, we provide an overview of tumour vascular characteristics that limit the effect of radiation and establish a rationale for exercise as adjunct therapy during PCa radiotherapy. Further, we summarise the existing literature on exercise as a modulator of tumour perfusion and hypoxia and outline potential future research directions. Results Preclinical research has shown that exercise can reduce intratumoral hypoxia—a major limiting factor in radiotherapy—by improving tumour perfusion and vascularisation. In addition, preliminary evidence suggests that exercise training can improve radiotherapy treatment outcomes by increasing natural killer cell infiltration in a murine PCa model. Conclusions Exercise is a potentially promising adjunct therapy for men with PCa undergoing radiotherapy that may increase its effectiveness. However, exercise-induced tumour radiosensitisation remains to be confirmed in preclinical and clinical trials, as does the optimal exercise prescription to elicit such effects.
Collapse
Affiliation(s)
- Oliver Schumacher
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia. .,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.
| | - Daniel A Galvão
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Dennis R Taaffe
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Raphael Chee
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,GenesisCare, Joondalup, WA, Australia
| | - Nigel Spry
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,GenesisCare, Joondalup, WA, Australia
| | - Robert U Newton
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
16
|
Affiliation(s)
- Claudia Hill
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Robert Carlisle
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| |
Collapse
|
17
|
Marabelle A, Andtbacka R, Harrington K, Melero I, Leidner R, de Baere T, Robert C, Ascierto PA, Baurain JF, Imperiale M, Rahimian S, Tersago D, Klumper E, Hendriks M, Kumar R, Stern M, Öhrling K, Massacesi C, Tchakov I, Tse A, Douillard JY, Tabernero J, Haanen J, Brody J. Starting the fight in the tumor: expert recommendations for the development of human intratumoral immunotherapy (HIT-IT). Ann Oncol 2018; 29:2163-2174. [PMID: 30295695 PMCID: PMC6290929 DOI: 10.1093/annonc/mdy423] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
A European Society for Medical Oncology (ESMO)-sponsored expert meeting was held in Paris on 8 March 2018 which comprised 11 experts from academia, 11 experts from the pharmaceutical industry and 2 clinicians who were representatives of ESMO. The focus of the meeting was exclusively on the intratumoral injection/delivery of immunostimulatory agents with the aim of harmonizing the standard terms and methodologies used in the reporting of human intratumoral immunotherapy (HIT-IT) clinical trials to ensure quality assurance and avoid a blurring of the data reported from different studies. The goal was to provide a reference document, endorsed by the panel members that could provide guidance to clinical investigators, pharmaceutical companies, ethics committees, independent review boards, patient advocates and the regulatory authorities and promote an increase in the number and quality of HIT-IT clinical trials in the future. Particular emphasis was placed not only on the development of precise definitions to facilitate a better understanding between investigators but also on the importance of systematic serial biopsies as a driver for translational research and the need for the recording and reporting of data, to facilitate a better understanding of the key processes involved.
Collapse
Affiliation(s)
- A Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| | - R Andtbacka
- Surgical Oncology Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, USA
| | - K Harrington
- The Royal Marsden/The Institute of Cancer Research, National Institute for Health Research Biomedical Centre, London, UK
| | - I Melero
- Clinica Universidad de Navarra and CIBERONC, Pamplona, Spain
| | - R Leidner
- Providence Cancer Center, Earle A. Chiles Research Institute, Portland, USA
| | - T de Baere
- Department of Image Guided Therapy, Gustave Roussy, Université Paris-Saclay, Villejuif
| | - C Robert
- Department of Dermatology, Institute Gustave-Roussy, Paris, France
| | - P A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - J-F Baurain
- King Albert II Cancer Institute, Cliniques Universitaires St Luc, Université Catholique de Louvain, Brussels, Belgium
| | | | | | - D Tersago
- Clinical Development, Bioncotech Therapeutics, Madrid, Spain
| | | | - M Hendriks
- Aduro Biotech, Eindhoven, The Netherlands
| | - R Kumar
- MedImmune, LLC, Gaithersburg, USA
| | | | | | - C Massacesi
- Global Product Development Oncology, Pfizer, USA
| | | | - A Tse
- Oncology Early Development, Merck & Co., Inc, Kenilworth, USA
| | | | - J Tabernero
- Medical Oncology Department, Vall d' Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - J Haanen
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J Brody
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai Hospital, New York, USA
| |
Collapse
|
18
|
For the Success of Oncolytic Viruses: Single Cycle Cures or Repeat Treatments? (One Cycle Should Be Enough). Mol Ther 2018; 26:1876-1880. [PMID: 30029891 DOI: 10.1016/j.ymthe.2018.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
19
|
Felt SA, Grdzelishvili VZ. Recent advances in vesicular stomatitis virus-based oncolytic virotherapy: a 5-year update. J Gen Virol 2017; 98:2895-2911. [PMID: 29143726 DOI: 10.1099/jgv.0.000980] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oncolytic virus (OV) therapy is an anti-cancer approach that uses viruses that preferentially infect, replicate in and kill cancer cells. Vesicular stomatitis virus (VSV, a rhabdovirus) is an OV that is currently being tested in the USA in several phase I clinical trials against different malignancies. Several factors make VSV a promising OV: lack of pre-existing human immunity against VSV, a small and easy to manipulate genome, cytoplasmic replication without risk of host cell transformation, independence of cell cycle and rapid growth to high titres in a broad range of cell lines facilitating large-scale virus production. While significant advances have been made in VSV-based OV therapy, room for improvement remains. Here we review recent studies (published in the last 5 years) that address 'old' and 'new' challenges of VSV-based OV therapy. These studies focused on improving VSV safety, oncoselectivity and oncotoxicity; breaking resistance of some cancers to VSV; preventing premature clearance of VSV; and stimulating tumour-specific immunity. Many of these approaches were based on combining VSV with other therapeutics. This review also discusses another rhabdovirus closely related to VSV, Maraba virus, which is currently being tested in Canada in phase I/II clinical trials.
Collapse
Affiliation(s)
- Sébastien A Felt
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Valery Z Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| |
Collapse
|
20
|
Yokoda R, Nagalo BM, Vernon B, Oklu R, Albadawi H, DeLeon TT, Zhou Y, Egan JB, Duda DG, Borad MJ. Oncolytic virus delivery: from nano-pharmacodynamics to enhanced oncolytic effect. Oncolytic Virother 2017; 6:39-49. [PMID: 29184854 PMCID: PMC5687448 DOI: 10.2147/ov.s145262] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
With the advancement of a growing number of oncolytic viruses (OVs) to clinical development, drug delivery is becoming an important barrier to overcome for optimal therapeutic benefits. Host immunity, tumor microenvironment and abnormal vascularity contribute to inefficient vector delivery. A number of novel approaches for enhanced OV delivery are under evaluation, including use of nanoparticles, immunomodulatory agents and complex viral–particle ligands along with manipulations of the tumor microenvironment. This field of OV delivery has quickly evolved to bioengineering of complex nanoparticles that could be deposited within the tumor using minimal invasive image-guided delivery. Some of the strategies include ultrasound (US)-mediated cavitation-enhanced extravasation, magnetic viral complexes delivery, image-guided infusions with focused US and targeting photodynamic virotherapy. In addition, strategies that modulate tumor microenvironment to decrease extracellular matrix deposition and increase viral propagation are being used to improve tumor penetration by OVs. Some involve modification of the viral genome to enhance their tumoral penetration potential. Here, we highlight the barriers to oncolytic viral delivery, and discuss the challenges to improving it and the perspectives of establishing new modes of active delivery to achieve enhanced oncolytic effects.
Collapse
Affiliation(s)
- Raquel Yokoda
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Bolni M Nagalo
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Brent Vernon
- Department of Biomedical Engineering, Arizona State University, Tempe
| | - Rahmi Oklu
- Division of Vascular and Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, AZ
| | - Hassan Albadawi
- Division of Vascular and Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, AZ
| | - Thomas T DeLeon
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Yumei Zhou
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Jan B Egan
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Dan G Duda
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Mitesh J Borad
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| |
Collapse
|
21
|
Zhang L, Steele MB, Jenks N, Grell J, Suksanpaisan L, Naik S, Federspiel MJ, Lacy MQ, Russell SJ, Peng KW. Safety Studies in Tumor and Non-Tumor-Bearing Mice in Support of Clinical Trials Using Oncolytic VSV-IFNβ-NIS. HUM GENE THER CL DEV 2017; 27:111-22. [PMID: 27532609 DOI: 10.1089/humc.2016.061] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Oncolytic VSV-IFNβ-NIS is selectively destructive to tumors. Here, we present the IND enabling preclinical rodent studies in support of clinical testing of vesicular stomatitis virus (VSV) as a systemic therapy. Efficacy studies showed dose-dependent tumor regression in C57BL/KaLwRij mice bearing syngeneic 5TGM1 plasmacytomas after systemic VSV administration. In contrast, the virus was effective at all doses tested against human KAS6/1 xenografts in SCID mice. Intravenous administration of VSV-mIFNβ-NIS is well tolerated in C57BL/6 mice up to 5 × 10(10) TCID50 (50% tissue culture infective dose)/kg with no neurovirulence, no cytokine storm, and no abnormalities in tissues. Dose-limiting toxicities included elevated transaminases, thrombocytopenia, and lymphopenia. Inactivated viral particles did not cause hepatic toxicity. Intravenously administered VSV was preferentially sequestered by macrophages in the spleen and liver. Quantitative RT-PCR analysis for total viral RNA on days 2, 7, 21, and 58 showed highest VSV RNA in day 2 samples; highest in spleen, liver, lung, lymph node, kidney, gonad, and bone marrow. No infectious virus was recovered from tissues at any time point. The no observable adverse event level and maximum tolerated dose of VSV-mIFNβ-NIS in C57BL/6 mice are 10(10) TCID50/kg and 5 × 10(10) TCID50/kg, respectively. Clinical translation of VSV-IFNβ-NIS is underway in companion dogs with cancer and in human patients with relapsed hematological malignancies and endometrial cancer.
Collapse
Affiliation(s)
- Lianwen Zhang
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota
| | - Michael B Steele
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,2 Toxicology and Pharmacology Laboratory, Mayo Clinic , Rochester, Minnesota
| | - Nathan Jenks
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,2 Toxicology and Pharmacology Laboratory, Mayo Clinic , Rochester, Minnesota
| | - Jacquelyn Grell
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,2 Toxicology and Pharmacology Laboratory, Mayo Clinic , Rochester, Minnesota
| | | | - Shruthi Naik
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota
| | - Mark J Federspiel
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,4 Viral Vector Production Laboratory, Mayo Clinic , Rochester, Minnesota
| | - Martha Q Lacy
- 5 Division of Hematology, Mayo Clinic , Rochester, Minnesota
| | - Stephen J Russell
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,5 Division of Hematology, Mayo Clinic , Rochester, Minnesota
| | - Kah-Whye Peng
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,2 Toxicology and Pharmacology Laboratory, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
22
|
Guo ZS, Liu Z, Kowalsky S, Feist M, Kalinski P, Lu B, Storkus WJ, Bartlett DL. Oncolytic Immunotherapy: Conceptual Evolution, Current Strategies, and Future Perspectives. Front Immunol 2017; 8:555. [PMID: 28555136 PMCID: PMC5430078 DOI: 10.3389/fimmu.2017.00555] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/25/2017] [Indexed: 12/17/2022] Open
Abstract
The concept of oncolytic virus (OV)-mediated cancer therapy has been shifted from an operational virotherapy paradigm to an immunotherapy. OVs often induce immunogenic cell death (ICD) of cancer cells, and they may interact directly with immune cells as well to prime antitumor immunity. We and others have developed a number of strategies to further stimulate antitumor immunity and to productively modulate the tumor microenvironment (TME) for potent and sustained antitumor immune cell activity. First, OVs have been engineered or combined with other ICD inducers to promote more effective T cell cross-priming, and in many cases, the breaking of functional immune tolerance. Second, OVs may be armed to express Th1-stimulatory cytokines/chemokines or costimulators to recruit and sustain the potent antitumor immunity into the TME to focus their therapeutic activity within the sites of disease. Third, combinations of OV with immunomodulatory drugs or antibodies that recondition the TME have proven to be highly promising in early studies. Fourth, combinations of OVs with other immunotherapeutic regimens (such as prime-boost cancer vaccines, CAR T cells; armed with bispecific T-cell engagers) have also yielded promising preliminary findings. Finally, OVs have been combined with immune checkpoint blockade, with robust antitumor efficacy being observed in pilot evaluations. Despite some expected hurdles for the rapid translation of OV-based state-of-the-art protocols, we believe that a cohort of these novel approaches will join the repertoire of standard cancer treatment options in the near future.
Collapse
Affiliation(s)
- Zong Sheng Guo
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zuqiang Liu
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stacy Kowalsky
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mathilde Feist
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Surgery, CCM/CVK, Charité – Universitaetsmedizin Berlin, Berlin, Germany
| | - Pawel Kalinski
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Binfeng Lu
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Walter J. Storkus
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L. Bartlett
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
23
|
Chia SL, Lei J, Ferguson DJP, Dyer A, Fisher KD, Seymour LW. Group B adenovirus enadenotucirev infects polarised colorectal cancer cells efficiently from the basolateral surface expected to be encountered during intravenous delivery to treat disseminated cancer. Virology 2017; 505:162-171. [PMID: 28260622 DOI: 10.1016/j.virol.2017.02.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/03/2017] [Accepted: 02/14/2017] [Indexed: 12/28/2022]
Abstract
Enadenotucirev (EnAd) is a group B oncolytic adenovirus developed for systemic delivery and currently undergoing clinical evaluation for advanced cancer therapy. For differentiated carcinomas, systemic delivery would likely expose virus particles to the basolateral surface of cancer cells rather than the apical surface encountered during natural infection. Here, we compare the ability of EnAd and adenovirus type-5 (Ad5) to infect polarised colorectal carcinoma cells from the apical or basolateral surfaces. Whereas Ad5 infection was more efficient via the apical than basolateral surface, EnAd readily infected cells from either surface. Progeny particles from EnAd were released preferentially via the apical surface for all cell lines and routes of infection. These data further support the utility of group B adenoviruses for systemic delivery and suggest that progeny virus are more likely to be released into the tumour rather than back through the basolateral surface into the blood stream.
Collapse
Affiliation(s)
- Suet-Lin Chia
- Department of Oncology, University of Oxford, Oxford, United Kingdom; Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Janet Lei
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - David J P Ferguson
- Nuffield Department of Clinical Laboratory Science, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Arthur Dyer
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Kerry D Fisher
- Department of Oncology, University of Oxford, Oxford, United Kingdom; PsiOxus Therapeutics, Abingdon, United Kingdom
| | - Leonard W Seymour
- Department of Oncology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
24
|
Maroun J, Muñoz-Alía M, Ammayappan A, Schulze A, Peng KW, Russell S. Designing and building oncolytic viruses. Future Virol 2017; 12:193-213. [PMID: 29387140 PMCID: PMC5779534 DOI: 10.2217/fvl-2016-0129] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/30/2017] [Indexed: 02/07/2023]
Abstract
Oncolytic viruses (OVs) are engineered and/or evolved to propagate selectively in cancerous tissues. They have a dual mechanism of action; direct killing of infected cancer cells cross-primes anticancer immunity to boost the killing of uninfected cancer cells. The goal of the field is to develop OVs that are easily manufactured, efficiently delivered to disseminated sites of cancer growth, undergo rapid intratumoral spread, selectively kill tumor cells, cause no collateral damage and pose no risk of transmission in the population. Here we discuss the many virus engineering strategies that are being pursued to optimize delivery, intratumoral spread and safety of OVs derived from different virus families. With continued progress, OVs have the potential to transform the paradigm of cancer care.
Collapse
Affiliation(s)
- Justin Maroun
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Miguel Muñoz-Alía
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Arun Ammayappan
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Autumn Schulze
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Stephen Russell
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
25
|
Seymour LW, Fisher KD. Under Pressure: Elevated Blood Pressure Enhances Targeting of Tumors by Oncolytic Viruses. Mol Ther 2016; 24:204-205. [PMID: 26906615 PMCID: PMC4817829 DOI: 10.1038/mt.2016.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
| | - Kerry D Fisher
- Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Peters C, Nigim F, Chiocca EA, Rabkin SD. Oncolytic viruses on the cusp of success?: proceedings of the 9th International Conference on Oncolytic Virus Therapeutics. MOLECULAR THERAPY-ONCOLYTICS 2016. [PMCID: PMC4898404 DOI: 10.1038/mto.2016.16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Boston, Massachusetts, was the site of the 9th International Conference on Oncolytic Virus Therapeutics held 13–16 June 2015. An overarching theme of the meeting was the continued development of combinatorial treatment regimens to bolster the therapeutic potential of oncolytic viruses (OVs). Several talks focused on combining OVs with immune checkpoint inhibitors in a wide array of tumors, signaling an experimental and thematic shift toward driving immune activation to clear a tumor versus relying on direct viral oncolysis. An important aspect of the meeting was the variety of ongoing OV clinical trials. Topics ranged from basic virology to clinical trials and from academic research to intellectual property and biotechnology. There was much excitement due to the US Food and Drug Administration’s recent consideration of talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma (T-VEC was approved in October, following the conference). Here, we summarize the meeting’s primary themes, which reflect the current state of the field.
Collapse
|