1
|
Parthasarathy A, Frost M, Gillard AG, Alonso MM, Gomez-Manzano C, Fueyo J. The multilayered control of acetylation during adenovirus-based immunotherapy of cancer. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200976. [PMID: 40236995 PMCID: PMC11999684 DOI: 10.1016/j.omton.2025.200976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Adenoviruses are highly immunogenic agents that have shown promise first as gene delivery vectors and later as oncolytic viruses. Currently, oncolytic adenoviruses are featured in over 30% of cancer virotherapy clinical trials. Due to their effective cellular uptake and hijack of cellular machinery, replication-competent adenoviruses are promising therapeutic agents for treating a wide range of tumors. Adenoviral influence on host cell acetylome regulation has regained attention, as these viruses redirect or suppress acetylation during replication, making them potentially desirable therapeutic agents for cancers driven by epigenetic modifications. In this review, we aim to cover the viral processes influencing the acetylome of the host genome. In addition, we shall discuss the effect of differential acetylation on the antiviral defense mounted by the host immune system. Lastly, we will discuss the opportunities for combining acetylation modifiers with oncolytic adenoviruses to improve further outcomes for patients treated with viroimmunotherapy.
Collapse
Affiliation(s)
- Akhila Parthasarathy
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria Frost
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew G. Gillard
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marta M. Alonso
- Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
2
|
Kulbay M, Tuli N, Mazza M, Jaffer A, Juntipwong S, Marcotte E, Tanya SM, Nguyen AXL, Burnier MN, Demirci H. Oncolytic Viruses and Immunotherapy for the Treatment of Uveal Melanoma and Retinoblastoma: The Current Landscape and Novel Advances. Biomedicines 2025; 13:108. [PMID: 39857692 PMCID: PMC11762644 DOI: 10.3390/biomedicines13010108] [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: 12/09/2024] [Revised: 12/25/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Intraocular malignant tumors are rare; however, they can cause serious life-threatening complications. Uveal melanoma (UM) and retinoblastoma (RB) are the most common intraocular tumors in adults and children, respectively, and come with a great disease burden. For many years, several different treatment modalities for UM and RB have been proposed, with chemotherapy for RB cases and plaque radiation therapy for localized UM as first-line treatment options. Extraocular extension, recurrence, and metastasis constitute the major challenges of conventional treatments. To overcome these obstacles, immunotherapy, which encompasses different treatment options such as oncolytic viruses, antibody-mediated immune modulations, and targeted immunotherapy, has shown great potential as a novel therapeutic tool for cancer therapy. These anti-cancer treatment options provide numerous advantages such as selective cancer cell death and the promotion of an anti-tumor immune response, and they prove useful in preventing vision impairment due to macular and/or optic disc involvement. Numerous factors such as the vector choice, route of administration, dosing, and patient characteristics must be considered when engineering an oncolytic virus or other forms of immunotherapy vectors. This manuscript provides an in-depth review of the molecular design of oncolytic viruses (e.g., virus capsid proteins and encapsulation technologies, vectors for delivery, cell targeting) and immunotherapy. The most recent advances in preclinical- and clinical-phase studies are further summarized. The recent developments in virus-like drug conjugates (i.e., AU011), oncolytic viruses for metastatic UM, and targeted immunotherapies have shown great results in clinical trials for the future clinical application of these novel technologies in the treatment algorithm of certain intraocular tumors.
Collapse
Affiliation(s)
- Merve Kulbay
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3J1, Canada; (M.K.)
| | - Nicolas Tuli
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | - Massimo Mazza
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | - Armaan Jaffer
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 2V5, Canada
- Research Excellence Cluster in Vision, University of British Columbia, Vancouver, BC V5Z 3N9, Canada
| | - Sarinee Juntipwong
- Kellogg Eye Center, Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, MI 48105, USA
| | - Emily Marcotte
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Stuti Misty Tanya
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3J1, Canada; (M.K.)
| | - Anne Xuan-Lan Nguyen
- Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Miguel N. Burnier
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3J1, Canada; (M.K.)
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Hakan Demirci
- Kellogg Eye Center, Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
3
|
Costa-Garcia M, Moya-Borrego L, Alemany Bonastre R, Moreno Olié R. Optimized protocol for culturing menstrual blood-derived MSCs for combination with oncolytic adenoviruses in cancer treatment. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200907. [PMID: 39758253 PMCID: PMC11697545 DOI: 10.1016/j.omton.2024.200907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 01/07/2025]
Abstract
Oncolytic viruses (OVs) are a promising therapeutic approach for cancer, although their systemic administration faces significant challenges. Mesenchymal stem cells have emerged as potential carriers to overcome these obstacles due to their tumor-tropic properties. This study investigates the use of menstrual blood-derived mesenchymal stem cells (MenSCs) as carriers for OVs in cancer therapy, focusing on enhancing their efficacy through different culture conditions. MenSCs were isolated from donors of different ages and cultured under normoxic and hypoxic conditions, with varying adherence capacities. Hypoxic conditions significantly improved MenSCs proliferation and tumor migration capabilities, as demonstrated by proliferation assays and RNA-sequencing analysis, which revealed upregulation of genes related to cell division and tumor tropism. In vivo studies using a lung adenocarcinoma mouse model confirmed that hypoxia-conditioned MenSCs had superior tumor-homing abilities. The study also demonstrated the feasibility of establishing a master and working cell bank from a single menstrual blood donation. These findings suggest that hypoxia-conditioned MenSCs could be highly effective as OV carriers, potentially leading to better clinical outcomes in cancer treatment by enhancing tumor targeting and therapeutic efficacy.
Collapse
Affiliation(s)
- Marcel Costa-Garcia
- Cancer Immunotherapy Group, Oncobell and iProCURE programs, IDIBELL-Institut Català d'Oncologia, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Laura Moya-Borrego
- Cancer Immunotherapy Group, Oncobell and iProCURE programs, IDIBELL-Institut Català d'Oncologia, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ramon Alemany Bonastre
- Cancer Immunotherapy Group, Oncobell and iProCURE programs, IDIBELL-Institut Català d'Oncologia, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael Moreno Olié
- Cancer Immunotherapy Group, Oncobell and iProCURE programs, IDIBELL-Institut Català d'Oncologia, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
4
|
Morales-Molina A, Rodriguez-Milla MA, Garcia-Rodriguez P, Hidalgo L, Alemany R, Garcia-Castro J. Deletion of the RGD motif from the penton base in oncolytic adenoviruses enhances antitumor efficacy of combined CAR T cell therapy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200863. [PMID: 39290319 PMCID: PMC11406095 DOI: 10.1016/j.omton.2024.200863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024]
Abstract
Oncolytic viruses often face challenges in achieving optimal antitumor immunity as standalone therapies. The penton base RGD-integrin interactions play a significant role in wild-type adenovirus-induced innate immune responses. To modify these responses, we present ISC301, a novel oncolytic adenovirus engineered by deleting the natural RGD motifs in the penton base while incorporating artificial RGD motifs in the fiber knobs. ISC301 demonstrated comparable in vitro infectivity, cytotoxic effects, and signaling profiles across various cell types to its parental ICOVIR-5, which retains the penton base RGD motif. In immunodeficient and immunocompetent mouse models, ISC301 exhibited similar in vivo antitumor efficacy to ICOVIR-5. However, ISC301 induced higher intratumoral inflammation through NF-κB activation, leading to increased levels of tumor-infiltrating leukocytes and higher proportion of cytotoxic CD8+ T cells. In addition, ISC301 elicits a heightened pro-inflammatory response in peripheral blood. Importantly, when combined with CAR T cell therapy, ISC301 exhibited superior antitumor efficacy, surpassing monotherapy outcomes. These findings emphasize the impact of adenoviral modifications on antitumor immune responses. The deletion of penton base RGD motifs enhances ISC301's pro-inflammatory profile and boosts CAR T cell therapy efficacy. This study enhances understanding of oncolytic virus engineering strategies, positioning ISC301 as a promising candidate for combined immunotherapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
| | | | - Patricia Garcia-Rodriguez
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain
- Universidad Nacional de Educación a Distancia, UNED, 28015 Madrid, Spain
| | - Laura Hidalgo
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Ramon Alemany
- Oncobell and ProCure Programs, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Javier Garcia-Castro
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain
- Instituto de Investigación de Enfermedades Raras (IIER) & Departamento de Desarrollo de Medicamentos de Terapias Avanzadas (DDMTA), Instituto de Salud Carlos III, 28220 Madrid, Spain
| |
Collapse
|
5
|
Kato Y, Rice N, Pokrass M, Jeong J, Rodriguez R, Field JJ, Nowyhed H. Nonclinical characterization of ICVB-1042 as a selective oncolytic adenovirus for solid tumor treatment. Commun Biol 2024; 7:1132. [PMID: 39271928 PMCID: PMC11399272 DOI: 10.1038/s42003-024-06839-6] [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: 01/08/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
ICVB-1042 is an oncolytic adenovirus containing modifications to enhance replication, lysis, and viral spreading in tumor cells. The anti-tumor activity, immune activation, tropism, selectivity, and mechanism of action were evaluated in preparation for a first-in-human study. ICVB-1042 was at least 100-fold more cytotoxic in A549 cells than in normal primary cells tested, demonstrating its high tumor selectivity and a low likelihood of targeting primary tissues. ICVB-1042 administered to mice intravenously or intratumorally was effective in reducing tumor burden. Its intravenous administration also inhibited tumor growth in orthotopic models. ICVB-1042 was well tolerated in mice compared to HAdV-C5 (Wt Ad5), with reduced liver sequestration, supporting safety of the drug for systemic delivery. These preclinical data demonstrating the safety and potency of ICVB-1042 for treatment of various solid tumors support the ongoing clinical investigation (NCT05904236).
Collapse
Affiliation(s)
- Yu Kato
- IconOVir Bio, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
6
|
Polychronopoulos PA, Bedoya-Reina OC, Johnsen JI. The Neuroblastoma Microenvironment, Heterogeneity and Immunotherapeutic Approaches. Cancers (Basel) 2024; 16:1863. [PMID: 38791942 PMCID: PMC11119056 DOI: 10.3390/cancers16101863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Neuroblastoma is a peripheral nervous system tumor that almost exclusively occurs in young children. Although intensified treatment modalities have led to increased patient survival, the prognosis for patients with high-risk disease is still around 50%, signifying neuroblastoma as a leading cause of cancer-related deaths in children. Neuroblastoma is an embryonal tumor and is shaped by its origin from cells within the neural crest. Hence, neuroblastoma usually presents with a low mutational burden and is, in the majority of cases, driven by epigenetically deregulated transcription networks. The recent development of Omic techniques has given us detailed knowledge of neuroblastoma evolution, heterogeneity, and plasticity, as well as intra- and intercellular molecular communication networks within the neuroblastoma microenvironment. Here, we discuss the potential of these recent discoveries with emphasis on new treatment modalities, including immunotherapies which hold promise for better future treatment regimens.
Collapse
Affiliation(s)
- Panagiotis Alkinoos Polychronopoulos
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 11883 Stockholm, Sweden; (P.A.P.); (O.C.B.-R.)
| | - Oscar C. Bedoya-Reina
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 11883 Stockholm, Sweden; (P.A.P.); (O.C.B.-R.)
- School of Medical Sciences, Örebro University, 70182 Örebro, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 11883 Stockholm, Sweden; (P.A.P.); (O.C.B.-R.)
| |
Collapse
|
7
|
Hu M, Liao X, Tao Y, Chen Y. Advances in oncolytic herpes simplex virus and adenovirus therapy for recurrent glioma. Front Immunol 2023; 14:1285113. [PMID: 38022620 PMCID: PMC10652401 DOI: 10.3389/fimmu.2023.1285113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Recurrent glioma treatment is challenging due to molecular heterogeneity and treatment resistance commonly observed in these tumors. Researchers are actively pursuing new therapeutic strategies. Oncolytic viruses have emerged as a promising option. Oncolytic viruses selectively replicate within tumor cells, destroying them and stimulating the immune system for an enhanced anticancer response. Among Oncolytic viruses investigated for recurrent gliomas, oncolytic herpes simplex virus and oncolytic adenovirus show notable potential. Genetic modifications play a crucial role in optimizing their therapeutic efficacy. Different generations of replicative conditioned oncolytic human adenovirus and oncolytic HSV have been developed, incorporating specific modifications to enhance tumor selectivity, replication efficiency, and immune activation. This review article summarizes these genetic modifications, offering insights into the underlying mechanisms of Oncolytic viruses' therapy. It also aims to identify strategies for further enhancing the therapeutic benefits of Oncolytic viruses. However, it is important to acknowledge that additional research and clinical trials are necessary to establish the safety, efficacy, and optimal utilization of Oncolytic viruses in treating recurrent glioblastoma.
Collapse
Affiliation(s)
- Mingming Hu
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - XuLiang Liao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Tao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yaohui Chen
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Analysis of the Prevalence of Binding and Neutralizing Antibodies against 39 Human Adenovirus Types in Student Cohorts Reveals Low-Prevalence Types and a Decline in Binding Antibody Levels during the SARS-CoV-2 Pandemic. J Virol 2022; 96:e0113322. [DOI: 10.1128/jvi.01133-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Vectors based on human adenoviruses (HAdVs) are important for the development of novel immunizations, oncolytic therapies, and gene therapies. The use of HAdV-based vaccines against Ebola virus, the rapid adaptation of the vector technology for vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and their very good efficacy have shown the great potential of HAdV-based vaccines.
Collapse
|
9
|
Recent Advances and Challenges in Uveal Melanoma Immunotherapy. Cancers (Basel) 2022; 14:cancers14133094. [PMID: 35804863 PMCID: PMC9264803 DOI: 10.3390/cancers14133094] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Uveal melanoma is the most common primary intraocular malignancy in adults. Although it can be controlled locally, half of the patients still develop metastases. To date, there have been no standard therapeutic strategies for the prevention or treatment of metastases. Existing therapies, such as chemotherapy and targeted therapies, induce only minimal responses. This review focuses on newly published research on immunotherapy. We highlight expanding treatments and their clinical outcomes, as well as propose promising new treatments and feasible checkpoints. Based on these findings, we provide innovative insights into feasible strategies for the treatment of patients with uveal melanoma. Abstract Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. Compared to cutaneous melanoma (CM), which mainly harbors BRAF or NRAS mutations, UM predominantly harbors GNAQ or GNA11 mutations. Although primary UM can be controlled locally, approximately 50% of patients still develop metastases. To date, there have been no standard therapeutic strategies for the prevention or treatment of metastases. Unfortunately, chemotherapy and targeted therapies only induce minimal responses in patients with metastatic UM, with a median survival time of only 4–5 months after metastasis detection. Immunotherapy agents, such as immune checkpoint inhibitors, have achieved pioneering outcomes in CM but have shown limited effects in UM. Researchers have explored several feasible checkpoints to identify options for future therapies. Cancer vaccines have shown little in the way of therapeutic benefit in patients with UM, and there are few ongoing trials providing favorable evidence, but adoptive cell transfer-related therapies seem promising and deserve further investigation. More recently, the immune-mobilizing monoclonal T-cell receptor against the cancer molecule tebentafusp showed impressive antitumor effects. Meanwhile, oncolytic viruses and small molecule inhibitors have also gained ground. This review highlights recent progress in burgeoning treatments and provides innovative insights on feasible strategies for the treatment of UM.
Collapse
|
10
|
Naumenko VA, Stepanenko AA, Lipatova AV, Vishnevskiy DA, Chekhonin VP. Infection of non-cancer cells: A barrier or support for oncolytic virotherapy? MOLECULAR THERAPY - ONCOLYTICS 2022; 24:663-682. [PMID: 35284629 PMCID: PMC8898763 DOI: 10.1016/j.omto.2022.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oncolytic viruses are designed to specifically target cancer cells, sparing normal cells. Although numerous studies demonstrate the ability of oncolytic viruses to infect a wide range of non-tumor cells, the significance of this phenomenon for cancer virotherapy is poorly understood. To fill the gap, we summarize the data on infection of non-cancer targets by oncolytic viruses with a special focus on tumor microenvironment and secondary lymphoid tissues. The review aims to address two major questions: how do attenuated viruses manage to infect normal cells, and whether it is of importance for oncolytic virotherapy.
Collapse
Affiliation(s)
- Victor A. Naumenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Corresponding author Victor A. Naumenko, PhD, V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia.
| | - Aleksei A. Stepanenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Anastasiia V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Daniil A. Vishnevskiy
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
| | - Vladimir P. Chekhonin
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| |
Collapse
|
11
|
Bazan-Peregrino M, Garcia-Carbonero R, Laquente B, Álvarez R, Mato-Berciano A, Gimenez-Alejandre M, Morgado S, Rodríguez-García A, Maliandi MV, Riesco MC, Moreno R, Ginestà MM, Perez-Carreras M, Gornals JB, Prados S, Perea S, Capella G, Alemany R, Salazar R, Blasi E, Blasco C, Cascallo M, Hidalgo M. VCN-01 disrupts pancreatic cancer stroma and exerts antitumor effects. J Immunother Cancer 2022; 9:jitc-2021-003254. [PMID: 35149591 PMCID: PMC8578996 DOI: 10.1136/jitc-2021-003254] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2021] [Indexed: 12/16/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is characterized by dense desmoplastic stroma that limits the delivery of anticancer agents. VCN-01 is an oncolytic adenovirus designed to replicate in cancer cells with a dysfunctional RB1 pathway and express hyaluronidase. Here, we evaluated the mechanism of action of VCN-01 in preclinical models and in patients with pancreatic cancer. Methods VCN-01 replication and antitumor efficacy were evaluated alone and in combination with standard chemotherapy in immunodeficient and immunocompetent preclinical models using intravenous or intratumoral administration. Hyaluronidase activity was evaluated by histochemical staining and by measuring drug delivery into tumors. In a proof-of-concept clinical trial, VCN-01 was administered intratumorally to patients with PDAC at doses up to 1×1011 viral particles in combination with chemotherapy. Hyaluronidase expression was measured in serum by an ELISA and its activity within tumors by endoscopic ultrasound elastography. Results VCN-01 replicated in PDAC models and exerted antitumor effects which were improved when combined with chemotherapy. Hyaluronidase expression by VCN-01 degraded tumor stroma and facilitated delivery of a variety of therapeutic agents such as chemotherapy and therapeutic antibodies. Clinically, treatment was generally well-tolerated and resulted in disease stabilization of injected lesions. VCN-01 was detected in blood as secondary peaks and in post-treatment tumor biopsies, indicating virus replication. Patients had increasing levels of hyaluronidase in sera over time and decreased tumor stiffness, suggesting stromal disruption. Conclusions VCN-01 is an oncolytic adenovirus with direct antitumor effects and stromal disruption capabilities, representing a new therapeutic agent for cancers with dense stroma. Trial registration number EudraCT number: 2012-005556-42 and NCT02045589.
Collapse
Affiliation(s)
| | - Rocio Garcia-Carbonero
- Oncology Department, Hospital Universitario 12 de Octubre, Imas12, UCM, CNIO, CIBERONC, Madrid, Spain
| | - Berta Laquente
- Medical Oncology Department, IDIBELL-Institut Catala d' Oncologia, L'Hospitalet de Llobregat, Barcelona, 08908, Spain
| | - Rafael Álvarez
- Centro Integral Oncológico Clara Campal (CIOCC), Oña 10, 28050, Madrid, Spain
| | | | | | - Sara Morgado
- VCN Biosciences, Sant Cugat del Valles, Barcelona, 08174, Spain
| | - Alba Rodríguez-García
- Virotherapy and Gene Therapy Group, Oncobell and ProCure Programs, IDIBELL-Instituto Catalan d'Oncología, L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Hematology and Oncology, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
| | | | - M Carmen Riesco
- Oncology Department, Hospital Universitario 12 de Octubre, Imas12, UCM, CNIO, CIBERONC, Madrid, Spain
| | - Rafael Moreno
- Virotherapy and Gene Therapy Group, Oncobell and ProCure Programs, IDIBELL-Instituto Catalan d'Oncología, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mireia M Ginestà
- Hereditary Cancer Program, Oncobell Program, CIBERONC, IDIBELL-Instituto Catalan d'Oncología, l'Hospitalet de Llobregat, Barcelona, Spain
| | - Mercedes Perez-Carreras
- Endoscopic Unit, Servicio Aparato Digestivo, University Hospital 12 De Octubre, Madrid, Spain
| | - Joan B Gornals
- Hospital Universitari de Bellvitge, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Susana Prados
- Centro Integral Oncológico Clara Campal (CIOCC), Oña 10, 28050, Madrid, Spain
| | - Sofía Perea
- Centro Integral Oncológico Clara Campal (CIOCC), Oña 10, 28050, Madrid, Spain
| | - Gabriel Capella
- Hereditary Cancer Program, Oncobell Program, CIBERONC, IDIBELL-Instituto Catalan d'Oncología, l'Hospitalet de Llobregat, Barcelona, Spain
| | - Ramon Alemany
- Virotherapy and Gene Therapy Group, Oncobell and ProCure Programs, IDIBELL-Instituto Catalan d'Oncología, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ramon Salazar
- Medical Oncology Department, IDIBELL-Institut Catala d' Oncologia, L'Hospitalet de Llobregat, Barcelona, 08908, Spain
| | - Emma Blasi
- VCN Biosciences, Sant Cugat del Valles, Barcelona, 08174, Spain
| | - Carmen Blasco
- VCN Biosciences, Sant Cugat del Valles, Barcelona, 08174, Spain
| | - Manel Cascallo
- VCN Biosciences, Sant Cugat del Valles, Barcelona, 08174, Spain
| | - Manuel Hidalgo
- Centro Integral Oncológico Clara Campal (CIOCC), Oña 10, 28050, Madrid, Spain .,Div. of Hematology and Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
12
|
Chen XT, Dai SY, Zhan Y, Yang R, Chen DQ, Li Y, Zhou EQ, Dong R. Progress of oncolytic virotherapy for neuroblastoma. Front Pediatr 2022; 10:1055729. [PMID: 36467495 PMCID: PMC9716318 DOI: 10.3389/fped.2022.1055729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022] Open
Abstract
As a neuroendocrine tumor derived from the neural crest, neuroblastoma (NB) is the most common extracranial solid tumor in children. The prognosis in patients with low- and intermediate-risk NB is favorable while that in high-risk patients is often detrimental. However, the management of the considerably large proportion of high-risk patients remains challenging in clinical practice. Among various new approaches, oncolytic virus (OV) therapy offers great advantages in tumor treatment, especially for high-risk NB. Genetic modified OVs can target NB specifically without affecting normal tissue and avoid the widespread drug resistance issue in anticancer monotherapy. Meanwhile, its safety profile provides great potential in combination therapy with chemo-, radio-, and immunotherapy. The therapeutic efficacy of OV for NB is impressive from bench to bedside. The effectiveness and safety of OVs have been demonstrated and reported in studies on children with NB. Furthermore, clinical trials on some OVs (Celyvir, Pexa-Vec (JX-594) and Seneca Valley Virus (NTX-010)) have reported great results. This review summarizes the latest evidence in the therapeutic application of OVs in NB, including those generated in cell lines, animal models and clinical trials.
Collapse
Affiliation(s)
- Xiao-Tong Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Shu-Yang Dai
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Yong Zhan
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Ran Yang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - De-Qian Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Yi Li
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - En-Qing Zhou
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| |
Collapse
|
13
|
Concepts in Oncolytic Adenovirus Therapy. Int J Mol Sci 2021; 22:ijms221910522. [PMID: 34638863 PMCID: PMC8508870 DOI: 10.3390/ijms221910522] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 02/07/2023] Open
Abstract
Oncolytic adenovirus therapy is gaining importance as a novel treatment option for the management of various cancers. Different concepts of modification within the adenovirus vector have been identified that define the mode of action against and the interaction with the tumour. Adenoviral vectors allow for genetic manipulations that restrict tumour specificity and also the expression of specific transgenes in order to support the anti-tumour effect. Additionally, replication of the virus and reinfection of neighbouring tumour cells amplify the therapeutic effect. Another important aspect in oncolytic adenovirus therapy is the virus induced cell death which is a process that activates the immune system against the tumour. This review describes which elements in adenovirus vectors have been identified for modification not only to utilize oncolytic adenovirus vectors into conditionally replicating adenoviruses (CRAds) that allow replication specifically in tumour cells but also to confer specific characteristics to these viruses. These advances in development resulted in clinical trials that are summarized based on the conceptual design.
Collapse
|
14
|
Li M, Zhu Y, Bai B, Fang J, Yao W, Li Y, Li S, Li X, Jin N, Jiang R. Suppression effect of a dual cancer-specific oncolytic adenovirus on luciferase-labeled human melanoma cells in vitro and in vivo. Cancer Biomark 2021; 32:251-262. [PMID: 34459386 DOI: 10.3233/cbm-203150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND To explore the suppressive effect of Apoptin-loaded oncolytic adenovirus (Ad-VT) on luciferase-labeled human melanoma cells in vitro and in vivo. METHODS The stable luciferase-expressing human melanoma cells A375-luc or M14-luc were obtained by transfecting the plasmid pGL4.51 and selection with G418, followed by luciferase activity, genetic stability and bioluminescence intensity assays. In vitro, the inhibitory effects of Ad-VT on A375-luc or M14-luc were evaluated using the MTS cell proliferation, FITC-Annexin V apoptosis detection, transwell migration, Matrigel invasion and scratch assays. The inhibition pathway in Ad-VT-infected A375-luc and M14-luc cells were analyzed by JC-1 staining and Western-blot detection of mitochondrial apoptosis-related proteins. In vivo, the suppressive effects of Ad-VT on A375-luc or M14-luc were assessed by living imaging technology, tumor volume, bioluminescence intensity, survival curves and immunohistochemical analysis of the tumors from the xenograft tumor model BALB/c nude mice. RESULTS The growth and migration of A375-luc and M14-luc were significantly inhibited by Ad-VT in vitro. The evaluations of A375-luc and M14-luc tumor models in BALB/c nude mice were successfully performed using living imaging technology. Ad-VT had an anti-tumor effect by reducing tumor growth and increasing survival in vivo. Ad-VT significantly changed the mitochondrial membrane potential by triggering the the mitochondrial release of apoptosis-related proteins, AIF (apoptosis inducing factor), ARTS (Apoptosis-Related Proteins), and Cyto-c (cytochrome c) from the mitochondria. CONCLUSION Ad-VT reduced the mitochondrial membrane potential in A375-luc or M14-luc cells and induced the mitochondrial release of AIF, ARTS and Cyto-C. Ad-VT induced apoptosis in A375-luc or M14-luc cells via the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Min Li
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yilong Zhu
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Bing Bai
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jinbo Fang
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wei Yao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China.,Center for Disease Control and Prevention, Agency for Offices Administration, Central Military Commission, Beijing, China
| | - Yiquan Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Shanzhi Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiao Li
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.,Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Ningyi Jin
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.,Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Rihua Jiang
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
15
|
Puigdelloses M, Garcia-Moure M, Labiano S, Laspidea V, Gonzalez-Huarriz M, Zalacain M, Marrodan L, Martinez-Velez N, De la Nava D, Ausejo I, Hervás-Stubbs S, Herrador G, Chen Z, Hambardzumyan D, Patino Garcia A, Jiang H, Gomez-Manzano C, Fueyo J, Gállego Pérez-Larraya J, Alonso M. CD137 and PD-L1 targeting with immunovirotherapy induces a potent and durable antitumor immune response in glioblastoma models. J Immunother Cancer 2021; 9:jitc-2021-002644. [PMID: 34281988 PMCID: PMC8291319 DOI: 10.1136/jitc-2021-002644] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2021] [Indexed: 01/09/2023] Open
Abstract
Background Glioblastoma (GBM) is a devastating primary brain tumor with a highly immunosuppressive tumor microenvironment, and treatment with oncolytic viruses (OVs) has emerged as a promising strategy for these tumors. Our group constructed a new OV named Delta-24-ACT, which was based on the Delta-24-RGD platform armed with 4-1BB ligand (4-1BBL). In this study, we evaluated the antitumor effect of Delta-24-ACT alone or in combination with an immune checkpoint inhibitor (ICI) in preclinical models of glioma. Methods The in vitro effect of Delta-24-ACT was characterized through analyses of its infectivity, replication and cytotoxicity by flow cytometry, immunofluorescence (IF) and MTS assays, respectively. The antitumor effect and therapeutic mechanism were evaluated in vivo using several immunocompetent murine glioma models. The tumor microenvironment was studied by flow cytometry, immunohistochemistry and IF. Results Delta-24-ACT was able to infect and exert a cytotoxic effect on murine and human glioma cell lines. Moreover, Delta-24-ACT expressed functional 4-1BBL that was able to costimulate T lymphocytes in vitro and in vivo. Delta-24-ACT elicited a more potent antitumor effect in GBM murine models than Delta-24-RGD, as demonstrated by significant increases in median survival and the percentage of long-term survivors. Furthermore, Delta-24-ACT modulated the tumor microenvironment, which led to lymphocyte infiltration and alteration of their immune phenotype, as characterized by increases in the expression of Programmed Death 1 (PD-1) on T cells and Programmed Death-ligand 1 (PD-L1) on different myeloid cell populations. Because Delta-24-ACT did not induce an immune memory response in long-term survivors, as indicated by rechallenge experiments, we combined Delta-24-ACT with an anti-PD-L1 antibody. In GL261 tumor-bearing mice, this combination showed superior efficacy compared with either monotherapy. Specifically, this combination not only increased the median survival but also generated immune memory, which allowed long-term survival and thus tumor rejection on rechallenge. Conclusions In summary, our data demonstrated the efficacy of Delta-24-ACT combined with a PD-L1 inhibitor in murine glioma models. Moreover, the data underscore the potential to combine local immunovirotherapy with ICIs as an effective therapy for poorly infiltrated tumors.
Collapse
Affiliation(s)
- Montserrat Puigdelloses
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marc Garcia-Moure
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sara Labiano
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Virginia Laspidea
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marisol Gonzalez-Huarriz
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marta Zalacain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Lucia Marrodan
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Naiara Martinez-Velez
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Daniel De la Nava
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Iker Ausejo
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sandra Hervás-Stubbs
- Program in Immunology and Immunotherapy, Foundation for the Applied Medical Research, Pamplona, Spain
| | - Guillermo Herrador
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - ZhiHong Chen
- Department of Oncological Sciences, The Tisch Cancer Institut and Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institut and Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Ana Patino Garcia
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Hong Jiang
- Department of NeuroOncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Candelaria Gomez-Manzano
- Department of NeuroOncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juan Fueyo
- Department of NeuroOncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jaime Gállego Pérez-Larraya
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain .,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marta Alonso
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain .,Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
16
|
Lin W, Zhao Y, Zhong L. Current strategies of virotherapy in clinical trials for cancer treatment. J Med Virol 2021; 93:4668-4692. [PMID: 33738818 DOI: 10.1002/jmv.26947] [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] [Received: 11/28/2020] [Revised: 02/12/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022]
Abstract
As a novel immune-active agent for cancer treatment, viruses have the ability of infecting and replicating in tumor cells. The safety and efficacy of viruses has been tested and confirmed in preclinical and clinical trials. In the last decade, virotherapy has been adopted as a monotherapy or combined therapy with immunotherapy, chemotherapy, or radiotherapy, showing promising outcomes against cancer. In this review, the current strategies of viruses used in clinical trials are classified and described. Besides this, the challenge and future prospects of virotherapy in the management for cancer patients are discussed in this review.
Collapse
Affiliation(s)
- Weijian Lin
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Yongxiang Zhao
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Liping Zhong
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| |
Collapse
|
17
|
Morales-Molina A, Gambera S, Leo A, García-Castro J. Combination immunotherapy using G-CSF and oncolytic virotherapy reduces tumor growth in osteosarcoma. J Immunother Cancer 2021; 9:e001703. [PMID: 33737338 PMCID: PMC7978281 DOI: 10.1136/jitc-2020-001703] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Osteosarcoma is the most common malignant solid tumor that affects bones, however, survival rates of patients with relapsed osteosarcoma have not improved in the last 30 years. Oncolytic virotherapy, which uses viruses designed to selectively replicate in cancer cells, has emerged as a promising treatment for solid tumors. Our group uses mesenchymal stem cells (MSCs) to transport oncolytic adenoviruses (OAds) to the tumor site, a therapeutic strategy called Celyvir. This treatment has been already applied in human patients, canine patients and different mouse models. In parallel, previous results have probed that administration of granulocyte-colony stimulating factor (G-CSF) increased immune infiltration in tumors. We then hypothesized that the mobilization of immune cells by G-CSF may increase the antitumor efficacy of Celyvir treatment by increasing the immune infiltration into the tumors. METHODS In this study, we use a murine version of Celyvir consisting in murine MSCs carrying the murine OAd dlE102-here called OAd-MSCs-in an immunocompetent model of osteosarcoma. We tested the antitumoral efficacy of the combination of OAd-MSCs plus G-CSF. RESULTS Our results show that treatment with OAd-MSCs or the union of OAd-MSCs with G-CSF (Combination) significantly reduced tumor growth of osteosarcoma in vivo. Moreover, treated tumors presented higher tumor infiltration of immune cells-especially tumor-infiltrating lymphocytes-and reduced T cell exhaustion, which seems to be enhanced in tumors treated with the Combination. The comparison of our results to those obtained from a cohort of pediatric osteosarcoma patients showed that the virotherapy induces immunological changes similar to those observed in patients with good prognosis. CONCLUSIONS The results open the possibility of using cellular virotherapy for the treatment of bone cancers. Indeed, its combination with G-CSF may be considered for the improvement of the therapy.
Collapse
Affiliation(s)
| | - Stefano Gambera
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, Madrid, Spain
| | - Angela Leo
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Firenze, Italy
| | | |
Collapse
|
18
|
Bulcha JT, Wang Y, Ma H, Tai PWL, Gao G. Viral vector platforms within the gene therapy landscape. Signal Transduct Target Ther 2021; 6:53. [PMID: 33558455 PMCID: PMC7868676 DOI: 10.1038/s41392-021-00487-6] [Citation(s) in RCA: 725] [Impact Index Per Article: 181.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/05/2020] [Accepted: 10/23/2020] [Indexed: 01/30/2023] Open
Abstract
Throughout its 40-year history, the field of gene therapy has been marked by many transitions. It has seen great strides in combating human disease, has given hope to patients and families with limited treatment options, but has also been subject to many setbacks. Treatment of patients with this class of investigational drugs has resulted in severe adverse effects and, even in rare cases, death. At the heart of this dichotomous field are the viral-based vectors, the delivery vehicles that have allowed researchers and clinicians to develop powerful drug platforms, and have radically changed the face of medicine. Within the past 5 years, the gene therapy field has seen a wave of drugs based on viral vectors that have gained regulatory approval that come in a variety of designs and purposes. These modalities range from vector-based cancer therapies, to treating monogenic diseases with life-altering outcomes. At present, the three key vector strategies are based on adenoviruses, adeno-associated viruses, and lentiviruses. They have led the way in preclinical and clinical successes in the past two decades. However, despite these successes, many challenges still limit these approaches from attaining their full potential. To review the viral vector-based gene therapy landscape, we focus on these three highly regarded vector platforms and describe mechanisms of action and their roles in treating human disease.
Collapse
Affiliation(s)
- Jote T Bulcha
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic medical sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hong Ma
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Phillip W L Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA.
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
19
|
Moreno R. Mesenchymal stem cells and oncolytic viruses: joining forces against cancer. J Immunother Cancer 2021; 9:e001684. [PMID: 33558278 PMCID: PMC7871674 DOI: 10.1136/jitc-2020-001684] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
The development of oncolytic viruses (OVs) has increased significantly in the past 20 years, with many candidates entering clinical trials and three of them receiving approval for some indications. Recently, OVs have also gathered interest as candidates to use in combination with immunotherapies for cancer due to their immunogenic properties, which include immunogenic cell death and the possibility to carry therapeutic transgenes in their genomes. OVs transform non-immunogenic 'cold' tumors into inflamed immunogenic 'hot' tumors, where immunotherapies show the highest efficacy. However, in monotherapy or in combination with immunotherapy, OVs face numerous challenges that limit their successful application, in particular upon systemic administration, such as liver sequestration, neutralizing interactions in blood, physical barriers to infection, and fast clearance by the immune system. In this regard, the use of mesenchymal stem cells (MSCs) as cells carrier for OV delivery addresses many of these obstacles acting as virus carriers and factories, expressing additional transgenes, and modulating the immune system. Here, I review the current progress of OVs-loaded MSCs in cancer, focusing on their interaction with the immune system, and discuss new strategies to improve their therapeutic efficacy.
Collapse
Affiliation(s)
- Rafael Moreno
- Virotherapy and immunotherapy group, ProCURE Program, Catalan Institute of Oncology - ICO, L'Hospitalet de Llobregat, Spain
- Cancer Virotherapy group, Oncobell Program, Institutd'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Spain
| |
Collapse
|
20
|
Gómez A, Sardón D, Cejalvo T, Vázquez F, García-Castro J, Perisé-Barrios AJ. Biodistribution Analysis of Oncolytic Adenoviruses in Canine Patient Necropsy Samples Treated with Cellular Virotherapy. Mol Ther Oncolytics 2020; 18:525-534. [PMID: 32995478 PMCID: PMC7490470 DOI: 10.1016/j.omto.2020.08.006] [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: 04/05/2020] [Accepted: 08/10/2020] [Indexed: 11/17/2022] Open
Abstract
Oncolytic immunotherapy with competent viruses is an emerging approach in cancer treatment. The clinical safety of many types of oncolytic viruses (OVs) has been demonstrated. However, there is a lack of information about viral biodistribution in patients. The available data about oncolytic adenovirus biodistribution in human subjects treated intravenously consists of virus detection in body fluids, a few tumor biopsies, and a single report of patient necropsy samples. There is no information about adenoviral biodistribution in patients treated intravenously with cellular vehicles carrying an oncolytic adenovirus. We previously published reports regarding the efficacy and clinical safety of infusing mesenchymal stem cells (MSCs) infected with an OV in human and canine patients. In this study, we performed necropsies on 12 canine patients treated with dCelyvir, canine MSCs infected with ICOCAV17, a canine oncolytic adenovirus. The prevalence of microscopic lesions, especially chronic inflammatory responses in different organs, was higher than expected. Concomitantly, we found a positive immunoreaction to ICOCAV17 in analyzed samples. These findings support a possible role of the virus in development of histopathological alterations and ongoing systemic viral replication of ICOCAV17 in the period after therapy administration.
Collapse
Affiliation(s)
- Ana Gómez
- Veterinary Pathology Unit, Universidad Alfonso X el Sabio, 28691 Madrid, Spain
| | - David Sardón
- Veterinary Pathology Unit, Universidad Alfonso X el Sabio, 28691 Madrid, Spain
| | - Teresa Cejalvo
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Fernando Vázquez
- Veterinary Pathology Unit, Universidad Alfonso X el Sabio, 28691 Madrid, Spain
| | - Javier García-Castro
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain
- Biomedical Research Unit, Universidad Alfonso X el Sabio, 28691 Madrid, Spain
| | - Ana Judith Perisé-Barrios
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain
- Biomedical Research Unit, Universidad Alfonso X el Sabio, 28691 Madrid, Spain
| |
Collapse
|
21
|
Cellular Virotherapy Increases Tumor-Infiltrating Lymphocytes (TIL) and Decreases their PD-1 + Subsets in Mouse Immunocompetent Models. Cancers (Basel) 2020; 12:cancers12071920. [PMID: 32708639 PMCID: PMC7409201 DOI: 10.3390/cancers12071920] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Oncolytic virotherapy uses viruses designed to selectively replicate in cancer cells. An alternative to intratumoral administration is to use mesenchymal stem cells (MSCs) to transport the oncolytic viruses to the tumor site. Following this strategy, our group has already applied this treatment to children and adults in a human clinical trial and a veterinary trial, with good clinical responses and excellent safety profiles. However, the development of immunocompetent cancer mouse models is still necessary for the study and improvement of oncolytic viroimmunotherapies. Here we have studied the antitumor efficacy, immune response, and mechanism of action of a complete murine version of our cellular virotherapy in mouse models of renal adenocarcinoma and melanoma. We used mouse MSCs infected with the mouse oncolytic adenovirus dlE102 (OAd-MSCs). In both models, treatment with OAd-MSCs significantly reduced tumor volumes by 50% and induced a pro-inflammatory tumor microenvironment. Furthermore, treated mice harboring renal adenocarcinoma and melanoma tumors presented increased infiltration of tumor-associated macrophages (TAMs), natural killer cells, and tumor-infiltrating lymphocytes (TILs). Treated mice also presented lower percentage of TILs expressing programmed cell death protein 1 (PD-1)-the major regulator of T cell exhaustion. In conclusion, treatment with OAd-MSCs significantly reduced tumor volume and induced changes in tumor-infiltrating populations of melanoma and renal cancer.
Collapse
|
22
|
Rodríguez-Milla MÁ, Morales-Molina A, Perisé-Barrios AJ, Cejalvo T, García-Castro J. AKT and JUN are differentially activated in mesenchymal stem cells after infection with human and canine oncolytic adenoviruses. Cancer Gene Ther 2020; 28:64-73. [PMID: 32457488 DOI: 10.1038/s41417-020-0184-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023]
Abstract
There is increasing evidence about the use of oncolytic adenoviruses (Ads) as promising immunotherapy agents. We have previously demonstrated the clinical efficiency of mesenchymal stem cells (MSCs) infected with oncolytic Ads as an antitumoral immunotherapy (called Celyvir) in human and canine patients, using ICOVIR-5 or ICOCAV17 as human and canine oncolytic Ads, respectively. Considering the better clinical outcomes of canine patients, in this study we searched for differences in cellular responses of human and canine MSCs to Ad infection that may help understand the mechanisms leading to higher antitumor immune response. We found that infection of human and canine MSCs with ICOVIR-5 or ICOCAV17 did not activate the NF-κB pathway or the interferon regulatory factors IRF3 and IRF7. However, we observed differences in the profile of cytokines secretion, as infection of canine MSCs with ICOCAV17 resulted in lower secretion of several cytokines. Moreover, we showed that infection of human MSCs with ICOVIR-5 increased the phosphorylation of a number of proteins, including AKT and c-JUN. Finally, we demonstrated that differences in regulation of AKT and c-JUN in human and canine MSCs by ICOVIR-5 or ICOCAV17 are intrinsic to each virus. Our findings suggest that ICOCAV17 induces a more limited host response in canine MSCs, which may be related to a better clinical outcome. This result opens the possibility to develop new human oncolytic Ads with these specific properties. In addition, this improvement could be imitated by selecting specific human MSC on the basis of a limited host response after Ad infection.
Collapse
Affiliation(s)
| | | | - Ana Judith Perisé-Barrios
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220, Madrid, Spain.,Biomedical Research Unit, Universidad Alfonso X el Sabio, 28691, Madrid, Spain
| | - Teresa Cejalvo
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220, Madrid, Spain
| | - Javier García-Castro
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220, Madrid, Spain.
| |
Collapse
|
23
|
Franco-Luzón L, García-Mulero S, Sanz-Pamplona R, Melen G, Ruano D, Lassaletta Á, Madero L, González-Murillo Á, Ramírez M. Genetic and Immune Changes Associated with Disease Progression under the Pressure of Oncolytic Therapy in A Neuroblastoma Outlier Patient. Cancers (Basel) 2020; 12:cancers12051104. [PMID: 32354143 PMCID: PMC7281487 DOI: 10.3390/cancers12051104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/25/2020] [Accepted: 04/26/2020] [Indexed: 12/19/2022] Open
Abstract
Little is known about the effect of oncolytic adenovirotherapy on pediatric tumors. Here we present the clinical case of a refractory neuroblastoma that responded positively to Celyvir (ICOVIR-5 oncolytic adenovirus delivered by autologous mesenchymal stem cells) for several months. We analyzed samples during tumor evolution in order to identify molecular and mutational features that could explain the interactions between treatment and tumor and how the balance between both of them evolved. We identified a higher adaptive immune infiltration during stabilized disease compared to progression, and also a higher mutational rate and T-cell receptor (TCR) diversity during disease progression. Our results indicate an initial active role of the immune system controlling tumor growth during Celyvir therapy. The tumor eventually escaped from the control exerted by virotherapy through acquisition of resistance by the tumor microenvironment that exhausted the initial T cell response.
Collapse
Affiliation(s)
- Lidia Franco-Luzón
- Children Oncohematology Foundation, 28079 Madrid, Spain; (L.F.-L.); (L.M.)
| | - Sandra García-Mulero
- Department of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain;
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, L’Hospitalet de Llobregat, 08908 Barcelona, Spain;
| | - Rebeca Sanz-Pamplona
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, L’Hospitalet de Llobregat, 08908 Barcelona, Spain;
| | - Gustavo Melen
- Biomedical Research Foundation, Niño Jesús Children Hospital, 28009 Madrid, Spain; (G.M.); (Á.G.-M.)
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
| | - David Ruano
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
| | - Álvaro Lassaletta
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
| | - Luís Madero
- Children Oncohematology Foundation, 28079 Madrid, Spain; (L.F.-L.); (L.M.)
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
- Oncohematology Unit, Hospital Infantil Universitario Niño Jesús, 28009 Madrid, Spain
| | - África González-Murillo
- Biomedical Research Foundation, Niño Jesús Children Hospital, 28009 Madrid, Spain; (G.M.); (Á.G.-M.)
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
| | - Manuel Ramírez
- Biomedical Research Foundation, Niño Jesús Children Hospital, 28009 Madrid, Spain; (G.M.); (Á.G.-M.)
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
- Correspondence: ; Tel.: +34-9150-35938
| |
Collapse
|
24
|
Pascual-Pasto G, Bazan-Peregrino M, Olaciregui NG, Restrepo-Perdomo CA, Mato-Berciano A, Ottaviani D, Weber K, Correa G, Paco S, Vila-Ubach M, Cuadrado-Vilanova M, Castillo-Ecija H, Botteri G, Garcia-Gerique L, Moreno-Gilabert H, Gimenez-Alejandre M, Alonso-Lopez P, Farrera-Sal M, Torres-Manjon S, Ramos-Lozano D, Moreno R, Aerts I, Doz F, Cassoux N, Chapeaublanc E, Torrebadell M, Roldan M, König A, Suñol M, Claverol J, Lavarino C, Carmen de T, Fu L, Radvanyi F, Munier FL, Catalá-Mora J, Mora J, Alemany R, Cascalló M, Chantada GL, Carcaboso AM. Therapeutic targeting of the RB1 pathway in retinoblastoma with the oncolytic adenovirus VCN-01. Sci Transl Med 2020; 11:11/476/eaat9321. [PMID: 30674657 DOI: 10.1126/scitranslmed.aat9321] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/22/2018] [Indexed: 12/12/2022]
Abstract
Retinoblastoma is a pediatric solid tumor of the retina activated upon homozygous inactivation of the tumor suppressor RB1 VCN-01 is an oncolytic adenovirus designed to replicate selectively in tumor cells with high abundance of free E2F-1, a consequence of a dysfunctional RB1 pathway. Thus, we reasoned that VCN-01 could provide targeted therapeutic activity against even chemoresistant retinoblastoma. In vitro, VCN-01 effectively killed patient-derived retinoblastoma models. In mice, intravitreous administration of VCN-01 in retinoblastoma xenografts induced tumor necrosis, improved ocular survival compared with standard-of-care chemotherapy, and prevented micrometastatic dissemination into the brain. In juvenile immunocompetent rabbits, VCN-01 did not replicate in retinas, induced minor local side effects, and only leaked slightly and for a short time into the blood. Initial phase 1 data in patients showed the feasibility of the administration of intravitreous VCN-01 and resulted in antitumor activity in retinoblastoma vitreous seeds and evidence of viral replication markers in tumor cells. The treatment caused local vitreous inflammation but no systemic complications. Thus, oncolytic adenoviruses targeting RB1 might provide a tumor-selective and chemotherapy-independent treatment option for retinoblastoma.
Collapse
Affiliation(s)
- Guillem Pascual-Pasto
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | | | - Nagore G Olaciregui
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | | | | | - Daniela Ottaviani
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Institut Curie, PSL Research University, 75248 Paris, France
| | - Klaus Weber
- AnaPath GmbH, Oberbuchsiten 4625, Switzerland
| | - Genoveva Correa
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Sonia Paco
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Monica Vila-Ubach
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Maria Cuadrado-Vilanova
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Helena Castillo-Ecija
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Gaia Botteri
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Laura Garcia-Gerique
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Helena Moreno-Gilabert
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | | | | | | | - Silvia Torres-Manjon
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Dolores Ramos-Lozano
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael Moreno
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Isabelle Aerts
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Institut Curie, PSL Research University, 75248 Paris, France
| | - François Doz
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Paris Descartes University, 75006 Paris, France
| | - Nathalie Cassoux
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Paris Descartes University, 75006 Paris, France.,Institut Curie, Ophthalmic Oncology, 75248 Paris, France
| | - Elodie Chapeaublanc
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Institut Curie, PSL Research University, 75248 Paris, France
| | - Montserrat Torrebadell
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Monica Roldan
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pathology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Andrés König
- Vivotecnia Research S.L., Tres Cantos, Madrid 28760, Spain
| | - Mariona Suñol
- Pathology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Joana Claverol
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Clinical Trials Unit, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Cinzia Lavarino
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Torres Carmen de
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Ligia Fu
- Pediatric Hematology-Oncology, Hospital Escuela Universitario, Tegucigalpa, Honduras
| | - François Radvanyi
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Institut Curie, PSL Research University, 75248 Paris, France
| | | | | | - Jaume Mora
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Ramón Alemany
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Manel Cascalló
- VCN Biosciences, Sant Cugat del Valles, Barcelona 08174, Spain
| | - Guillermo L Chantada
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain.,Hospital de Pediatria JP Garrahan, Buenos Aires 1245, Argentina.,CONICET, Buenos Aires 1245, Argentina
| | - Angel M Carcaboso
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain. .,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| |
Collapse
|
25
|
Ruano D, López-Martín JA, Moreno L, Lassaletta Á, Bautista F, Andión M, Hernández C, González-Murillo Á, Melen G, Alemany R, Madero L, García-Castro J, Ramírez M. First-in-Human, First-in-Child Trial of Autologous MSCs Carrying the Oncolytic Virus Icovir-5 in Patients with Advanced Tumors. Mol Ther 2020; 28:1033-1042. [PMID: 32053771 DOI: 10.1016/j.ymthe.2020.01.019] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
We present here the results of a first-in-human, first-in-child trial for patients with relapsed/refractory solid tumors using Celyvir, an advanced therapy medicine that combines autologous mesenchymal stem cells (MSCs) carrying an oncolytic adenovirus. Celyvir was manufactured from a bone marrow aspirate and then given intravenously. Patients received weekly infusions for 6 weeks at a dose of 2 × 106 cells/kg (children) or 0.5-1 × 106 cells/kg (adults), 2 × 104 viral particles per cell. Fifteen pediatric and 19 adult patients were recruited, but 18 were screen failures, mainly because rapid disease progression before Celyvir was available. No grade 2-5 toxicities were reported. Adenoviral replication detected by PCR was found in all but 2 pediatric patient and in none of the adult ones. Absolute numbers of circulating leukocytes suffered minor changes along therapy, but some subsets showed differences comparing the pediatric versus the adult cohorts. Two patients with neuroblastoma showed disease stabilization, and one of them continued on treatment for up to 6 additional weeks. Celyvir, the combination of MSCs and oncolytic adenovirus, is safe and warrants further evaluation in a phase 2 setting. The use of MSCs may be a strategy to increase the amount of oncolytic virus administered to patients, minimizing toxicities and avoiding direct tumor injections.
Collapse
Affiliation(s)
- David Ruano
- Servicio de Oncología, Hospital Universitario Niño Jesús, Madrid, Spain
| | - José A López-Martín
- Servicio de Oncología Médica, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Lucas Moreno
- Servicio de Oncología, Hospital Universitario Niño Jesús, Madrid, Spain
| | - Álvaro Lassaletta
- Servicio de Oncología, Hospital Universitario Niño Jesús, Madrid, Spain
| | | | - Maitane Andión
- Servicio de Oncología, Hospital Universitario Niño Jesús, Madrid, Spain
| | - Carmen Hernández
- Servicio de Oncología, Hospital Universitario Niño Jesús, Madrid, Spain
| | | | - Gustavo Melen
- Fundación de Investigación Biomédica, Hospital Universitario Niño Jesús, Madrid, Spain
| | - Ramón Alemany
- Institut Català d'Oncologia-IDIBELL, Barcelona, Spain
| | - Luis Madero
- Servicio de Oncología, Hospital Universitario Niño Jesús, Madrid, Spain
| | | | - Manuel Ramírez
- Servicio de Oncología, Hospital Universitario Niño Jesús, Madrid, Spain.
| |
Collapse
|
26
|
Garcia-Moure M, Martinez-Velez N, Gonzalez-Huarriz M, Marrodán L, Cascallo M, Alemany R, Patiño-García A, Alonso MM. The oncolytic adenovirus VCN-01 promotes anti-tumor effect in primitive neuroectodermal tumor models. Sci Rep 2019; 9:14368. [PMID: 31591461 PMCID: PMC6779892 DOI: 10.1038/s41598-019-51014-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/23/2019] [Indexed: 01/14/2023] Open
Abstract
Last advances in the treatment of pediatric tumors has led to an increase of survival rates of children affected by primitive neuroectodermal tumors, however, still a significant amount of the patients do not overcome the disease. In addition, the survivors might suffer from severe side effects caused by the current standard treatments. Oncolytic virotherapy has emerged in the last years as a promising alternative for the treatment of solid tumors. In this work, we study the anti-tumor effect mediated by the oncolytic adenovirus VCN-01 in CNS-PNET models. VCN-01 is able to infect and replicate in PNET cell cultures, leading to a cytotoxicity and immunogenic cell death. In vivo, VCN-01 increased significantly the median survival of mice and led to long-term survivors in two orthotopic models of PNETs. In summary, these results underscore the therapeutic effect of VCN-01 for rare pediatric cancers such as PNETs, and warrants further exploration on the use of this virus to treat them.
Collapse
Affiliation(s)
- Marc Garcia-Moure
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, 31008, Spain
| | - Naiara Martinez-Velez
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, 31008, Spain
| | - Marisol Gonzalez-Huarriz
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, 31008, Spain
| | - Lucía Marrodán
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, 31008, Spain
| | - Manel Cascallo
- VCN Biosciences, Sant Cugat del Vallés, 08174, Barcelona, Spain
| | - Ramón Alemany
- Translational Research Laboratory, IDIBELL-Institut Catalá d'Oncologia, L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Ana Patiño-García
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, 31008, Spain
| | - Marta M Alonso
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain. .,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain. .,Department of Pediatrics, University Hospital of Navarra, Pamplona, 31008, Spain.
| |
Collapse
|
27
|
Brachtlova T, van Beusechem VW. Unleashing the Full Potential of Oncolytic Adenoviruses against Cancer by Applying RNA Interference: The Force Awakens. Cells 2018; 7:cells7120228. [PMID: 30477117 PMCID: PMC6315459 DOI: 10.3390/cells7120228] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/23/2022] Open
Abstract
Oncolytic virus therapy of cancer is an actively pursued field of research. Viruses that were once considered as pathogens threatening the wellbeing of humans and animals alike are with every passing decade more prominently regarded as vehicles for genetic and oncolytic therapies. Oncolytic viruses kill cancer cells, sparing healthy tissues, and provoke an anticancer immune response. Among these viruses, recombinant adenoviruses are particularly attractive agents for oncolytic immunotherapy of cancer. Different approaches are currently examined to maximize their therapeutic effect. Here, knowledge of virus–host interactions may lead the way. In this regard, viral and host microRNAs are of particular interest. In addition, cellular factors inhibiting viral replication or dampening immune responses are being discovered. Therefore, applying RNA interference is an attractive approach to strengthen the anticancer efficacy of oncolytic viruses gaining attention in recent years. RNA interference can be used to fortify the virus’ cancer cell-killing and immune-stimulating properties and to suppress cellular pathways to cripple the tumor. In this review, we discuss different ways of how RNA interference may be utilized to increase the efficacy of oncolytic adenoviruses, to reveal their full potential.
Collapse
Affiliation(s)
- Tereza Brachtlova
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, De Boelelaan 1117, 1007 MB Amsterdam, The Netherlands.
| | - Victor W van Beusechem
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, De Boelelaan 1117, 1007 MB Amsterdam, The Netherlands.
| |
Collapse
|
28
|
García M, Moreno R, Gil-Martin M, Cascallò M, de Olza MO, Cuadra C, Piulats JM, Navarro V, Domenech M, Alemany R, Salazar R. A Phase 1 Trial of Oncolytic Adenovirus ICOVIR-5 Administered Intravenously to Cutaneous and Uveal Melanoma Patients. Hum Gene Ther 2018; 30:352-364. [PMID: 30234393 DOI: 10.1089/hum.2018.107] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Oncolytic viruses represent a unique type of agents that combine self-amplification, lytic, and immunostimulatory properties against tumors. A local and locoregional clinical benefit has been demonstrated upon intratumoral injections of an oncolytic herpes virus in melanoma patients, leading to its approval in the United States and Europe for patients without visceral disease (up to stage IVM1a). However, in order to debulk and change the local immunosuppressive environment of tumors that cannot be injected directly, oncolyitc viruses need to be administered systemically. Among different viruses, adenovirus has been extensively used in clinical trials but with few evidences of activity upon systemic administration. Preclinical efficacy of a single intravenous administration of our oncolytic adenovirus ICOVIR5, an adenovirus type 5 responsive to the retinoblastoma pathway commonly deregulated in tumors, led us to use this virus in a dose-escalation phase 1 trial in metastatic melanoma patients. The results in 12 patients treated with a single infusion of a dose up to 1 × 1013 viral particles show that ICOVIR5 can reach melanoma metastases upon a single intravenous administration but fails to induce tumor regressions. These results support the systemic administration of armed oncolytic viruses to treat disseminated cancer.
Collapse
Affiliation(s)
- Margarita García
- 1 Clinical Research Unit, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Rafael Moreno
- 2 ProCure and Oncobell Programs, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Marta Gil-Martin
- 3 Department of Medical Oncology, Oncobell Program, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Manel Cascallò
- 2 ProCure and Oncobell Programs, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain.,4 VCN Biosciences, Sant Cugat del Valles, Barcelona, Spain
| | - Maria Ochoa de Olza
- 3 Department of Medical Oncology, Oncobell Program, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Carmen Cuadra
- 1 Clinical Research Unit, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Josep Maria Piulats
- 3 Department of Medical Oncology, Oncobell Program, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Valentin Navarro
- 1 Clinical Research Unit, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Marta Domenech
- 3 Department of Medical Oncology, Oncobell Program, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Ramon Alemany
- 2 ProCure and Oncobell Programs, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Ramon Salazar
- 3 Department of Medical Oncology, Oncobell Program, Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| |
Collapse
|
29
|
Moreno R, Fajardo CA, Farrera-Sal M, Perisé-Barrios AJ, Morales-Molina A, Al-Zaher AA, García-Castro J, Alemany R. Enhanced Antitumor Efficacy of Oncolytic Adenovirus-loaded Menstrual Blood-derived Mesenchymal Stem Cells in Combination with Peripheral Blood Mononuclear Cells. Mol Cancer Ther 2018; 18:127-138. [PMID: 30322950 DOI: 10.1158/1535-7163.mct-18-0431] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/16/2018] [Accepted: 10/10/2018] [Indexed: 11/16/2022]
Abstract
Several studies have evaluated the efficacy of using human oncolytic adenovirus (OAdv)-loaded mesenchymal stem cells (MSC) for cancer treatment. For example, we have described the antitumor efficacy of CELYVIR, autologous bone marrow-mesenchymal stem cells infected with the OAdv ICOVIR-5, for treatment of patients with neuroblastoma. Results from this clinical trial point out the role of the immune system in the clinical outcome. In this context, a better understanding of the immunophenotypic changes of human MSCs upon adenoviral infection and how these changes affect human autologous or allogeneic peripheral blood mononuclear cells (PBMC) could guide strategies to improve the antitumor efficacy of infected MSCs. In this work, we show how infection by an OAdv induces toll-like receptor 9 overexpression and activation of the NFĸB pathway in menstrual blood-derived MSCs, leading to a specific cytokine secretion profile. Moreover, a proinflammatory environment, mainly mediated by monocyte activation that leads to the activation of both T cells and natural killer cells (NK cell), is generated when OAdv-loaded MSCs are cocultured with allogeneic PBMCs. This combination of allogeneic PBMCs and OAdv-loaded MSCs enhances antitumor efficacy both in vitro and in vivo, an effect partially mediated by monocytes and NK cells. Altogether our results demonstrate not only the importance of the immune system for the OAdv-loaded MSCs antitumor efficacy, but in particular the benefits of using allogeneic MSCs for this therapy.
Collapse
Affiliation(s)
- Rafael Moreno
- Virotherapy and Gene therapy Group, ProCure Program, Translational Research Laboratory, Instituto Catalan de Oncología-IDIBELL, Barcelona, Spain.
| | - Carlos Alberto Fajardo
- Virotherapy and Gene therapy Group, ProCure Program, Translational Research Laboratory, Instituto Catalan de Oncología-IDIBELL, Barcelona, Spain
| | - Marti Farrera-Sal
- Virotherapy and Gene therapy Group, ProCure Program, Translational Research Laboratory, Instituto Catalan de Oncología-IDIBELL, Barcelona, Spain
- VCN Biosciences S.L., Grifols Corporate Offices, Sant Cugat del Vallès, Spain
| | | | - Alvaro Morales-Molina
- Cellular Biotechnology Unit, Institute of Health Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Ahmed Abdullah Al-Zaher
- Virotherapy and Gene therapy Group, ProCure Program, Translational Research Laboratory, Instituto Catalan de Oncología-IDIBELL, Barcelona, Spain
| | - Javier García-Castro
- Cellular Biotechnology Unit, Institute of Health Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Ramon Alemany
- Virotherapy and Gene therapy Group, ProCure Program, Translational Research Laboratory, Instituto Catalan de Oncología-IDIBELL, Barcelona, Spain
| |
Collapse
|
30
|
Morales-Molina Á, Gambera S, Cejalvo T, Moreno R, Rodríguez-Milla MÁ, Perisé-Barrios AJ, García-Castro J. Antitumor virotherapy using syngeneic or allogeneic mesenchymal stem cell carriers induces systemic immune response and intratumoral leukocyte infiltration in mice. Cancer Immunol Immunother 2018; 67:1589-1602. [PMID: 30066102 PMCID: PMC11028294 DOI: 10.1007/s00262-018-2220-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 07/27/2018] [Indexed: 12/29/2022]
Abstract
Oncolytic virotherapy uses oncolytic viruses that selectively replicate in cancer cells. The use of cellular vehicles with migration ability to tumors has been considered to increase their delivery to target sites. Following this approach, the antitumor efficacy of the treatment Celyvir (mesenchymal stem cells infected with the oncolytic adenovirus ICOVIR-5) has been demonstrated in patients with neuroblastoma. However, the better efficacy of syngeneic or allogeneic mesenchymal stem cells as cell carriers and the specific role of the immune system in this therapy are still unknown. In this study we use our virotherapy Celyvir with syngeneic and allogeneic mouse mesenchymal stem cells to determine their antitumor efficacy in a C57BL/6 murine adenocarcinoma model. Adoptive transfer of splenocytes from treated mice to new tumor-bearing mice followed by a secondary adoptive transfer to a third group was performed. Similar reduction of tumor growth and systemic activation of the innate and adaptive immune system was observed in groups treated with syngeneic or allogeneic mesenchymal stem cells loaded with ICOVIR-5. Moreover, a different pattern of infiltration was observed by immunofluorescence in Celyvir-treated groups. While non-treated tumors presented higher density of infiltrating immune cells in the periphery of the tumor, both syngeneic and allogeneic Celyvir-treated groups presented higher infiltration of CD45+ cells in the core of the tumor. Therefore, these results suggest that syngeneic and allogeneic Celyvir induce systemic activation of the immune system, similar antitumor effect and a higher intratumoral infiltration of leukocytes.
Collapse
Affiliation(s)
- Álvaro Morales-Molina
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, lab. 51-00-031, Ctra Majadahonda-Pozuelo, Km 2, Majadahonda, 28220, Madrid, Spain
| | - Stefano Gambera
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, lab. 51-00-031, Ctra Majadahonda-Pozuelo, Km 2, Majadahonda, 28220, Madrid, Spain
| | - Teresa Cejalvo
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, lab. 51-00-031, Ctra Majadahonda-Pozuelo, Km 2, Majadahonda, 28220, Madrid, Spain
| | - Rafael Moreno
- Virotherapy and Gene therapy Group, ProCure Program, Translational Research Laboratory, Instituto Catalan de Oncologia-IDIBELL, Barcelona, Spain
| | - Miguel Ángel Rodríguez-Milla
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, lab. 51-00-031, Ctra Majadahonda-Pozuelo, Km 2, Majadahonda, 28220, Madrid, Spain
| | - Ana Judith Perisé-Barrios
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, lab. 51-00-031, Ctra Majadahonda-Pozuelo, Km 2, Majadahonda, 28220, Madrid, Spain
| | - Javier García-Castro
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, lab. 51-00-031, Ctra Majadahonda-Pozuelo, Km 2, Majadahonda, 28220, Madrid, Spain.
| |
Collapse
|
31
|
Cejalvo T, Perisé-Barrios AJ, del Portillo I, Laborda E, Rodriguez-Milla MA, Cubillo I, Vázquez F, Sardón D, Ramirez M, Alemany R, del Castillo N, García-Castro J. Remission of Spontaneous Canine Tumors after Systemic Cellular Viroimmunotherapy. Cancer Res 2018; 78:4891-4901. [DOI: 10.1158/0008-5472.can-17-3754] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/17/2018] [Accepted: 07/05/2018] [Indexed: 11/16/2022]
|
32
|
Rincón E, Cejalvo T, Kanojia D, Alfranca A, Rodríguez-Milla MÁ, Gil Hoyos RA, Han Y, Zhang L, Alemany R, Lesniak MS, García-Castro J. Mesenchymal stem cell carriers enhance antitumor efficacy of oncolytic adenoviruses in an immunocompetent mouse model. Oncotarget 2018; 8:45415-45431. [PMID: 28525366 PMCID: PMC5542197 DOI: 10.18632/oncotarget.17557] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 04/18/2017] [Indexed: 12/28/2022] Open
Abstract
Oncolytic virotherapy represents a promising alternative for cancer treatment; however, viral delivery to the tumor represents a major challenge. Mesenchymal stem cells (MSCs) chemotax to tumors, and can serve as a viral delivery tool. Previously, we demonstrated antitumor therapeutic efficacy for mesenchymal stem cells (MSCs) infected with the oncolytic human adenovirus ICOVIR5 (Celyvir) for treatment of neuroblastoma patients. Given the lack of suitable immunocompetent preclinical models, the mechanism underlying Celyvir antitumor activity remains unknown. In this study, we used the syngeneic murine CMT64 cell line as a human adenovirus-semi-permissive tumor model and demonstrate the homing capacity of mouse Celyvir (mCelyvir) to CMT64 tumors. We found that the combined treatment of mCelyvir and intratumoral injections (i.t.) of ICOVIR5 was more effective than treatment with i.t. ICOVIR5 alone. Interestingly, the superior therapeutic effect of the combined therapy was associated with a higher tumor infiltration of CD8+ and CD4+ T cells. Our findings suggest that the use of MSCs as carriers of oncolytic adenovirus can improve the clinical efficacy of anti-cancer virotherapy, not only by driving the adenovirus to tumors, but also through their potential to recruit T cells.
Collapse
Affiliation(s)
- Esther Rincón
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Madrid, Spain.,The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA
| | - Teresa Cejalvo
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Madrid, Spain
| | - Deepak Kanojia
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA
| | - Arantzazu Alfranca
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Yu Han
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA
| | - Lingjiao Zhang
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA
| | - Ramón Alemany
- Institut Català d´Oncologia, IDIBELL, Barcelona, Spain
| | - Maciej S Lesniak
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
33
|
Promotion of Cell-Based Therapy: Special Focus on the Cooperation of Mesenchymal Stem Cell Therapy and Gene Therapy for Clinical Trial Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1119:103-118. [PMID: 30155859 DOI: 10.1007/5584_2018_256] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Regenerative medicine (RM) is a promising new field of medicine that has mobilized several new tools to repair or replace lost or damaged cells or tissues by stimulating natural regenerative mechanisms nearby cell and tissue-based therapy approaches. However, mesenchymal stem cell (MSC) based therapy has been shown to be safe and effective to a certain degree in multiple clinical trial studies (CTSs) of several diseases, in most MSC CTSs the efficacy of treatment has been reported low. Therefore, researchers have focused on efficacy enhancing of MSC to improve migratory and homing, survival, stemness, differentiation and other therapeutic applicable properties by using different approaches. Gene therapy is one of the experimental technique tools that uses genes to change cells for therapeutic and investigation purposes. In this study has been focused on genetically modified MSCs for use in RM with an emphasis on CTSs. We highlight the basic concept of genetic modifications and also discuss recent clinical studies aspects. Recently reviewed studies show that MSC therapy with assistant gene therapy can be used in cancer therapy, heart diseases, Fanconi anemia and several other diseases.
Collapse
|
34
|
Human Menstrual Blood-Derived Mesenchymal Stem Cells as Potential Cell Carriers for Oncolytic Adenovirus. Stem Cells Int 2017; 2017:3615729. [PMID: 28781596 PMCID: PMC5525077 DOI: 10.1155/2017/3615729] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/19/2017] [Accepted: 05/04/2017] [Indexed: 12/21/2022] Open
Abstract
Antitumor efficacy of systemically administered oncolytic adenoviruses (OAdv) is limited due to diverse factors such as liver sequestration, neutralizing interactions in blood, elimination by the immune system, and physical barriers in tumors. It is therefore of clinical relevance to improve OAdv bioavailability and tumor delivery. Among the variety of tumor-targeting strategies, the use of stem cells and specifically bone marrow-derived mesenchymal stem cells (BM-MSCs) is of particular interest due to their tumor tropism and immunomodulatory properties. Nonetheless, the invasive methods to obtain these cells, the low number of MSCs present in the bone marrow, and their restricted in vitro expansion represent major obstacles for their use in cancer treatments, pointing out the necessity to identify an alternative source of MSCs. Here, we have evaluated the use of menstrual blood-derived mesenchymal stem cells (MenSCs) as cell carriers for regional delivery of an OAdv in the tumor. Our results indicate that MenSCs can be isolated without invasive methods, they have an increased proliferation rate compared to BM-MSCs, and they can be efficiently infected with different serotype 5-based capsid-modified adenoviruses, leading to viral replication and release. In addition, our in vivo studies confirmed the tumor-homing properties of MenSCs after regional administration.
Collapse
|
35
|
Kaczorowski A, Hammer K, Liu L, Villhauer S, Nwaeburu C, Fan P, Zhao Z, Gladkich J, Groß W, Nettelbeck DM, Herr I. Delivery of improved oncolytic adenoviruses by mesenchymal stromal cells for elimination of tumorigenic pancreatic cancer cells. Oncotarget 2016; 7:9046-59. [PMID: 26824985 PMCID: PMC4891025 DOI: 10.18632/oncotarget.7031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/15/2016] [Indexed: 01/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is one of the most aggressive malignancies and has poor therapeutic options. We evaluated improved oncolytic adenoviruses (OAds), in which the adenoviral gene E1B19K was deleted or a TRAIL transgene was inserted. Bone marrow mesenchymal stromal cells (MSCs) served as carriers for protected and tumor-specific virus transfers. The infection competence, tumor migration, and oncolysis were measured in cancer stem cell (CSC) models of primary and established tumor cells and in tumor xenografts. All OAds infected and lysed CSCs and prevented colony formation. MSCs migrated into PDA spheroids without impaired homing capacity. Xenotransplantation of non-infected PDA cells mixed with infected tumor cells strongly reduced the tumor volume and the expression of the proliferation marker Ki67 along with a necrotic morphology. Adenoviral capsid protein was detected in tumor xenograft tissue after intravenous injection of infected MSCs, but not in normal tissue, implying tumor-specific migration. Likewise, direct in vivo treatment correlated with a strongly reduced tumor volume, lower expression of Ki67 and CD24, and enhanced activity of caspase 3. These data demonstrate that the improved OAds induced efficient oncolysis with the OAd-TRAIL as most promising candidate for future clinical application.
Collapse
Affiliation(s)
- Adam Kaczorowski
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katharina Hammer
- Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Li Liu
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Villhauer
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clifford Nwaeburu
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pei Fan
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Zhefu Zhao
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jury Gladkich
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Groß
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk M Nettelbeck
- Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ingrid Herr
- Surgical Research Section, Molecular OncoSurgery, Department of General and Transplantation Surgery, University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
36
|
Yumul R, Richter M, Lu ZZ, Saydaminova K, Wang H, Wang CHK, Carter D, Lieber A. Epithelial Junction Opener Improves Oncolytic Adenovirus Therapy in Mouse Tumor Models. Hum Gene Ther 2016; 27:325-37. [PMID: 26993072 DOI: 10.1089/hum.2016.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A central resistance mechanism in solid tumors is the maintenance of epithelial junctions between malignant cells that prevent drug penetration into the tumor. Human adenoviruses (Ads) have evolved mechanisms to breach epithelial barriers. For example, during Ad serotype 3 (Ad3) infection of epithelial tumor cells, massive amounts of subviral penton-dodecahedral particles (PtDd) are produced and released from infected cells to trigger the transient opening of epithelial junctions, thus facilitating lateral virus spread. We show here that an Ad3 mutant that is disabled for PtDd production is significantly less effective in killing of epithelial human xenograft tumors than the wild-type Ad3 virus. Intratumoral spread and therapeutic effect of the Ad3 mutant was enhanced by co-administration of a small recombinant protein (JO; produced in Escherichia coli) that incorporated the minimal junction opening domains of PtDd. We then demonstrated that co-administration of JO with replication-competent Ads that do not produce PtDd (Ad5, Ad35) resulted in greater attenuation of tumor growth than virus injection alone. Furthermore, we genetically modified a conditionally replicating Ad5-based oncolytic Ad (Ad5Δ24) to express a secreted form of JO upon replication in tumor cells. The JO-expressing virus had a significantly greater antitumor effect than the unmodified AdΔ24 version. Our findings indicate that epithelial junctions limit the efficacy of oncolytic Ads and that this problem can be address by co-injection or expression of JO. JO has also the potential for improving cancer therapy with other types of oncolytic viruses.
Collapse
Affiliation(s)
- Roma Yumul
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington
| | - Maximilian Richter
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington
| | - Zhuo-Zhuang Lu
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington.,2 National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing, PR China
| | - Kamola Saydaminova
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington
| | - Hongjie Wang
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington
| | | | - Darrick Carter
- 4 Compliment Corp. , Seattle, Washington.,5 PAI Life Sciences Inc. , Seattle, Washington
| | - André Lieber
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington.,4 Compliment Corp. , Seattle, Washington.,6 Department of Pathology, University of Washington , Seattle, Washington
| |
Collapse
|
37
|
Abstract
For decades, effective cancer gene therapy has been a tantalising prospect; for a therapeutic modality potentially able to elicit highly effective and selective responses, definitive efficacy outcomes have often seemed out of reach. However, steady progress in vector development and accumulated experience from previous clinical studies has finally led the field to its first licensed therapy. Following a pivotal phase III trial, Imlygic (talimogene laherparepvec/T-Vec) received US approval as a treatment for cutaneous and subcutaneous melanoma in October 2015, followed several weeks later by its European authorisation. These represent the first approvals for an oncolytic virotherapy. Imlygic is an advanced-generation herpesvirus-based vector optimised for oncolytic and immunomodulatory activities. Many other oncolytic agents currently remain in development, providing hope that current success will be followed by other diverse vectors that may ultimately come to constitute a new class of clinical anti-cancer agents. In this review, we discuss some of the key oncolytic viral agents developed in the adenovirus and herpesvirus classes, and the prospects for further enhancing their efficacy by combining them with novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alan E. Bilsland
- Institute of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, G61 1QH, UK
| | | | - T. R. Jeffry Evans
- Institute of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, G61 1QH, UK
| |
Collapse
|
38
|
Kasala D, Yoon AR, Hong J, Kim SW, Yun CO. Evolving lessons on nanomaterial-coated viral vectors for local and systemic gene therapy. Nanomedicine (Lond) 2016; 11:1689-713. [PMID: 27348247 DOI: 10.2217/nnm-2016-0060] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Viral vectors are promising gene carriers for cancer therapy. However, virus-mediated gene therapies have demonstrated insufficient therapeutic efficacy in clinical trials due to rapid dissemination to nontarget tissues and to the immunogenicity of viral vectors, resulting in poor retention at the disease locus and induction of adverse inflammatory responses in patients. Further, the limited tropism of viral vectors prevents efficient gene delivery to target tissues. In this regard, modification of the viral surface with nanomaterials is a promising strategy to augment vector accumulation at the target tissue, circumvent the host immune response, and avoid nonspecific interactions with the reticuloendothelial system or serum complement. In the present review, we discuss various chemical modification strategies to enhance the therapeutic efficacy of viral vectors delivered either locally or systemically. We conclude by highlighting the salient features of various nanomaterial-coated viral vectors and their prospects and directions for future research.
Collapse
Affiliation(s)
- Dayananda Kasala
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Jinwoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Sung Wan Kim
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea.,Department of Pharmaceutics & Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| |
Collapse
|
39
|
Kumar MD, Dravid A, Kumar A, Sen D. Gene therapy as a potential tool for treating neuroblastoma-a focused review. Cancer Gene Ther 2016; 23:115-24. [PMID: 27080224 DOI: 10.1038/cgt.2016.16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/12/2022]
Abstract
Neuroblastoma, a solid tumor caused by rapid division of undifferentiated neuroblasts, is the most common childhood malignancy affecting children aged <5 years. Several approaches and strategies developed and tested to cure neuroblastoma have met with limited success due to different reasons. Many oncogenes are deregulated during the onset and development of neuroblastoma and thus offer an opportunity to circumvent this disease if the expression of these genes is restored to normalcy. Gene therapy is a powerful tool with the potential to inhibit the deleterious effects of oncogenes by inserting corrected/normal genes into the genome. Both viral and non-viral vector-based gene therapies have been developed and adopted to deliver the target genes into neuroblastoma cells. These attempts have given hope to bringing in a new regime of treatment against neuroblastoma. A few gene-therapy-based treatment strategies have been tested in limited clinical trials yielding some positive results. This mini review is an attempt to provide an overview of the available options of gene therapy to treat neuroblastoma.
Collapse
Affiliation(s)
- M D Kumar
- School of Biosciences and Technology, Vellore Institute of Technology University, Vellore, Tamil Nadu, India
| | - A Dravid
- School of Biosciences and Technology, Vellore Institute of Technology University, Vellore, Tamil Nadu, India
| | - A Kumar
- School of Biosciences and Technology, Vellore Institute of Technology University, Vellore, Tamil Nadu, India
| | - D Sen
- School of Biosciences and Technology, Vellore Institute of Technology University, Vellore, Tamil Nadu, India.,Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology University, Vellore, Tamil Nadu, India
| |
Collapse
|
40
|
Yoon AR, Hong J, Kim SW, Yun CO. Redirecting adenovirus tropism by genetic, chemical, and mechanical modification of the adenovirus surface for cancer gene therapy. Expert Opin Drug Deliv 2016; 13:843-58. [PMID: 26967319 DOI: 10.1517/17425247.2016.1158707] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Despite remarkable advancements, clinical evaluations of adenovirus (Ad)-mediated cancer gene therapies have highlighted the need for improved delivery and targeting. AREA COVERED Genetic modification of Ad capsid proteins has been extensively attempted. Although genetic modification enhances the therapeutic potential of Ad, it is difficult to successfully incorporate extraneous moieties into the capsid and the engineering process is laborious. Recently, chemical modification of the Ad surface with nanomaterials and targeting moieties has been found to enhance Ad internalization into the target by both passive and active mechanisms. Alternatively, external stimulus-mediated targeting can result in selective accumulation of Ad in the tumor and prevent dissemination of Ad into surrounding nontarget tissues. In the present review, we discuss various genetic, chemical, and mechanical engineering strategies for overcoming the challenges that hinder the therapeutic efficacy of Ad-based approaches. EXPERT OPINION Surface modification of Ad by genetic, chemical, or mechanical engineering strategies enables Ad to overcome the shortcomings of conventional Ad and enhances delivery efficiency through distinct and unique mechanisms that unmodified Ad cannot mimic. However, although the therapeutic potential of Ad-mediated gene therapy has been enhanced by various surface modification strategies, each strategy still possesses innate limitations that must be addressed, requiring innovative ideas and designs.
Collapse
Affiliation(s)
- A-Rum Yoon
- a Department of Bioengineering, College of Engineering , Hanyang University , Seoul , Korea
| | - Jinwoo Hong
- a Department of Bioengineering, College of Engineering , Hanyang University , Seoul , Korea
| | - Sung Wan Kim
- a Department of Bioengineering, College of Engineering , Hanyang University , Seoul , Korea.,b Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry , University of Utah , Salt Lake City , UT , USA
| | - Chae-Ok Yun
- a Department of Bioengineering, College of Engineering , Hanyang University , Seoul , Korea
| |
Collapse
|
41
|
Influence of carrier cells on the clinical outcome of children with neuroblastoma treated with high dose of oncolytic adenovirus delivered in mesenchymal stem cells. Cancer Lett 2016; 371:161-70. [DOI: 10.1016/j.canlet.2015.11.036] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/24/2015] [Accepted: 11/27/2015] [Indexed: 12/22/2022]
|
42
|
Vera B, Martínez-Vélez N, Xipell E, Acanda de la Rocha A, Patiño-García A, Saez-Castresana J, Gonzalez-Huarriz M, Cascallo M, Alemany R, Alonso MM. Characterization of the Antiglioma Effect of the Oncolytic Adenovirus VCN-01. PLoS One 2016; 11:e0147211. [PMID: 26808201 PMCID: PMC4726573 DOI: 10.1371/journal.pone.0147211] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/30/2015] [Indexed: 02/04/2023] Open
Abstract
Despite the recent advances in the development of antitumor therapies, the prognosis for patients with malignant gliomas remains dismal. Therapy with tumor-selective viruses is emerging as a treatment option for this devastating disease. In this study we characterize the anti-glioma effect of VCN-01, an improved hyaluronidase-armed pRB-pathway-selective oncolytic adenovirus that has proven safe and effective in the treatment of several solid tumors. VCN-01 displayed a significant cytotoxic effect on glioma cells in vitro. In vivo, in two different orthotopic glioma models, a single intra-tumoral administration of VCN-01 increased overall survival significantly and led to long-term survivors free of disease.
Collapse
Affiliation(s)
- Beatriz Vera
- Navarra’s Health Research Institute (IDISNA) Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Dpt of Medical Oncology, University Hospital of Navarra, Pamplona 31008, Spain
| | - Naiara Martínez-Vélez
- Navarra’s Health Research Institute (IDISNA) Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Dpt of Medical Oncology, University Hospital of Navarra, Pamplona 31008, Spain
| | - Enric Xipell
- Navarra’s Health Research Institute (IDISNA) Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Dpt of Medical Oncology, University Hospital of Navarra, Pamplona 31008, Spain
| | - Arlet Acanda de la Rocha
- Navarra’s Health Research Institute (IDISNA) Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Dpt of Medical Oncology, University Hospital of Navarra, Pamplona 31008, Spain
| | - Ana Patiño-García
- Navarra’s Health Research Institute (IDISNA) Pamplona, Spain
- Dpt of Pediatrics, University Hospital of Navarra, Pamplona 31008, Spain
| | - Javier Saez-Castresana
- Navarra’s Health Research Institute (IDISNA) Pamplona, Spain
- Brain Tumor Biology Unit, University of Navarra School of Sciences, 31008 Pamplona, Spain
| | - Marisol Gonzalez-Huarriz
- Navarra’s Health Research Institute (IDISNA) Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Dpt of Medical Oncology, University Hospital of Navarra, Pamplona 31008, Spain
| | - Manel Cascallo
- VCN Biosciences, Sant Cugat del Vallés, 08174 Barcelona, Spain
| | - Ramón Alemany
- Translational Research Laboratory, IDIBELL-Institut Catalá d’Oncologia, L’Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Marta M. Alonso
- Navarra’s Health Research Institute (IDISNA) Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Dpt of Medical Oncology, University Hospital of Navarra, Pamplona 31008, Spain
- * E-mail:
| |
Collapse
|
43
|
Tsun A, Miao XN, Wang CM, Yu DC. Oncolytic Immunotherapy for Treatment of Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 909:241-83. [PMID: 27240460 DOI: 10.1007/978-94-017-7555-7_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Immunotherapy entails the treatment of disease by modulation of the immune system. As detailed in the previous chapters, the different modes of achieving immune modulation are many, including the use of small/large molecules, cellular therapy, and radiation. Oncolytic viruses that can specifically attack, replicate within, and destroy tumors represent one of the most promising classes of agents for cancer immunotherapy (recently termed as oncolytic immunotherapy). The notion of oncolytic immunotherapy is considered as the way in which virus-induced tumor cell death (known as immunogenic cancer cell death (ICD)) allows the immune system to recognize tumor cells and provide long-lasting antitumor immunity. Both immune responses toward the virus and ICD together contribute toward successful antitumor efficacy. What is now becoming increasingly clear is that monotherapies, through any of the modalities detailed in this book, are neither sufficient in eradicating tumors nor in providing long-lasting antitumor immune responses and that combination therapies may deliver enhanced efficacy. After the rise of the genetic engineering era, it has been possible to engineer viruses to harbor combination-like characteristics to enhance their potency in cancer immunotherapy. This chapter provides a historical background on oncolytic virotherapy and its future application in cancer immunotherapy, especially as a combination therapy with other treatment modalities.
Collapse
Affiliation(s)
- A Tsun
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - X N Miao
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - C M Wang
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - D C Yu
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China.
| |
Collapse
|
44
|
Uusi-Kerttula H, Hulin-Curtis S, Davies J, Parker AL. Oncolytic Adenovirus: Strategies and Insights for Vector Design and Immuno-Oncolytic Applications. Viruses 2015; 7:6009-42. [PMID: 26610547 PMCID: PMC4664994 DOI: 10.3390/v7112923] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 02/06/2023] Open
Abstract
Adenoviruses (Ad) are commonly used both experimentally and clinically, including oncolytic virotherapy applications. In the clinical area, efficacy is frequently hampered by the high rates of neutralizing immunity, estimated as high as 90% in some populations that promote vector clearance and limit bioavailability for tumor targeting following systemic delivery. Active tumor targeting is also hampered by the ubiquitous nature of the Ad5 receptor, hCAR, as well as the lack of highly tumor-selective targeting ligands and suitable targeting strategies. Furthermore, significant off-target interactions between the viral vector and cellular and proteinaceous components of the bloodstream have been documented that promote uptake into non-target cells and determine dose-limiting toxicities. Novel strategies are therefore needed to overcome the obstacles that prevent efficacious Ad deployment for wider clinical applications. The use of less seroprevalent Ad serotypes, non-human serotypes, capsid pseudotyping, chemical shielding and genetic masking by heterologous peptide incorporation are all potential strategies to achieve efficient vector escape from humoral immune recognition. Conversely, selective vector arming with immunostimulatory agents can be utilized to enhance their oncolytic potential by activation of cancer-specific immune responses against the malignant tissues. This review presents recent advantages and pitfalls occurring in the field of adenoviral oncolytic therapies.
Collapse
Affiliation(s)
- Hanni Uusi-Kerttula
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| | - Sarah Hulin-Curtis
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| | - James Davies
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| | - Alan L Parker
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
45
|
Ruf B, Lauer UM. Assessment of current virotherapeutic application schemes: "hit hard and early" versus "killing softly"? Mol Ther Oncolytics 2015; 2:15018. [PMID: 27119110 PMCID: PMC4782955 DOI: 10.1038/mto.2015.18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/20/2015] [Accepted: 07/20/2015] [Indexed: 12/31/2022] Open
Abstract
Over the past two decades, a considerable amount of oncolytic vector families has entered numerous clinical trials. However, to this date, the field has not yet been able to come to a common understanding regarding the best possible ways to administer oncolytic viruses to cancer patients. This is mainly due to the fact that so far clinical trials being designed for head-to-head comparisons (such as using two different virotherapeutics originating from two distinct virus families being applied via identical routes in the same types of cancer) are still missing. Hence, there is no consent (i) on the best route of virotherapeutics administration (e.g., systemic versus intratumoral), (ii) on the virus dosages to be applied, (iii) on dosing intervals, and (iv) on the numbers of repetitive courses of virus administration. As the detailed comparison of clinical virotherapy trial regimens is time-consuming and complex, we here present an overview of current state-of-the-art virotherapeutic application schemes. Notably, our comprehensive assessment culminates in raising two rough classifications of virotherapeutic strategies, i.e., "hit hard and early" versus "killing softly". In order to find out which one of these two gross alternatives might be most successful for each and every tumor entity, we here suggest the implementation of phase 1/2 studies, which primarily aim at a repetitive sampling and analysis of tumor samples in cancer patients treated with oncolytic viruses reading out (i) virus-specific, (ii) tumor-specific as well as (iii) immunotherapeutic parameters. On this basis, a rational design of significantly improved virotherapeutic application schemes should be possible in the future.
Collapse
Affiliation(s)
- Benjamin Ruf
- Department of Internal Medicine I, University Hospital Tuebingen, Tuebingen, Germany
| | - Ulrich M Lauer
- Department of Internal Medicine I, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
46
|
Ramírez M, García-Castro J, Melen GJ, González-Murillo Á, Franco-Luzón L. Patient-derived mesenchymal stem cells as delivery vehicles for oncolytic virotherapy: novel state-of-the-art technology. Oncolytic Virother 2015; 4:149-55. [PMID: 27512678 PMCID: PMC4918392 DOI: 10.2147/ov.s66010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Oncolytic virotherapy is gaining interest in the clinic as a new weapon against cancer. In vivo administration of oncolytic viruses showed important limitations that decrease their effectiveness very significantly: the antiviral immune response causes the elimination of the therapeutic effect, and the poor natural ability of oncolytic viruses to infect micrometastatic lesions significantly minimizes the effective dose of virus. This review will focus on updating the technical and scientific foundations of one of the strategies developed to overcome these limitations, ie, using cells as vehicles for oncolytic viruses. Among many candidates, a special type of adult stem cell, mesenchymal stem cells (MSCs), have already been used in the clinic as cell vehicles for oncolytic viruses, partly due to the fact that these cells are actively being evaluated for other indications. MSC carrier cells are used as Trojan horses loaded with oncoviruses, are administered systemically, and release their cargos at the right places. MSCs are equipped with an array of molecules involved in cell arrest in the capillaries (integrins and selectins), migration toward specific parenchymal locations within tissues (chemokine receptors), and invasion and degradation of the extracellular matrix (proteases). In addition to anatomical targeting capacity, MSCs have a well-recognized role in modulating immune responses by affecting cells of the innate (antigen-presenting cells, natural killer cells) and adaptive immune system (effector and regulatory lymphocytes). Therefore, carrier MSCs may also modulate the immune responses taking place after therapy, ie, the antiviral and the antitumor immune responses.
Collapse
Affiliation(s)
- Manuel Ramírez
- Oncohematología, Hospital Universitario Niño Jesús, Madrid, Spain
| | | | - Gustavo J Melen
- Oncohematología, Hospital Universitario Niño Jesús, Madrid, Spain
| | | | | |
Collapse
|
47
|
Rodríguez-García A, Giménez-Alejandre M, Rojas JJ, Moreno R, Bazan-Peregrino M, Cascalló M, Alemany R. Safety and efficacy of VCN-01, an oncolytic adenovirus combining fiber HSG-binding domain replacement with RGD and hyaluronidase expression. Clin Cancer Res 2014; 21:1406-18. [PMID: 25391696 DOI: 10.1158/1078-0432.ccr-14-2213] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor targeting upon intravenous administration and subsequent intratumoral virus dissemination are key features to improve oncolytic adenovirus therapy. VCN-01 is a novel oncolytic adenovirus that combines selective replication conditional to pRB pathway deregulation, replacement of the heparan sulfate glycosaminoglycan putative-binding site KKTK of the fiber shaft with an integrin-binding motif RGDK for tumor targeting, and expression of hyaluronidase to degrade the extracellular matrix. In this study, we evaluate the safety and efficacy profile of this novel oncolytic adenovirus. EXPERIMENTAL DESIGN VCN-01 replication and potency were assessed in a panel of tumor cell lines. VCN-01 tumor-selective replication was evaluated in human fibroblasts and pancreatic islets. Preclinical toxicity, biodistribution, and efficacy studies were conducted in mice and Syrian hamsters. RESULTS Toxicity and biodistribution preclinical studies support the selectivity and safety of VCN-01. Antitumor activity after intravenous or intratumoral administration of the virus was observed in all tumor models tested, including melanoma and pancreatic adenocarcinoma, both in immunodeficient mice and immunocompetent hamsters. CONCLUSIONS Oncolytic adenovirus VCN-01 characterized by the expression of hyaluronidase and the RGD shaft retargeting ligand shows an efficacy-toxicity prolife in mice and hamsters by intravenous and intratumoral administration that warrants clinical testing.
Collapse
Affiliation(s)
- Alba Rodríguez-García
- Translational Research Laboratory, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Juan J Rojas
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rafael Moreno
- Translational Research Laboratory, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Manel Cascalló
- VCN Biosciences, Sant Cugat del Vallès, Barcelona, Spain
| | - Ramon Alemany
- Translational Research Laboratory, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
48
|
Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Cremer I, Erbs P, Limacher JM, Preville X, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch:: Oncolytic viruses for cancer therapy. Oncoimmunology 2014; 3:e28694. [PMID: 25097804 PMCID: PMC4091053 DOI: 10.4161/onci.28694] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 12/11/2022] Open
Abstract
Oncolytic viruses are natural or genetically modified viral species that selectively infect and kill neoplastic cells. Such an innate or exogenously conferred specificity has generated considerable interest around the possibility to employ oncolytic viruses as highly targeted agents that would mediate cancer cell-autonomous anticancer effects. Accumulating evidence, however, suggests that the therapeutic potential of oncolytic virotherapy is not a simple consequence of the cytopathic effect, but strongly relies on the induction of an endogenous immune response against transformed cells. In line with this notion, superior anticancer effects are being observed when oncolytic viruses are engineered to express (or co-administered with) immunostimulatory molecules. Although multiple studies have shown that oncolytic viruses are well tolerated by cancer patients, the full-blown therapeutic potential of oncolytic virotherapy, especially when implemented in the absence of immunostimulatory interventions, remains unclear. Here, we cover the latest advances in this active area of translational investigation, summarizing high-impact studies that have been published during the last 12 months and discussing clinical trials that have been initiated in the same period to assess the therapeutic potential of oncolytic virotherapy in oncological indications.
Collapse
Affiliation(s)
- Jonathan Pol
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Norma Bloy
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Florine Obrist
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | | | - Jérôme Galon
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers; Paris, France
| | - Isabelle Cremer
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | | | | | | | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
| |
Collapse
|
49
|
Bauzon M, Hermiston T. Armed therapeutic viruses - a disruptive therapy on the horizon of cancer immunotherapy. Front Immunol 2014; 5:74. [PMID: 24605114 PMCID: PMC3932422 DOI: 10.3389/fimmu.2014.00074] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/11/2014] [Indexed: 12/17/2022] Open
Abstract
For the past 150 years cancer immunotherapy has been largely a theoretical hope that recently has begun to show potential as a highly impactful treatment for various cancers. In particular, the identification and targeting of immune checkpoints have given rise to exciting data suggesting that this strategy has the potential to activate sustained antitumor immunity. It is likely that this approach, like other anti-cancer strategies before it, will benefit from co-administration with an additional therapeutic and that it is this combination therapy that may generate the greatest clinical outcome for the patient. In this regard, oncolytic viruses are a therapeutic moiety that is well suited to deliver and augment these immune-modulating therapies in a highly targeted and economically advantageous way over current treatment. In this review, we discuss the blockade of immune checkpoints, how oncolytic viruses complement and extend these therapies, and speculate on how this combination will uniquely impact the future of cancer immunotherapy.
Collapse
Affiliation(s)
- Maxine Bauzon
- Bayer HealthCare, US Innovation Center, Biologics Research , San Francisco, CA , USA
| | - Terry Hermiston
- Bayer HealthCare, US Innovation Center, Biologics Research , San Francisco, CA , USA
| |
Collapse
|
50
|
Uchino J, Curiel DT, Ugai H. Species D human adenovirus type 9 exhibits better virus-spread ability for antitumor efficacy among alternative serotypes. PLoS One 2014; 9:e87342. [PMID: 24503714 PMCID: PMC3913592 DOI: 10.1371/journal.pone.0087342] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/20/2013] [Indexed: 11/19/2022] Open
Abstract
Species C human adenovirus serotype 5 (HAdV-C5) is widely used as a vector for cancer gene therapy, because it efficiently transduces target cells. A variety of HAdV-C5 vectors have been developed and tested in vitro and in vivo for cancer gene therapy. While clinical trials with HAdV-C5 vectors resulted in effective responses in many cancer patients, administration of HAdV-C5 vectors to solid tumors showed responses in a limited area. A biological barrier in tumor mass is considered to hinder viral spread of HAdV-C5 vectors from infected cells. Therefore, efficient virus-spread from an infected tumor cell to surrounding tumor cells is required for successful cancer gene therapy. In this study, we compared HAdV-C5 to sixteen other HAdV serotypes selected from species A to G for virus-spread ability in vitro. HAdV-D9 showed better virus-spread ability than other serotypes, and its viral progeny were efficiently released from infected cells during viral replication. Although the HAdV-D9 fiber protein contains a binding site for coxsackie B virus and adenovirus receptor (CAR), HAdV-D9 showed expanded tropism for infection due to human CAR (hCAR)-independent attachment to target cells. HAdV-D9 infection effectively killed hCAR-negative cancer cells as well as hCAR-positive cancer cells. These results suggest that HADV-D9, with its better virus-spread ability, could have improved therapeutic efficacy in solid tumors compared to HAdV-C5.
Collapse
Affiliation(s)
- Junji Uchino
- Cancer Biology Division, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - David T. Curiel
- Cancer Biology Division, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Biologic Therapeutics Center, Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Hideyo Ugai
- Cancer Biology Division, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|