1
|
Hua X, Xuan S, Tang Y, You S, Zhao S, Qiu Y, Li Y, Li Y, Su Y, Qu P. Progression of oncolytic virus in liver cancer treatment. Front Oncol 2024; 14:1446085. [PMID: 39391253 PMCID: PMC11464341 DOI: 10.3389/fonc.2024.1446085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
The liver plays a crucrial role in detoxification, metabolism, and nutrient storage. Because liver cancer ranks among the top three leading causes of death globally, there is an urgent need for developing treatment strategies for liver cancer. Although traditional approaches such as radiation, chemotherapy, surgical removal, and transplantation are widely practiced, the number of patients with liver cancer continues to increase rapidly each year. Some novel therapeutics for liver cancer have been studied for many years. In the past decade, oncolytic therapy has emerged, in which viruses selectively infect and destroy cancer cells while sparing normal cells. However, oncolytic virotherapy for liver cancer remains relatively obscure due to the aggressive nature of the disease and the limited effectiveness of treatment. To keep pace with the latest developments in oncolytic tumor therapy for liver cancer, this review summarizes basic science studies and clinical trials conducted within 5 years, focusing on the efficacy and safety profiles of the five most commonly used oncolytic viruses: herpes simplex virus, adenovirus, influenza virus, vaccinia virus, and coxsackievirus.
Collapse
Affiliation(s)
- Xuesi Hua
- School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Siyu Xuan
- Department of Histology and Embryology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yangyang Tang
- Department of Histology and Embryology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shilin You
- Department of Pharmacy, Changchun University of Traditional Chinese Medicine Innovation Practice Center, Changchun, Jilin, China
| | - Shang Zhao
- Department of Histology and Embryology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ye Qiu
- Department of Pharmacy, Changchun University of Traditional Chinese Medicine Innovation Practice Center, Changchun, Jilin, China
| | - Yinqing Li
- Department of Pharmacy, Changchun University of Traditional Chinese Medicine Innovation Practice Center, Changchun, Jilin, China
| | - Yongqing Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing, China
| | - Yanping Su
- Department of Histology and Embryology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Peng Qu
- Department of Histology and Embryology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Pharmacy, Changchun University of Traditional Chinese Medicine Innovation Practice Center, Changchun, Jilin, China
- Department of Pharmacy, Zhejiang University of Technology Fuyang Yinhu Institute of Innovation and Entrepreneurship, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Asif M, Iqbal W, Fakhar-e-Alam M, Hussain Z, Saadullah M, Hassan M, Rehman J, Dahlous KA, Al-Qahtani NH. Synthesis and Characterization of Chemically and Green-Synthesized Silver Oxide Particles for Evaluation of Antiviral and Anticancer Activity. Pharmaceuticals (Basel) 2024; 17:908. [PMID: 39065758 PMCID: PMC11280290 DOI: 10.3390/ph17070908] [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: 05/04/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Silver oxide (Ag2O) particles are wonderful candidates due to their unique properties, and their use in a wide range of research, industrial and biomedical applications is rapidly increasing. This makes it fundamental to develop simple, environmentally friendly methods with possible scaling. Herein, sodium borohydride and Datura innoxia leaf extract were applied as chemical and biological stabilizing and reducing agents to develop Ag2O particles. The primary aim was to evaluate the anticancer and antiviral activity of Ag2O particles prepared via two methods. XRD, UV-visible and SEM analyses were used to examine the crystallite structure, optical properties and morphology, respectively. The resulting green-synthesized Ag2O particles exhibited small size, spherically agglomerated shape, and high anticancer and antiviral activities compared to chemically synthesized Ag2O particles. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium-bromide) assay of green-synthesized Ag2O particles showed high anticancer activity against MCF-7 cells with IC50 = 17.908 µg/mL compared to chemically synthesized Ag2O particles with IC50 = 23.856 µg/mL. The antiviral activity of green-synthesized Ag2O particles and chemically synthesized Ag2O particles was also evaluated by a plaque-forming assay, and green-synthesized Ag2O particles showed higher antiviral ability with IC50 = 0.618 µg/mL as compared to chemically synthesized Ag2O particles with IC50 = 6.129 µg/mL. We propose the use of green-synthesized Ag2O particles in cancer treatment and drug delivery.
Collapse
Affiliation(s)
- Muhammad Asif
- Department of Physics, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Wajeeha Iqbal
- Department of Physics, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Fakhar-e-Alam
- Department of Physics, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Zahid Hussain
- Department of Physics, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Malik Saadullah
- Department of Pharmaceutical Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Mudassir Hassan
- Department of Zoology, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Javed Rehman
- State Key Laboratory of Metastable Materials Science and Technology, School of Materials Science and Engineering, Yanshan University, Qinhuangdao 066004, China
- MEU Research Unit, Middle East University, Amman 11831, Jordan
| | - Kholood A. Dahlous
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | | |
Collapse
|
3
|
Joo V, Abdelhamid K, Noto A, Latifyan S, Martina F, Daoudlarian D, De Micheli R, Pruijm M, Peters S, Hullin R, Gaide O, Pantaleo G, Obeid M. Primary prophylaxis with mTOR inhibitor enhances T cell effector function and prevents heart transplant rejection during talimogene laherparepvec therapy of squamous cell carcinoma. Nat Commun 2024; 15:3664. [PMID: 38693123 PMCID: PMC11063183 DOI: 10.1038/s41467-024-47965-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 04/15/2024] [Indexed: 05/03/2024] Open
Abstract
The application of mammalian target of rapamycin inhibition (mTORi) as primary prophylactic therapy to optimize T cell effector function while preserving allograft tolerance remains challenging. Here, we present a comprehensive two-step therapeutic approach in a male patient with metastatic cutaneous squamous cell carcinoma and heart transplantation followed with concomitant longitudinal analysis of systemic immunologic changes. In the first step, calcineurin inhibitor/ mycophenolic acid is replaced by the mTORi everolimus to achieve an improved effector T cell status with increased cytotoxic activity (perforin, granzyme), enhanced proliferation (Ki67) and upregulated activation markers (CD38, CD69). In the second step, talimogene laherparepvec (T-VEC) injection further enhances effector function by switching CD4 and CD8 cells from central memory to effector memory profiles, enhancing Th1 responses, and boosting cytotoxic and proliferative activities. In addition, cytokine release (IL-6, IL-18, sCD25, CCL-2, CCL-4) is enhanced and the frequency of circulating regulatory T cells is increased. Notably, no histologic signs of allograft rejection are observed in consecutive end-myocardial biopsies. These findings provide valuable insights into the dynamics of T cell activation and differentiation and suggest that timely initiation of mTORi-based primary prophylaxis may provide a dual benefit of revitalizing T cell function while maintaining allograft tolerance.
Collapse
Affiliation(s)
- Victor Joo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Karim Abdelhamid
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Alessandra Noto
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Sofiya Latifyan
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Federica Martina
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Douglas Daoudlarian
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Rita De Micheli
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Menno Pruijm
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Nephrology Division, Rue du Bugnon 17, CH-1011, Lausanne, Switzerland
| | - Solange Peters
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Roger Hullin
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Cardiology, Cardiovascular Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Olivier Gaide
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Dermatology Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Michel Obeid
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland.
| |
Collapse
|
4
|
Li Z, Chen Y, Li L, Xue M, Feng L. Different Infectivity of Swine Enteric Coronaviruses in Cells of Various Species. Pathogens 2024; 13:174. [PMID: 38392912 PMCID: PMC10891669 DOI: 10.3390/pathogens13020174] [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: 12/19/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Swine enteric coronaviruses (SECoVs), including porcine deltacoronavirus (PDCoV), transmissible gastroenteritis virus (TGEV), porcine epidemic diarrhea virus (PEDV), and swine acute diarrhea syndrome coronavirus (SADS-CoV), have caused high mortality in piglets and, therefore, pose serious threats to the pork industry. Coronaviruses exhibit a trend of interspecies transmission, and understanding the host range of SECoVs is crucial for improving our ability to predict and control future epidemics. Here, the replication of PDCoV, TGEV, and PEDV in cells from different host species was compared by measuring viral genomic RNA transcription and protein synthesis. We demonstrated that PDCoV had a higher efficiency in infecting human lung adenocarcinoma cells (A549), Madin-Darby bovine kidney cells (MDBK), Madin-Darby canine kidney cells (MDCK), and chicken embryonic fibroblast cells (DF-1) than PEDV and TGEV. Moreover, trypsin can enhance the infectivity of PDCoV to MDCK cells that are nonsusceptible to TGEV. Additionally, structural analyses of the receptor ectodomain indicate that PDCoV S1 engages Aminopeptidase N (APN) via domain II, which is highly conserved among animal species of different vertebrates. Our findings provide a basis for understanding the interspecies transmission potential of these three porcine coronaviruses.
Collapse
Affiliation(s)
| | | | | | - Mei Xue
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Li Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| |
Collapse
|
5
|
Synowiec A, Dąbrowska A, Pachota M, Baouche M, Owczarek K, Niżański W, Pyrc K. Feline herpesvirus 1 (FHV-1) enters the cell by receptor-mediated endocytosis. J Virol 2023; 97:e0068123. [PMID: 37493545 PMCID: PMC10506464 DOI: 10.1128/jvi.00681-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/13/2023] [Indexed: 07/27/2023] Open
Abstract
Feline herpesvirus type 1 (FHV-1) is an enveloped dsDNA virus belonging to the Herpesviridae family and is considered one of the two primary viral etiological factors of feline upper respiratory tract disease. In this study, we investigated the entry of FHV-1 into host cells using two models: the AK-D cell line and primary feline skin fibroblasts (FSFs). We employed confocal microscopy, siRNA silencing, and selective inhibitors of various entry pathways. Our observations revealed that the virus enters cells via pH and dynamin-dependent endocytosis, as the infection was significantly inhibited by NH4Cl, bafilomycin A1, dynasore, and mitmab. Additionally, genistein, nystatin, and filipin treatments, siRNA knock-down of caveolin-1, as well as FHV-1 and caveolin-1 colocalization suggest the involvement of caveolin-mediated endocytosis during the entry process. siRNA knock-down of clathrin heavy chain and analysis of virus particle colocalization with clathrin indicated that clathrin-mediated endocytosis also takes part in the primary cells. This is the first study to systematically examine FHV-1 entry into host cells, and for the first time, we describe FHV-1 replication in AK-D and FSFs. IMPORTANCE Feline herpesvirus 1 (FHV-1) is one of the most prevalent viruses in cats, causing feline viral rhinotracheitis, which is responsible for over half of viral upper respiratory diseases in cats and can lead to ocular lesions resulting in loss of sight. Although the available vaccine reduces the severity of the disease, it does not prevent infection or limit virus shedding. Despite the clinical relevance, the entry mechanisms of FHV-1 have not been thoroughly studied. Considering the limitations of commonly used models based on immortalized cells, we sought to verify our findings using primary feline skin fibroblasts, the natural target for infection in cats.
Collapse
Affiliation(s)
- Aleksandra Synowiec
- ViroGenetics - BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
- Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Agnieszka Dąbrowska
- ViroGenetics - BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Pachota
- ViroGenetics - BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Meriem Baouche
- Department of Reproduction and Clinic of Farm Animals, University of Environmental Science, Wrocław, Poland
| | - Katarzyna Owczarek
- ViroGenetics - BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Wojciech Niżański
- Department of Reproduction and Clinic of Farm Animals, University of Environmental Science, Wrocław, Poland
| | - Krzysztof Pyrc
- ViroGenetics - BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
6
|
Lin D, Shen Y, Liang T. Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther 2023; 8:156. [PMID: 37041165 PMCID: PMC10090134 DOI: 10.1038/s41392-023-01407-6] [Citation(s) in RCA: 142] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 04/13/2023] Open
Abstract
Oncolytic viruses (OVs) have attracted growing awareness in the twenty-first century, as they are generally considered to have direct oncolysis and cancer immune effects. With the progress in genetic engineering technology, OVs have been adopted as versatile platforms for developing novel antitumor strategies, used alone or in combination with other therapies. Recent studies have yielded eye-catching results that delineate the promising clinical outcomes that OVs would bring about in the future. In this review, we summarized the basic principles of OVs in terms of their classifications, as well as the recent advances in OV-modification strategies based on their characteristics, biofunctions, and cancer hallmarks. Candidate OVs are expected to be designed as "qualified soldiers" first by improving target fidelity and safety, and then equipped with "cold weapons" for a proper cytocidal effect, "hot weapons" capable of activating cancer immunotherapy, or "auxiliary weapons" by harnessing tactics such as anti-angiogenesis, reversed metabolic reprogramming and decomposing extracellular matrix around tumors. Combinations with other cancer therapeutic agents have also been elaborated to show encouraging antitumor effects. Robust results from clinical trials using OV as a treatment congruously suggested its significance in future application directions and challenges in developing OVs as novel weapons for tactical decisions in cancer treatment.
Collapse
Affiliation(s)
- Danni Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yinan Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Zhou H, Astore C, Skolnick J. PHEVIR: an artificial intelligence algorithm that predicts the molecular role of pathogens in complex human diseases. Sci Rep 2022; 12:20889. [PMID: 36463386 PMCID: PMC9719543 DOI: 10.1038/s41598-022-25412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
Infectious diseases are known to cause a wide variety of post-infection complications. However, it's been challenging to identify which diseases are most associated with a given pathogen infection. Using the recently developed LeMeDISCO approach that predicts comorbid diseases associated with a given set of putative mode of action (MOA) proteins and pathogen-human protein interactomes, we developed PHEVIR, an algorithm which predicts the corresponding human disease comorbidities of 312 viruses and 57 bacteria. These predictions provide an understanding of the molecular bases of complications and means of identifying appropriate drug targets to treat them. As an illustration of its power, PHEVIR is applied to identify putative driver pathogens and corresponding human MOA proteins for Type 2 diabetes, atherosclerosis, Alzheimer's disease, and inflammatory bowel disease. Additionally, we explore the origins of the oncogenicity/oncolyticity of certain pathogens and the relationship between heart disease and influenza. The full PHEVIR database is available at https://sites.gatech.edu/cssb/phevir/ .
Collapse
Affiliation(s)
- Hongyi Zhou
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA, 30332, USA
| | - Courtney Astore
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA, 30332, USA
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA, 30332, USA.
| |
Collapse
|
8
|
Schwarze JK, Tijtgat J, Awada G, Cras L, Vasaturo A, Bagnall C, Forsyth R, Dufait I, Tuyaerts S, Van Riet I, Neyns B. Intratumoral administration of CD1c (BDCA-1) + and CD141 (BDCA-3) + myeloid dendritic cells in combination with talimogene laherparepvec in immune checkpoint blockade refractory advanced melanoma patients: a phase I clinical trial. J Immunother Cancer 2022; 10:jitc-2022-005141. [PMID: 36113895 PMCID: PMC9486335 DOI: 10.1136/jitc-2022-005141] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Intratumoral (IT) myeloid dendritic cells (myDCs) play a pivotal role in initiating antitumor immune responses and relicensing of anti-tumor cytotoxic T lymphocytes within the tumor microenvironment. Talimogene laherparepvec (T-VEC) induces immunogenic cell death, thereby providing maturation signals and enhancing the release of tumor antigens that can be captured and processed by CD1c (BDCA-1)+ / CD141 (BDCA-3)+ myDCs, in order to reinvigorate the cancer-immunity cycle. METHODS In this phase I trial, patients with advanced melanoma who failed standard therapy were eligible for IT injections of ≥1 non-visceral metastases with T-VEC on day 1 followed by IT injection of CD1c (BDCA-1)+ myDCs +/- CD141 (BDCA-3)+ myDCs on day 2. T-VEC injections were repeated on day 21 and every 14 days thereafter. The number of IT administered CD1c (BDCA-1)+ myDCs was escalated from 0.5×106, to 1×106, to a maximum of 10×106 cells in three sequential cohorts. In cohort 4, all isolated CD1c (BDCA-1)+ / CD141 (BDCA-3)+ myDCs were used for IT injection. Primary objectives were safety and feasibility. Repetitive biopsies of treated lesions were performed. RESULTS In total, 13 patients were enrolled (cohort 1 n=2; cohort 2 n=2; cohort 3 n=3; cohort 4 n=6). Patients received a median of 6 (range 3-8) T-VEC injections. The treatment was safe with most frequent adverse events being fatigue (n=11 (85%)), fever (n=8 (62%)), and chills/influenza-like symptoms (n=6 (46%)). Nine (69%) and four patients (31%), respectively, experienced pain or redness at the injection-site. Clinical responses were documented in injected and non-injected lesions. Two patients (cohort 3) who previously progressed on anti-PD-1 therapy (and one patient also on anti-CTLA-4 therapy) developed a durable, pathologically confirmed complete response that is ongoing at 33 and 35 months following initiation of study treatment. One additional patient treated (cohort 4) had an unconfirmed partial response as best response; two additional patients had a mixed response (with durable complete responses of some injected and non-injected lesions). On-treatment biopsies revealed a strong infiltration by inflammatory cells in regressing lesions. CONCLUSIONS IT coinjection of autologous CD1c (BDCA-1)+ +/- CD141 (BDCA-3)+ myDCs with T-VEC is feasible, tolerable and resulted in encouraging early signs of antitumor activity in immune checkpoint inhibitor-refractory melanoma patients. TRIAL REGISTRATION NUMBER NCT03747744.
Collapse
Affiliation(s)
- Julia Katharina Schwarze
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Jens Tijtgat
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Gil Awada
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Louise Cras
- Department of Anatomopathology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | | | | | - Ramses Forsyth
- Department of Anatomopathology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Inès Dufait
- Department of Radiotherapy, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Sandra Tuyaerts
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Ivan Van Riet
- Stem Cell Laboratory, Department of Hematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Bart Neyns
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| |
Collapse
|
9
|
Wedge ME, Jennings VA, Crupi MJF, Poutou J, Jamieson T, Pelin A, Pugliese G, de Souza CT, Petryk J, Laight BJ, Boileau M, Taha Z, Alluqmani N, McKay HE, Pikor L, Khan ST, Azad T, Rezaei R, Austin B, He X, Mansfield D, Rose E, Brown EEF, Crawford N, Alkayyal A, Surendran A, Singaravelu R, Roy DG, Migneco G, McSweeney B, Cottee ML, Jacobus EJ, Keller BA, Yamaguchi TN, Boutros PC, Geoffrion M, Rayner KJ, Chatterjee A, Auer RC, Diallo JS, Gibbings D, tenOever BR, Melcher A, Bell JC, Ilkow CS. Virally programmed extracellular vesicles sensitize cancer cells to oncolytic virus and small molecule therapy. Nat Commun 2022; 13:1898. [PMID: 35393414 PMCID: PMC8990073 DOI: 10.1038/s41467-022-29526-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
Recent advances in cancer therapeutics clearly demonstrate the need for innovative multiplex therapies that attack the tumour on multiple fronts. Oncolytic or “cancer-killing” viruses (OVs) represent up-and-coming multi-mechanistic immunotherapeutic drugs for the treatment of cancer. In this study, we perform an in-vitro screen based on virus-encoded artificial microRNAs (amiRNAs) and find that a unique amiRNA, herein termed amiR-4, confers a replicative advantage to the VSVΔ51 OV platform. Target validation of amiR-4 reveals ARID1A, a protein involved in chromatin remodelling, as an important player in resistance to OV replication. Virus-directed targeting of ARID1A coupled with small-molecule inhibition of the methyltransferase EZH2 leads to the synthetic lethal killing of both infected and uninfected tumour cells. The bystander killing of uninfected cells is mediated by intercellular transfer of extracellular vesicles carrying amiR-4 cargo. Altogether, our findings establish that OVs can serve as replicating vehicles for amiRNA therapeutics with the potential for combination with small molecule and immune checkpoint inhibitor therapy. RNA-based viruses can be engineered to express artificial microRNAs (amiRNAs). Here, the authors identify a candidate amiRNA that confers a replicative advantage to oncolytic viruses, enhancing their anticancer potency, and show that intercellular transfer of extracellular vesicles carrying the amiRNA promotes bystander killing of uninfected cancer cells.
Collapse
Affiliation(s)
- Marie-Eve Wedge
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Victoria A Jennings
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Institute of Cancer Research, London, UK.,Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Mathieu J F Crupi
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Joanna Poutou
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Taylor Jamieson
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Adrian Pelin
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Giuseppe Pugliese
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | - Julia Petryk
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Brian J Laight
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Meaghan Boileau
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Zaid Taha
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Nouf Alluqmani
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Hayley E McKay
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Larissa Pikor
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Sarwat Tahsin Khan
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Taha Azad
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Reza Rezaei
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Bradley Austin
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Xiaohong He
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | - Elaine Rose
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Emily E F Brown
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Natalie Crawford
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Almohanad Alkayyal
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Abera Surendran
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ragunath Singaravelu
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Dominic G Roy
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Gemma Migneco
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Benjamin McSweeney
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Mary Lynn Cottee
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Egon J Jacobus
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Oncology, University of Oxford, Oxford, UK
| | - Brian A Keller
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Takafumi N Yamaguchi
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Paul C Boutros
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Urology, University of California, Los Angeles, Los Angeles, CA, USA.,Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Katey J Rayner
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Avijit Chatterjee
- The Ottawa Hospital, Division of Gastroenterology, Ottawa, Ontario, Canada
| | - Rebecca C Auer
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Jean-Simon Diallo
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Derrick Gibbings
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Benjamin R tenOever
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - John C Bell
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Carolina S Ilkow
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada. .,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
10
|
Ohadi L, Hosseinzadeh F, Dadkhahfar S, Nasiri S. Oncolytic effect of SARS-CoV-2 in a patient with mycosis fungoides: A case report. Clin Case Rep 2022; 10:e05682. [PMID: 35387287 PMCID: PMC8978791 DOI: 10.1002/ccr3.5682] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/06/2022] [Accepted: 03/15/2022] [Indexed: 12/19/2022] Open
Abstract
The most common variant of cutaneous T-cell lymphomas (CTCL) is mycosis fungoides (MF). Patients with MF often experience a chronic course of disease. The spontaneous regression (SR) of MF is rare, and the factors that predict SR have not been recognized yet. Here, we are reporting a case of persistent MF who had prominent remission after COVID-19. This case report supports the possible antineoplastic effect of SARS-CoV-2. Understanding the underlying etiology of such effect can result in development of new target therapies for MF.
Collapse
Affiliation(s)
- Laya Ohadi
- Shahid Beheshti University of Medical SciencesTehranIran
| | | | - Sahar Dadkhahfar
- Skin Research CenterShahid Beheshti Universiry of Medical SciencesTehranIran
| | - Soheila Nasiri
- Skin Research CenterShahid Beheshti Universiry of Medical SciencesTehranIran
| |
Collapse
|
11
|
Bezeljak U. Cancer gene therapy goes viral: viral vector platforms come of age. Radiol Oncol 2022; 56:1-13. [PMID: 35148469 PMCID: PMC8884858 DOI: 10.2478/raon-2022-0002] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/04/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Since the advent of viral vector gene therapy in 1990s, cancer treatment with viral vectors promised to revolutionize the field of oncology. Notably, viral vectors offer a unique combination of efficient gene delivery and engagement of the immune system for anti-tumour response. Despite the early potential, viral vector-based cancer treatments are only recently making a big impact, most prominently as gene delivery devices in approved CAR-T cell therapies, cancer vaccines and targeted oncolytic therapeutics. To reach this broad spectrum of applications, a number of challenges have been overcome - from our understanding of cancer biology to vector design, manufacture and engineering. Here, we take an overview of viral vector usage in cancer therapy and discuss the latest advancements. We also consider production platforms that enable mainstream adoption of viral vectors for cancer gene therapy. CONCLUSIONS Viral vectors offer numerous opportunities in cancer therapy. Recent advances in vector production platforms open new avenues in safe and efficient viral therapeutic strategies, streamlining the transition from lab bench to bedside. As viral vectors come of age, they could become a standard tool in the cancer treatment arsenal.
Collapse
|
12
|
Andretta E, Costa C, Longobardi C, Damiano S, Giordano A, Pagnini F, Montagnaro S, Quintiliani M, Lauritano C, Ciarcia R. Potential Approaches Versus Approved or Developing Chronic Myeloid Leukemia Therapy. Front Oncol 2022; 11:801779. [PMID: 34993151 PMCID: PMC8724906 DOI: 10.3389/fonc.2021.801779] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/12/2021] [Indexed: 12/22/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of patients with chronic myeloid leukemia (CML). However, continued use of these inhibitors has contributed to the increase in clinical resistance and the persistence of resistant leukemic stem cells (LSCs). So, there is an urgent need to introduce additional targeted and selective therapies to eradicate quiescent LSCs, and to avoid the relapse and disease progression. Here, we focused on emerging BCR-ABL targeted and non-BCR-ABL targeted drugs employed in clinical trials and on alternative CML treatments, including antioxidants, oncolytic virus, engineered exosomes, and natural products obtained from marine organisms that could pave the way for new therapeutic approaches for CML patients.
Collapse
Affiliation(s)
- Emanuela Andretta
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Caterina Costa
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Consiglia Longobardi
- Department of Mental, Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Largo Madonna delle Grazie, Naples, Italy
| | - Sara Damiano
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, Siena, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Francesco Pagnini
- Unit of Radiology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Serena Montagnaro
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | | | - Chiara Lauritano
- Marine Biotechnology Department, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
13
|
The Oncolytic Caprine Herpesvirus 1 (CpHV-1) Induces Apoptosis and Synergizes with Cisplatin in Mesothelioma Cell Lines: A New Potential Virotherapy Approach. Viruses 2021; 13:v13122458. [PMID: 34960727 PMCID: PMC8703924 DOI: 10.3390/v13122458] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/12/2021] [Accepted: 12/03/2021] [Indexed: 12/17/2022] Open
Abstract
Malignant mesothelioma (MM) is an aggressive asbestos-related cancer, against which no curative modalities exist. Oncolytic virotherapy is a promising therapeutic approach, for which MM is an ideal candidate; indeed, the pleural location provides direct access for the intra-tumoral injection of oncolytic viruses (OVs). Some non-human OVs offer advantages over human OVs, including the non-pathogenicity in humans and the absence of pre-existing immunity. We previously showed that caprine herpesvirus 1 (CpHV-1), a non-pathogenic virus for humans, can kill different human cancer cell lines. Here, we assessed CpHV-1 effects on MM (NCI-H28, MSTO, NCI-H2052) and non-tumor mesothelial (MET-5A) cells. We found that CpHV-1 reduced cell viability and clonogenic potential in all MM cell lines without affecting non-tumor cells, in which, indeed, we did not detect intracellular viral DNA after treatment. In particular, CpHV-1 induced MM cell apoptosis and accumulation in G0/G1 or S cell cycle phases. Moreover, CpHV-1 strongly synergized with cisplatin, the drug currently used in MM chemotherapy, and this agent combination did not affect normal mesothelial cells. Although further studies are required to elucidate the mechanisms underlying the selective CpHV-1 action on MM cells, our data suggest that the CpHV-1-cisplatin combination could be a feasible strategy against MM.
Collapse
|
14
|
Kai M, Marx AN, Liu DD, Shen Y, Gao H, Reuben JM, Whitman G, Krishnamurthy S, Ross MI, Litton JK, Lim B, Ibrahim N, Kogawa T, Ueno NT. A phase II study of talimogene laherparepvec for patients with inoperable locoregional recurrence of breast cancer. Sci Rep 2021; 11:22242. [PMID: 34782633 PMCID: PMC8593093 DOI: 10.1038/s41598-021-01473-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 10/05/2021] [Indexed: 01/03/2023] Open
Abstract
Talimogene laherparepvec (T-VEC) is an immunotherapy that generates local tumor lysis and systemic antitumor immune response. We studied the efficacy of intratumoral administration of T-VEC as monotherapy for inoperable locoregional recurrence of breast cancer. T-VEC was injected intratumorally at 106 PFU/mL on day 1 (cycle 1), 108 PFU/mL on day 22 (cycle 2), and 108 PFU/mL every 2 weeks thereafter (cycles ≥ 3). Nine patients were enrolled, 6 with only locoregional disease and 3 with both locoregional and distant disease. No patient completed the planned 10 cycles or achieved complete or partial response. The median number of cycles administered was 4 (range, 3-8). Seven patients withdrew prematurely because of uncontrolled disease progression, 1 withdrew after cycle 3 because of fatigue, and 1 withdrew after cycle 4 for reasons unrelated to study treatment. Median progression-free survival and overall survival were 77 days (95% CI, 63-NA) and 361 days (95% CI, 240-NA). Two patients received 8 cycles with clinically stable disease as the best response. The most common grade 2 or higher adverse event was injection site reaction (n = 7, 78%). Future studies could examine whether combining intratumoral T-VEC with concurrent systemic therapy produces better outcomes.
Collapse
Affiliation(s)
- Megumi Kai
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Angela N Marx
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Diane D Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Yu Shen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Hui Gao
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematopathology Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - James M Reuben
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematopathology Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Gary Whitman
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Savitri Krishnamurthy
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Merrick I Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Oncology/Medicine, Baylor College of Medicine, 7200 Cambridge St., Houston, TX, 77030, USA
| | - Nuhad Ibrahim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Takahiro Kogawa
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA.
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
Gao Y, Wu Y, Huan T, Wang X, Xu J, Xu Q, Yu F, Shi H. The application of oncolytic viruses in cancer therapy. Biotechnol Lett 2021; 43:1945-1954. [PMID: 34448096 DOI: 10.1007/s10529-021-03173-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 08/16/2021] [Indexed: 12/22/2022]
Abstract
Oncolytic therapy is a treatment method used to directly combat tumor cells by modifying the genes of naturally occurring low pathogenic viruses to form "rhizobia" virus. By taking the advantage of abnormal signal pathways in cancer cells, it selectively replicates in tumor cells leading to tumor cell lysis and death. At present, clinical studies widely employ biomolecular technology to transform oncolytic viruses to exert stronger oncolytic effects and reduce their adverse reactions. This review summarizes the current progresses and the molecular mechanism of oncolytic viruses towards tumor treatment and management.
Collapse
Affiliation(s)
- Yang Gao
- School of Life Sciences, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, People's Republic of China
| | - Yan Wu
- School of Life Sciences, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, People's Republic of China
| | - Tian Huan
- School of Life Sciences, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, People's Republic of China
| | - Xiaoyan Wang
- Department of Gastroenterology, The First People's Hospital of Suqian, Suqian, Jiangsu, People's Republic of China
| | - Jun Xu
- Department of Cognitive Neurology, China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing Tian Tan Hospital, Affiliated to Capital Medical University, Beijing, People's Republic of China
| | - Qinggang Xu
- School of Life Sciences, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, People's Republic of China
| | - Feng Yu
- School of Life Sciences, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, People's Republic of China.
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, People's Republic of China.
| |
Collapse
|
16
|
Oncolytic Herpes Simplex Virus-Based Therapies for Cancer. Cells 2021; 10:cells10061541. [PMID: 34207386 PMCID: PMC8235327 DOI: 10.3390/cells10061541] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
With the increased worldwide burden of cancer, including aggressive and resistant cancers, oncolytic virotherapy has emerged as a viable therapeutic option. Oncolytic herpes simplex virus (oHSV) can be genetically engineered to target cancer cells while sparing normal cells. This leads to the direct killing of cancer cells and the activation of the host immunity to recognize and attack the tumor. Different variants of oHSV have been developed to optimize its antitumor effects. In this review, we discuss the development of oHSV, its antitumor mechanism of action and the clinical trials that have employed oHSV variants to treat different types of tumor.
Collapse
|
17
|
Martini V, D'Avanzo F, Maggiora PM, Varughese FM, Sica A, Gennari A. Oncolytic virotherapy: new weapon for breast cancer treatment. Ecancermedicalscience 2020; 14:1149. [PMID: 33574894 PMCID: PMC7864690 DOI: 10.3332/ecancer.2020.1149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
The recent introduction of viruses as a weapon against cancer can be regarded as one of the most intriguing approaches in the context of precision medicine. The role of immune checkpoint inhibitors has been extensively studied in early and advanced cancer stages, with extraordinary results. Although there is a good tolerability profile, especially when compared with conventional chemotherapy, severe immune-related adverse events have emerged as a potential limitation. Moreover, there are still treatment-resistant cases and thus further treatment options need to be implemented. Several in vitro and in vivo studies have been conducted and are ongoing to develop oncolytic viruses (OVs) as a tool to modulate the immune system response. OVs are attenuated viruses that can kill cancer cells after having infected them, producing microenvironment remodelling and antitumour immune response. The potential of oncolytic virotherapy is to contrast the absence of T cell infiltrates, converting ‘cold’ tumours into ‘hot’ ones, thus improving the performance of the immune system. Breast cancer, the second most common cause of cancer-related deaths among women, is considered a ‘cold’ tumour. In this context, oncolytic virotherapy might well be considered as a promising strategy. This review summarises the current status, clinical applications and future development of OVs, focusing on breast cancer treatment.
Collapse
Affiliation(s)
- Veronica Martini
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases - CAAD, Novara 28100, Italy.,https://orcid.org/0000-0002-0887-4082
| | - Francesca D'Avanzo
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy
| | - Paola Maria Maggiora
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy
| | - Feba Maria Varughese
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases - CAAD, Novara 28100, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A Avogadro 28100, Italy.,Department of Inflammation and Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano (MI) 20089, Italy.,https://orcid.org/0000-0002-8342-7442
| | - Alessandra Gennari
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases - CAAD, Novara 28100, Italy.,https://orcid.org/0000-0002-0928-2281
| |
Collapse
|
18
|
Bitter EE, Townsend MH, Erickson R, Allen C, O'Neill KL. Thymidine kinase 1 through the ages: a comprehensive review. Cell Biosci 2020; 10:138. [PMID: 33292474 PMCID: PMC7694900 DOI: 10.1186/s13578-020-00493-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 11/09/2020] [Indexed: 12/23/2022] Open
Abstract
Proliferation markers, such as proliferating cell nuclear antigen (PCNA), Ki-67, and thymidine kinase 1 (TK1), have potential as diagnostic tools and as prognostic factors in assessing cancer treatment and disease progression. TK1 is involved in cellular proliferation through the recovery of the nucleotide thymidine in the DNA salvage pathway. TK1 upregulation has been found to be an early event in cancer development. In addition, serum levels of TK1 have been shown to be tied to cancer stage, so that higher levels of TK1 indicate a more serious prognosis. As a result of these findings and others, TK1 is not only a potentially viable biomarker for cancer recurrence, treatment monitoring, and survival, but is potentially more advantageous than current biomarkers. Compared to other proliferation markers, TK1 levels during S phase more accurately determine the rate of DNA synthesis in actively dividing tumors. Several reviews of TK1 elaborate on various assays that have been developed to measure levels in the serum of cancer patients in clinical settings. In this review, we include a brief history of important TK1 discoveries and findings, a comprehensive overview of TK1 regulation at DNA to protein levels, and recent findings that indicate TK1’s potential role in cancer pathogenesis and its growing potential as a tumor biomarker and therapeutic target.
Collapse
Affiliation(s)
- Eliza E Bitter
- Department of Microbiology and Molecular Biology, Brigham Young University, 701 E University Pkwy, LSB room 4007, Provo, UT, 84602, USA.
| | - Michelle H Townsend
- Department of Microbiology and Molecular Biology, Brigham Young University, 701 E University Pkwy, LSB room 4007, Provo, UT, 84602, USA
| | - Rachel Erickson
- Department of Microbiology and Molecular Biology, Brigham Young University, 701 E University Pkwy, LSB room 4007, Provo, UT, 84602, USA
| | - Carolyn Allen
- Department of Microbiology and Molecular Biology, Brigham Young University, 701 E University Pkwy, LSB room 4007, Provo, UT, 84602, USA
| | - Kim L O'Neill
- Department of Microbiology and Molecular Biology, Brigham Young University, 701 E University Pkwy, LSB room 4007, Provo, UT, 84602, USA.
| |
Collapse
|
19
|
Cunliffe TG, Bates EA, Parker AL. Hitting the Target but Missing the Point: Recent Progress towards Adenovirus-Based Precision Virotherapies. Cancers (Basel) 2020; 12:E3327. [PMID: 33187160 PMCID: PMC7696810 DOI: 10.3390/cancers12113327] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/31/2020] [Accepted: 11/09/2020] [Indexed: 12/23/2022] Open
Abstract
More people are surviving longer with cancer. Whilst this can be partially attributed to advances in early detection of cancers, there is little doubt that the improvement in survival statistics is also due to the expansion in the spectrum of treatments available for efficacious treatment. Transformative amongst those are immunotherapies, which have proven effective agents for treating immunogenic forms of cancer, although immunologically "cold" tumour types remain refractive. Oncolytic viruses, such as those based on adenovirus, have great potential as anti-cancer agents and have seen a resurgence of interest in recent years. Amongst their many advantages is their ability to induce immunogenic cell death (ICD) of infected tumour cells, thus providing the alluring potential to synergise with immunotherapies by turning immunologically "cold" tumours "hot". Additionally, enhanced immune mediated cell killing can be promoted through the local overexpression of immunological transgenes, encoded from within the engineered viral genome. To achieve this full potential requires the development of refined, tumour selective "precision virotherapies" that are extensively engineered to prevent off-target up take via native routes of infection and targeted to infect and replicate uniquely within malignantly transformed cells. Here, we review the latest advances towards this holy grail within the adenoviral field.
Collapse
Affiliation(s)
| | | | - Alan L. Parker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (T.G.C.); (E.A.B.)
| |
Collapse
|
20
|
Liu F, Dai S, Feng D, Peng X, Qin Z, Kearns AC, Huang W, Chen Y, Ergün S, Wang H, Rappaport J, Bryda EC, Chandrasekhar A, Aktas B, Hu H, Chang SL, Gao B, Qin X. Versatile cell ablation tools and their applications to study loss of cell functions. Cell Mol Life Sci 2019; 76:4725-4743. [PMID: 31359086 PMCID: PMC6858955 DOI: 10.1007/s00018-019-03243-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/22/2022]
Abstract
Targeted cell ablation is a powerful approach for studying the role of specific cell populations in a variety of organotypic functions, including cell differentiation, and organ generation and regeneration. Emerging tools for permanently or conditionally ablating targeted cell populations and transiently inhibiting neuronal activities exhibit a diversity of application and utility. Each tool has distinct features, and none can be universally applied to study different cell types in various tissue compartments. Although these tools have been developed for over 30 years, they require additional improvement. Currently, there is no consensus on how to select the tools to answer the specific scientific questions of interest. Selecting the appropriate cell ablation technique to study the function of a targeted cell population is less straightforward than selecting the method to study a gene's functions. In this review, we discuss the features of the various tools for targeted cell ablation and provide recommendations for optimal application of specific approaches.
Collapse
Affiliation(s)
- Fengming Liu
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Shen Dai
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiao Peng
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Zhongnan Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Alison C Kearns
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Wenfei Huang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Yong Chen
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, 261053, Weifang, People's Republic of China
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximillan University, 97070, Wurzburg, Germany
| | - Hong Wang
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Jay Rappaport
- Division of Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Elizabeth C Bryda
- Rat Resource and Research Center, University of Missouri, 4011 Discovery Drive, Columbia, MO, 65201, USA
| | - Anand Chandrasekhar
- Division of Biological Sciences, 340D Life Sciences Center, University of Missouri, 1201 Rollins St, Columbia, MO, USA
| | - Bertal Aktas
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sulie L Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuebin Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA.
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA.
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
21
|
Zhang KL, Li RP, Zhang BP, Gao ST, Li B, Huang CJ, Cao R, Cheng JY, Xie XD, Yu ZH, Feng XY. Efficacy of a new oncolytic adenovirus armed with IL-13 against oral carcinoma models. Onco Targets Ther 2019; 12:6515-6523. [PMID: 31616161 PMCID: PMC6699363 DOI: 10.2147/ott.s203638] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/09/2019] [Indexed: 12/27/2022] Open
Abstract
Purpose The efficacy of traditional therapies for oral carcinoma (OC) is limited. Oncolytic adenovirus, a novel strategy of cancer therapy, shows potential use in OC treatment. However, its clinical application is limited by pre-existing neutralizing antibodies. Thus, this study aimed to examine the efficacy of a new modified adenovirus against OC in vitro and in vivo. Materials and methods A multiple modified adenovirus (MMAD) armed with IL-13 (MMAD-IL-13) was constructed, and its effect on Cal-27 cells was examined. The potency of MMAD-IL-13 was examined in vitro and in vivo. For in vitro experiment, CCK-8 kit was used to determine the IC50 of MMAD-IL-3 in OC cell lines. For in vivo experiment, Cal-27 xenograft models were used to determine the antitumor effect of MMAD-IL-13. Apoptosis was measured in Cal-27 cells by Western blotting assay. Immunity response was detected in Cal-27 xenograft models 7 days after intratumoral injection with MMAD-IL-13. The potency of MMAD and MMAD-IL-13 was compared in Cal-27 peripheral blood mononuclear cells (PBMCs) models. Results MMAD-IL-13 was successfully constructed; the harvested virus could be replicated and they overexpressed human IL-13 in Cal-27 cells. Compared with MMAD, MMAD-IL-13 showed enhanced antitumor effect in vitro by inducing apoptosis and reducing percentage of M2 macrophages in tumor environment in vivo. MMAD-IL-13 also showed potent antitumor effect in Cal-27, SCC-4, and Tca8113 cells in vitro and in Cal-27 xenograft models in vivo. However, MMAD-IL13 did not harm normal human oral epithelial cells in vitro and exhibited no effect on body weight in Cal-27 xenograft models. In Cal-27 PBMC models, MMAD-IL-13 showed stronger antitumor effect than MMAD. Conclusion A new oncolytic adenovirus carrying the human IL-13 gene was constructed. This virus effectively led to remission of tumor development and death of OC cells in vivo and in vitro, showing its potential as a clinical cancer therapy.
Collapse
Affiliation(s)
- Kai-Liang Zhang
- Department of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Rui-Ping Li
- Department of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Bao-Ping Zhang
- Department of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.,Department of Civil Engineering and Mechanics, Lanzhou University, Gansu 730000, People's Republic of China
| | - Shu-Ting Gao
- Department of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Bo Li
- Department of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Chun-Juan Huang
- Department of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Rui Cao
- Department of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Jing-Yang Cheng
- Department of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xiao-Dong Xie
- Department of Basic Medical Sciences, Lanzhou University, Gansu 730000, People's Republic of China
| | - Zhan-Hai Yu
- Department of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xin-Yu Feng
- Department of Civil Engineering and Mechanics, Lanzhou University, Gansu 730000, People's Republic of China
| |
Collapse
|
22
|
Harrington K, Freeman DJ, Kelly B, Harper J, Soria JC. Optimizing oncolytic virotherapy in cancer treatment. Nat Rev Drug Discov 2019; 18:689-706. [PMID: 31292532 DOI: 10.1038/s41573-019-0029-0] [Citation(s) in RCA: 337] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
In the wake of the success of modern immunotherapy, oncolytic viruses (OVs) are currently seen as a potential therapeutic option for patients with cancer who do not respond or fail to achieve durable responses following treatment with immune checkpoint inhibitors. OVs offer a multifaceted therapeutic platform because they preferentially replicate in tumour cells, can be engineered to express transgenes that augment their cytotoxic and immunostimulatory activities, and modulate the tumour microenvironment to optimize immune-mediated tumour eradication, both at locoregional and systemic sites of disease. Lysis of tumour cells releases tumour-specific antigens that trigger both the innate and adaptive immune systems. OVs also represent attractive combination partners with other systemically delivered agents by virtue of their highly favourable safety profiles. Rational combinations of OVs with different immune modifiers and/or antitumour agents, based on mechanisms of tumour resistance to immune-mediated attack, may benefit the large, currently underserved, population of patients who respond poorly to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Kevin Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK.
| | | | - Beth Kelly
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Jean-Charles Soria
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA.,Department of Medicine and Medical Oncology, Université Paris-Sud, Orsay, France
| |
Collapse
|
23
|
Jahanban‐Esfahlan R, Seidi K, Majidinia M, Karimian A, Yousefi B, Nabavi SM, Astani A, Berindan‐Neagoe I, Gulei D, Fallarino F, Gargaro M, Manni G, Pirro M, Xu S, Sadeghi M, Nabavi SF, Shirooie S. Toll‐like receptors as novel therapeutic targets for herpes simplex virus infection. Rev Med Virol 2019; 29:e2048. [DOI: 10.1002/rmv.2048] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/12/2019] [Accepted: 03/19/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Rana Jahanban‐Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical SciencesTabriz University of Medical Sciences Tabriz Iran
- Drug Applied Research CenterTabriz University of Medical Sciences Tabriz Iran
| | - Khaled Seidi
- Immunology Research CenterTabriz University of Medical Sciences Tabriz Iran
| | - Maryam Majidinia
- Solid Tumor Research CenterUrmia University of Medical Sciences Urmia Iran
| | - Ansar Karimian
- Cellular and Molecular Biology Research Center, Health Research InstituteBabol University of Medical Sciences Babol Iran
| | - Bahman Yousefi
- Molecular Medicine Research CenterTabriz University of Medical Sciences Tabriz Iran
- Department of Biochemistry and Clinical Laboratories, Faculty of MedicineTabriz University of Medical Science Tabriz Iran
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research CenterBaqiyatallah University of Medical Sciences Tehran Iran
| | - Akram Astani
- Department of MicrobiologyShahid Sadoughi University of Medical Sciences Yazd Iran
| | - Ioana Berindan‐Neagoe
- MEDFUTURE ‐Research Center for Advanced Medicine“Iuliu‐Hatieganu” University of Medicine and Pharmacy Cluj‐Napoca Romania
- Research Centerfor Functional Genomics, Biomedicine and Translational Medicine“Iuliu‐Hatieganu” University of Medicine and Pharmacy Cluj‐Napoca Romania
- Department of Functional Genomics and Experimental PathologyThe Oncology Institute “Prof. Dr. Ion Chiricuţă” Cluj‐Napoca Romania
| | - Diana Gulei
- MEDFUTURE ‐Research Center for Advanced Medicine“Iuliu‐Hatieganu” University of Medicine and Pharmacy Cluj‐Napoca Romania
| | | | - Marco Gargaro
- Department of Experimental MedicineUniversity of Perugia Italy
| | - Giorgia Manni
- Department of Experimental MedicineUniversity of Perugia Italy
| | - Matteo Pirro
- Department of MedicineUniversity of Perugia Italy
| | - Suowen Xu
- Aab Cardiovascular Research InstituteUniversity of Rochester Rochester NY USA
| | - Mahmoud Sadeghi
- Department of Transplantation ImmunologyUniversity of Heidelberg Heidelberg Germany
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research CenterBaqiyatallah University of Medical Sciences Tehran Iran
| | - Samira Shirooie
- Department of Pharmacology, Faculty of PharmacyKermanshah University of Medical Sciences Kermanshah Iran
| |
Collapse
|
24
|
Wang Y, Zhou X, Wu Z, Hu H, Jin J, Hu Y, Dong Y, Zou J, Mao Z, Shi X, Huo Y, Lyu J, Fang Z, Zhang W, Zhu Y, Li B, Liu B. Preclinical Safety Evaluation of Oncolytic Herpes Simplex Virus Type 2. Hum Gene Ther 2019; 30:651-660. [PMID: 30499341 DOI: 10.1089/hum.2018.170] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Oncolytic virotherapy is a new and safe therapeutic strategy based on the inherent cytotoxicity of oncolytic viruses and their ability to replicate and spread within tumors in a selective manner. In a previous study, a new type of oncolytic herpes simplex virus type 2 (oHSV-2, named OH2) was constructed to treat human cancers. That study demonstrated that OH2 is genetically and biologically stable. Its antitumor activity was maintained, even after passaging the virus for >20 generations. To advance OH2 into a clinical trial, a systematic preclinical safety evaluation was performed, which included: an acute toxicity test of OH2 in BALB/c mice; repeated dose toxicity tests of OH2 in BALB/c mice and cynomolgus monkeys; and biodistribution assays of OH2 in BALB/c mice, tumor-bearing mice, tumor-bearing nude mice, and cynomolgus monkeys. The results of this preclinical safety evaluation of OH2 indicate that OH2 is safe and suitable for clinical trials.
Collapse
Affiliation(s)
- Yang Wang
- 1 National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Centre of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, P.R. China
| | - Xiaobing Zhou
- 2 National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control (NIFDC), Beijing, P.R. China
| | - Zhen Wu
- 1 National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Centre of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, P.R. China
| | - Han Hu
- 1 National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Centre of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, P.R. China
| | - Jing Jin
- 1 National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Centre of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, P.R. China
| | - Yanping Hu
- 2 National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control (NIFDC), Beijing, P.R. China
| | - Yuting Dong
- 3 Wuhan Binhui Biotechnology Co. Ltd., Wuhan, P.R. China
| | - Jianwen Zou
- 3 Wuhan Binhui Biotechnology Co. Ltd., Wuhan, P.R. China
| | - Zeyong Mao
- 3 Wuhan Binhui Biotechnology Co. Ltd., Wuhan, P.R. China
| | - Xiaotai Shi
- 3 Wuhan Binhui Biotechnology Co. Ltd., Wuhan, P.R. China
| | - Yan Huo
- 2 National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control (NIFDC), Beijing, P.R. China
| | - Jianjun Lyu
- 2 National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control (NIFDC), Beijing, P.R. China
| | - Zhizheng Fang
- 3 Wuhan Binhui Biotechnology Co. Ltd., Wuhan, P.R. China
| | - Wen Zhang
- 4 National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Pekin Union Medical College, Beijing, P.R. China
| | - Yujie Zhu
- 4 National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Pekin Union Medical College, Beijing, P.R. China
| | - Bo Li
- 2 National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control (NIFDC), Beijing, P.R. China
| | - Binlei Liu
- 1 National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Centre of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, P.R. China.,3 Wuhan Binhui Biotechnology Co. Ltd., Wuhan, P.R. China
| |
Collapse
|
25
|
Montagnaro S, Damiano S, Ciarcia R, Puzio MV, Ferrara G, Iovane V, Forte IM, Giordano A, Pagnini U. Caprine herpesvirus 1 (CpHV-1) as a potential candidate for oncolytic virotherapy. Cancer Biol Ther 2018; 20:42-51. [PMID: 30409104 DOI: 10.1080/15384047.2018.1504722] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Caprine Herpesvirus type 1 (CpHV-1) is a species-specific herpes virus able to induce apoptosis in several biological systems. In the present study we aimed to investigate the ability of CpHV-1 to reduce cells viability, to replicate and to cause cell death also in human cancer cell lines. We tested the CpHV-1 effects on HEL-299, Vero, MDA-MB-468, HeLa, U2OS, PC3, A549 and K562 neoplastic cell lines and on MDBK cells. Firstly, we evaluated the effect of CpHV-1 infection on cell viability by MTT assay and our data showed that CpHV-1 can induce a marked cytopathic effect (CPE) in most of cell lines tested, except for HEL-299, Vero and K562 cells. The reduction of cell viability was associated with a significant increase of viral production. We next investigated if CpHV-1 was able to induce cell death and so through western blotting analysis we evaluated cleaved caspase 3, LC3II and p62 protein levels after infection. Caspase 3 activation was detected in MDBK cells and, even if at different times p.i., also in MDA-MB-468, U2OS, and PC3 cell lines, while LC3II increase and concomitant p62 protein reduction were observed only in U2OS, and A549 cells, no significant alteration of these proteins was observed in the other cell lines tested. Finally, to confirm virus ability to trigger apoptosis we performed an Annexin-V apoptosis test after 24 h p.i. Although we need to further explore mechanisms underlying CpHV-1 treatment, this study could serve as the basis for the development of new treatment options aiming to fight several cancer types.
Collapse
Affiliation(s)
- Serena Montagnaro
- a Department of Veterinary Medicine and Animal Productions , University of Naples "Federico II" , Napoli , Italia
| | - Sara Damiano
- a Department of Veterinary Medicine and Animal Productions , University of Naples "Federico II" , Napoli , Italia
| | - Roberto Ciarcia
- a Department of Veterinary Medicine and Animal Productions , University of Naples "Federico II" , Napoli , Italia
| | - Maria Valeria Puzio
- a Department of Veterinary Medicine and Animal Productions , University of Naples "Federico II" , Napoli , Italia
| | - Gianmarco Ferrara
- a Department of Veterinary Medicine and Animal Productions , University of Naples "Federico II" , Napoli , Italia
| | - Valentina Iovane
- b Department of Pharmacy , University of Salerno , Fisciano (Na) , Italia
| | - Iris Maria Forte
- c OncologyResearch Center of Mercogliano (CROM) , Istituto Nazionale Tumori - IRCCS, "Fondazione G. Pascale" , Napoli , Italia
| | - Antonio Giordano
- d Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology , Temple University , Philadelphia , Pennsylvania , USA
| | - Ugo Pagnini
- a Department of Veterinary Medicine and Animal Productions , University of Naples "Federico II" , Napoli , Italia
| |
Collapse
|
26
|
Ebrahimi S, Teimoori A, Khanbabaei H, Tabasi M. Harnessing CRISPR/Cas 9 System for manipulation of DNA virus genome. Rev Med Virol 2018; 29:e2009. [PMID: 30260068 DOI: 10.1002/rmv.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/03/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022]
Abstract
The recent development of the Clustered Regularly Interspaced Palindromic Repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) system, a genome editing system, has many potential applications in virology. The possibility of introducing site specific breaks has provided new possibilities to precisely manipulate viral genomics. Here, we provide diagrams to summarize the steps involved in the process. We also systematically review recent applications of the CRISPR/Cas9 system for manipulation of DNA virus genomics and discuss the therapeutic potential of the system to treat viral diseases.
Collapse
Affiliation(s)
- Saeedeh Ebrahimi
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Virology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Teimoori
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Virology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hashem Khanbabaei
- Medical Physics Department, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Tabasi
- Department of Virology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
27
|
Liu SH, Yu J, Sanchez R, Liu X, Heidt D, Willey J, Nemunaitis J, Brunicardi FC. A novel synthetic human insulin super promoter for targeting PDX-1-expressing pancreatic cancer. Cancer Lett 2018; 418:75-83. [PMID: 29309817 DOI: 10.1016/j.canlet.2018.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/19/2017] [Accepted: 01/03/2018] [Indexed: 02/07/2023]
Abstract
Our previous studies have shown that a rat insulin promoter II fragment (RIP) was used to effectively target pancreatic adenocarcinoma (PDAC) and insulinoma that over-express pancreatic and duodenal homeobox-1 (PDX-1). To enhance the activity and specificity of the human insulin promoter, we engineered a synthetic human insulin super-promoter (SHIP). Reporter assay demonstrated that SHIP1 was the most powerful promoter among all of the SHIPs and had far greater activity than the endogenous human insulin promoters and RIP in PDAC expressing PDX-1. Over-expression, knockdown and competitive inhibition of PDX-1 expression assay proved that PDX-1 is a critical transcript factor to regulate the activity of SHIP1. SHIP1-driven viral thymidine kinase followed by ganciclovir (SHIP1-TK/GCV) resulted in cytotoxicity to PDAC cells in vitro. Systemic delivery of SHIP1-TK/GCV in PDAC xenograft mice significantly suppressed PANC-1 tumor growth in vivo greater than RIP-TK/GCV and CMV-TK/GCV controls (p < .05). These preclinical data suggest that SHIP1 is a powerful novel promoter that can be used to target human PDAC expressing PDX-1 in clinical trials. Furthermore, this novel strategy of engineering synthetic super-promoters could be used for other cancer targets.
Collapse
Affiliation(s)
- Shi-He Liu
- Department of Surgery, University of Toledo College of Medicine & Life Sciences, Toledo OH, USA
| | - Juehua Yu
- Department of Surgery, University of California at Los Angeles, CA, USA
| | - Robbi Sanchez
- Department of Surgery, University of California at Los Angeles, CA, USA
| | - Xiaochen Liu
- Department of Surgery, University of Toledo College of Medicine & Life Sciences, Toledo OH, USA
| | - David Heidt
- Department of Surgery, University of Toledo College of Medicine & Life Sciences, Toledo OH, USA
| | - James Willey
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo OH, USA
| | - John Nemunaitis
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo OH, USA
| | | |
Collapse
|
28
|
Chen X, Han J, Chu J, Zhang L, Zhang J, Chen C, Chen L, Wang Y, Wang H, Yi L, Elder JB, Wang QE, He X, Kaur B, Chiocca EA, Yu J. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 2017; 7:27764-77. [PMID: 27050072 PMCID: PMC5053686 DOI: 10.18632/oncotarget.8526] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/16/2016] [Indexed: 12/11/2022] Open
Abstract
Breast cancer brain metastases (BCBMs) are common in patients with metastatic breast cancer and indicate a poor prognosis. These tumors are especially resistant to currently available treatments due to multiple factors. However, the combination of chimeric antigen receptor (CAR)-modified immune cells and oncolytic herpes simplex virus (oHSV) has not yet been explored in this context. In this study, NK-92 cells and primary NK cells were engineered to express the second generation of EGFR-CAR. The efficacies of anti-BCBMs of EGFR-CAR NK cells, oHSV-1, and their combination were tested in vitro and in a breast cancer intracranial mouse model. In vitro, compared with mock-transduced NK-92 cells or primary NK cells, EGFR-CAR-engineered NK-92 cells and primary NK cells displayed enhanced cytotoxicity and IFN-γ production when co-cultured with breast cancer cell lines MDA-MB-231, MDA-MB-468, and MCF-7. oHSV-1 alone was also capable of lysing and destroying these cells. However, a higher cytolytic effect of EGFR-CAR NK-92 cells was observed when combined with oHSV-1 compared to the monotherapies. In the mice intracranially pre-inoculated with EGFR-expressing MDA-MB-231 cells, intratumoral administration of either EGFR-CAR-transduced NK-92 cells or oHSV-1 mitigated tumor growth. Notably, the combination of EGFR-CAR NK-92 cells with oHSV-1 resulted in more efficient killing of MDA-MB-231 tumor cells and significantly longer survival of tumor-bearing mice when compared to monotherapies. These results demonstrate that regional administration of EGFR-CAR NK-92 cells combined with oHSV-1 therapy is a potentially promising strategy to treat BCBMs.
Collapse
Affiliation(s)
- Xilin Chen
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA.,Lymphoma/Head and Neck Oncology Department, 307 Hospital, Beijing 100071, China
| | - Jianfeng Han
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - Jianhong Chu
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA.,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou 215000, China
| | - Lingling Zhang
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University, Columbus, Ohio 43210, USA
| | - Charlie Chen
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - Luxi Chen
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - Youwei Wang
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - Hongwei Wang
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - Long Yi
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - J Bradley Elder
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA.,Department of Neurological Surgery, The Ohio State University, Columbus, Ohio 43210, USA
| | - Qi-En Wang
- Department of Radiology, The Ohio State University, Columbus, Ohio 43210, USA
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Balveen Kaur
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA.,Department of Neurological Surgery, The Ohio State University, Columbus, Ohio 43210, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital and Harvey Cushing Neuro-oncology Laboratories, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA.,The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA.,The James Cancer Hospital, Columbus, OH 43210, USA
| |
Collapse
|
29
|
Mooney R, Abdul Majid A, Batalla J, Annala AJ, Aboody KS. Cell-mediated enzyme prodrug cancer therapies. Adv Drug Deliv Rev 2017; 118:35-51. [PMID: 28916493 DOI: 10.1016/j.addr.2017.09.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/15/2017] [Accepted: 09/06/2017] [Indexed: 02/08/2023]
Abstract
Cell-directed gene therapy is a promising new frontier for the field of targeted cancer therapies. Here we discuss the current pre-clinical and clinical use of cell-mediated enzyme prodrug therapy (EPT) directed against solid tumors and avenues for further development. We also discuss some of the challenges encountered upon translating these therapies to clinical trials. Upon sufficient development, cell-mediated enzyme prodrug therapy has the potential to maximize the distribution of therapeutic enzymes within the tumor environment, localizing conversion of prodrug to active drug at the tumor sites thereby decreasing off-target toxicities. New combinatorial possibilities are also promising. For example, when combined with viral gene-delivery vehicles, this may result in new hybrid vehicles that attain heretofore unmatched levels of therapeutic gene expression within the tumor.
Collapse
|
30
|
Aderibigbe BA. Metal-Based Nanoparticles for the Treatment of Infectious Diseases. Molecules 2017; 22:E1370. [PMID: 28820471 PMCID: PMC6152252 DOI: 10.3390/molecules22081370] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/15/2017] [Indexed: 11/16/2022] Open
Abstract
Infectious diseases can be transmitted and they cause a significant burden on public health globally. They are the greatest world killers and it is estimated that they are responsible for the demise of over 17 million people annually. The impact of these diseases is greater in the developing countries. People with compromised immune systems and children are the most affected. Infectious diseases may be caused by bacteria, viruses, and protozoa. The treatment of infectious diseases is hampered by simultaneous resistance to multiple drugs, indicating that there is a serious and pressing need to develop new therapeutics that can overcome drug resistance. This review will focus on the recent reports of metal-based nanoparticles that are potential therapeutics for the treatment of infectious diseases and their biological efficacy (in vitro and in vivo).
Collapse
Affiliation(s)
- Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa.
| |
Collapse
|
31
|
Asad AS, Moreno Ayala MA, Gottardo MF, Zuccato C, Nicola Candia AJ, Zanetti FA, Seilicovich A, Candolfi M. Viral gene therapy for breast cancer: progress and challenges. Expert Opin Biol Ther 2017; 17:945-959. [DOI: 10.1080/14712598.2017.1338684] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Antonela S. Asad
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariela A. Moreno Ayala
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M. Florencia Gottardo
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Camila Zuccato
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro Javier Nicola Candia
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flavia A. Zanetti
- Instituto de Ciencia y Tecnología César Milstein (ICT Milstein), Unidad Ejecutora del Consejo Nacional de Investigaciones Científicas y Técnicas, Fundación Pablo Cassará, Buenos Aires, Argentina
| | - Adriana Seilicovich
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Candolfi
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
32
|
Zaimy MA, Saffarzadeh N, Mohammadi A, Pourghadamyari H, Izadi P, Sarli A, Moghaddam LK, Paschepari SR, Azizi H, Torkamandi S, Tavakkoly-Bazzaz J. New methods in the diagnosis of cancer and gene therapy of cancer based on nanoparticles. Cancer Gene Ther 2017; 24:233-243. [PMID: 28574057 DOI: 10.1038/cgt.2017.16] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 03/17/2017] [Accepted: 04/12/2017] [Indexed: 12/31/2022]
Abstract
Cancer is one of the leading cause of death in the world with the prevalence of >10 million mortalities annually. Current cancer treatments include surgical intervention, radiation, and taking chemotherapeutic drugs, which often kill the healthy cells and result in toxicity in patients. Therefore, researchers are looking for ways to be able to eliminate just cancerous cells. Intra-tumor heterogeneity of cancerous cells is the main obstacle on the way of an effective cancer treatment. However, better comprehension of molecular basis of tumor and the advent of new diagnostic technologies can help to improve the treatment of various cancers. Therefore, study of epigenetic changes, gene expression of cancerous cells and employing methods that enable us to correct or minimize these changes is critically important. In this paper, we will review the recent advanced strategies being used in the field of cancer research.
Collapse
Affiliation(s)
- M A Zaimy
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - N Saffarzadeh
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - A Mohammadi
- Center of Excellence for Biodiversity, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - H Pourghadamyari
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - P Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - A Sarli
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - L K Moghaddam
- Department of Developmental Cell Biology, School of Biological Sciences, Azad University, Tehran North Branch, Tehran, Iran
| | - S R Paschepari
- Department of Developmental Cell Biology, School of Biological Sciences, Azad University, Tehran North Branch, Tehran, Iran
| | - H Azizi
- Department of Medical Parasitology, Zabol University of Medical Sciences, Zabol, Iran
| | - S Torkamandi
- Department of Medical Genetics and immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - J Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Sanchala DS, Bhatt LK, Prabhavalkar KS. Oncolytic Herpes Simplex Viral Therapy: A Stride toward Selective Targeting of Cancer Cells. Front Pharmacol 2017; 8:270. [PMID: 28559846 PMCID: PMC5432606 DOI: 10.3389/fphar.2017.00270] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/01/2017] [Indexed: 12/18/2022] Open
Abstract
Oncolytic viral therapy, which makes use of replication-competent lytic viruses, has emerged as a promising modality to treat malignancies. It has shown meaningful outcomes in both solid tumor and hematologic malignancies. Advancements during the last decade, mainly genetic engineering of oncolytic viruses have resulted in improved specificity and efficacy of oncolytic viruses in cancer therapeutics. Oncolytic viral therapy for treating cancer with herpes simplex virus-1 has been of particular interest owing to its range of benefits like: (a) large genome and power to infiltrate in the tumor, (b) easy access to manipulation with the flexibility to insert multiple transgenes, (c) infecting majority of the malignant cell types with quick replication in the infected cells and (d) as Anti-HSV agent to terminate HSV replication. This review provides an exhaustive list of oncolytic herpes simplex virus-1 along with their genetic alterations. It also encompasses the major developments in oncolytic herpes simplex-1 viral therapy and outlines the limitations and drawbacks of oncolytic herpes simplex viral therapy.
Collapse
Affiliation(s)
| | - Lokesh K. Bhatt
- Department of Pharmacology, Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W)Mumbai, India
| | | |
Collapse
|
34
|
Dummer R, Hoeller C, Gruter IP, Michielin O. Combining talimogene laherparepvec with immunotherapies in melanoma and other solid tumors. Cancer Immunol Immunother 2017; 66:683-695. [PMID: 28238174 PMCID: PMC5445176 DOI: 10.1007/s00262-017-1967-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/31/2017] [Indexed: 01/07/2023]
Abstract
Talimogene laherparepvec is a first-in-class intralesional oncolytic immunotherapy. In a recent Phase III trial (OPTiM), talimogene laherparepvec significantly improved durable response rate compared with subcutaneous granulocyte–macrophage colony-stimulating factor (GM-CSF). Overall response rate was also higher in the talimogene laherparepvec arm, and the greatest efficacy was demonstrated in patients with earlier-stage (IIIB, IIIC, or IVM1a) melanoma. Talimogene laherparepvec was well tolerated, with the majority (89%) of adverse events being grade 1 or 2. Preclinical studies have shown that talimogene laherparepvec exerts antitumor activity by selectively replicating within and destroying cancer cells, and through the release of tumor-associated antigens and expression of GM-CSF, which facilitates a wider antitumor immune response. It is hypothesized that combining talimogene laherparepvec with a systemic immunotherapy may, by bringing together complementary mechanisms of action, further enhance the efficacy of both agents. Indeed, talimogene laherparepvec is currently being assessed in combination with immune checkpoint inhibitors, including ipilimumab and pembrolizumab, in trials for melanoma and other solid tumors. Early results in melanoma indicate that the combination of talimogene laherparepvec with ipilimumab or pembrolizumab has greater efficacy than either therapy alone, without additional safety concerns above those expected for each monotherapy. In this review, we discuss the latest results from trials assessing talimogene laherparepvec in combination with other immunotherapies, provide an overview of ongoing and upcoming combination trials, and suggest future directions for talimogene laherparepvec in combination therapy for solid tumors.
Collapse
Affiliation(s)
- Reinhard Dummer
- Department of Dermatology, University of Zürich Hospital, Gloriastrasse 31, 8091, Zurich, Switzerland.
| | | | | | | |
Collapse
|
35
|
Bommareddy PK, Patel A, Hossain S, Kaufman HL. Talimogene Laherparepvec (T-VEC) and Other Oncolytic Viruses for the Treatment of Melanoma. Am J Clin Dermatol 2017; 18:1-15. [PMID: 27988837 DOI: 10.1007/s40257-016-0238-9] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Many mammalian viruses have properties that can be commandeered for the treatment of cancer. These characteristics include preferential infection and replication in tumor cells, the initiation of tumor cell lysis, and the induction of innate and adaptive anti-tumor immunity. Furthermore, viruses can be genetically engineered to reduce pathogenicity and increase immunogenicity resulting in minimally toxic therapeutic agents. Talimogene laherparepvec (T-VEC; Imlygic™), is a genetically modified herpes simplex virus, type 1, and is the first oncolytic virus therapy to be approved for the treatment of advanced melanoma by the US FDA. T-VEC is attenuated by the deletion of the herpes neurovirulence viral genes and enhanced for immunogenicity by the deletion of the viral ICP47 gene. Immunogenicity is further supported by expression of the human granulocyte-macrophage colony-stimulating factor (GM-CSF) gene, which helps promote the priming of T cell responses. T-VEC demonstrated significant improvement in durable response rate, objective response rate, and progression-free survival in a randomized phase III clinical trial for patients with advanced melanoma. This review will discuss the optimal selection of patients for such treatment and describe how therapy is optimally delivered. We will also discuss future directions for oncolytic virus immunotherapy, which will likely include combination T-VEC clinical trials, expansion of T-VEC to other types of non-melanoma skin cancers, and renewed efforts at oncolytic virus drug development with other viruses.
Collapse
|
36
|
Zhang Y, Wang J, Cheng H, Sun Y, Liu M, Wu Z, Pei R. Conditional control of suicide gene expression in tumor cells with theophylline-responsive ribozyme. Gene Ther 2016; 24:84-91. [DOI: 10.1038/gt.2016.78] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/06/2016] [Accepted: 11/11/2016] [Indexed: 12/18/2022]
|
37
|
Perny M, Muri L, Dawson H, Kleinlogel S. Chronic activation of the D156A point mutant of Channelrhodopsin-2 signals apoptotic cell death: the good and the bad. Cell Death Dis 2016; 7:e2447. [PMID: 27809305 PMCID: PMC5260891 DOI: 10.1038/cddis.2016.351] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/09/2016] [Accepted: 09/26/2016] [Indexed: 12/24/2022]
Abstract
Channelrhodopsin-2 (ChR2) has become a celebrated research tool and is considered a promising potential therapeutic for neurological disorders. While making its way into the clinic, concerns about the safety of chronic ChR2 activation have emerged; in particular as the high-intensity blue light illumination needed for ChR2 activation may be phototoxic. Here we set out to quantify for the first time the cytotoxic effects of chronic ChR2 activation. We studied the safety of prolonged illumination on ChR2(D156A)-expressing human melanoma cells as cancer cells are notorious for their resistance to killing. Three days of illumination eradicated the entire ChR2(D156A)-expressing cell population through mitochondria-mediated apoptosis, whereas blue light activation of non-expressing control cells did not significantly compromise cell viability. In other words, chronic high-intensity blue light illumination alone is not phototoxic, but prolonged ChR2 activation induces mitochondria-mediated apoptosis. The results are alarming for gain-of-function translational neurological studies but open the possibility to optogenetically manipulate the viability of non-excitable cells, such as cancer cells. In a second set of experiments we therefore evaluated the feasibility to put melanoma cell proliferation and apoptosis under the control of light by transdermally illuminating in vivo melanoma xenografts expressing ChR2(D156A). We show clear proof of principle that light treatment inhibits and even reverses tumor growth, rendering ChR2s potential tools for targeted light-therapy of cancers.
Collapse
Affiliation(s)
- Michael Perny
- Institute for Physiology, University of Bern, Bern 3012 Switzerland
| | - Lukas Muri
- Institute for Physiology, University of Bern, Bern 3012 Switzerland
| | - Heather Dawson
- Institute of Pathology, Clinical Pathology Division, University of Bern, Bern 3010 Switzerland
| | - Sonja Kleinlogel
- Institute for Physiology, University of Bern, Bern 3012 Switzerland
| |
Collapse
|
38
|
Rehman H, Silk AW, Kane MP, Kaufman HL. Into the clinic: Talimogene laherparepvec (T-VEC), a first-in-class intratumoral oncolytic viral therapy. J Immunother Cancer 2016; 4:53. [PMID: 27660707 PMCID: PMC5029010 DOI: 10.1186/s40425-016-0158-5] [Citation(s) in RCA: 281] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022] Open
Abstract
With the recent regulatory approval of Talimogene laherparepvec (T-VEC) for the treatment of advanced of melanoma in the United States, Europe and Australia, oncolytic virus immunotherapy has earned its place in the clinic. However, the adoption of T-VEC by the U.S. oncology community has been slow, and so far has been largely limited to specialized cancer centers. Limiting factors include the intratumoral route of administration, which is unfamiliar to medical oncologists, biosafety concerns related to the use of a live virus in the clinic, and the explosion of other therapeutic strategies now available for the treatment of advanced melanoma. Herein, we review the development of T-VEC, and suggest how it fits into the in the current clinical treatment paradigm, and provide pearls for drug preparation, administration, and monitoring of response to therapy.
Collapse
Affiliation(s)
- Hasan Rehman
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, Room 2508B, New Brunswick, NJ 08901 USA
| | - Ann W Silk
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, Room 2508B, New Brunswick, NJ 08901 USA
| | - Michael P Kane
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, Room 2508B, New Brunswick, NJ 08901 USA
| | - Howard L Kaufman
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, Room 2508B, New Brunswick, NJ 08901 USA
| |
Collapse
|
39
|
Harrington KJ, Puzanov I, Hecht JR, Hodi FS, Szabo Z, Murugappan S, Kaufman HL. Clinical development of talimogene laherparepvec (T-VEC): a modified herpes simplex virus type-1-derived oncolytic immunotherapy. Expert Rev Anticancer Ther 2016; 15:1389-403. [PMID: 26558498 DOI: 10.1586/14737140.2015.1115725] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tumor immunotherapy is emerging as a promising new treatment option for patients with cancer. T-VEC is an intralesional oncolytic virus therapy based on a modified herpes simplex virus type-1. T-VEC selectively targets tumor cells, causing regression in injected lesions and inducing immunologic responses that mediate regression at uninjected/distant sites. In a randomized phase III trial, T-VEC met its primary endpoint of improving the durable response rate vs granulocyte-macrophage colony-stimulating factor in patients with unresectable melanoma. Responses were observed in injected and uninjected regional and visceral lesions. Exploratory analyses suggested survival differences in favor of T-VEC in patients with untreated or stage IIIB/IIIC/IVM1a disease. T-VEC was generally well tolerated, the most common adverse events being flu-like symptoms. Here, we overview recent advances in cancer immunotherapy, focusing on the clinical development of T-VEC, from first-in-human studies and studies in other cancer types, to ongoing combination trials with checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Igor Puzanov
- a Division of Hematology-Oncology, Vanderbilt University Medical Center , Nashville , TN , USA
| | - J Randolph Hecht
- b David Geffen School of Medicine , UCLA , Los Angeles , CA , USA
| | - F Stephen Hodi
- c Melanoma Center and the Center for Immuno-Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Zsolt Szabo
- d Department of Oncology , Amgen (Europe) GmbH , Zug , Switzerland
| | - Swami Murugappan
- e Department of Oncology , Amgen Inc ., Thousand Oaks , CA , USA
| | - Howard L Kaufman
- f Division of Surgical Oncology , Rutgers Cancer Institute of New Jersey , New Brunswick , NJ , USA
| |
Collapse
|
40
|
Samudio I, Rezvani K, Shaim H, Hofs E, Ngom M, Bu L, Liu G, Lee JTC, Imren S, Lam V, Poon GFT, Ghaedi M, Takei F, Humphries K, Jia W, Krystal G. UV-inactivated HSV-1 potently activates NK cell killing of leukemic cells. Blood 2016; 127:2575-86. [PMID: 26941401 PMCID: PMC4892253 DOI: 10.1182/blood-2015-04-639088] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 02/26/2016] [Indexed: 11/20/2022] Open
Abstract
Herein we demonstrate that oncolytic herpes simplex virus-1 (HSV-1) potently activates human peripheral blood mononuclear cells (PBMCs) to lyse leukemic cell lines and primary acute myeloid leukemia samples, but not healthy allogeneic lymphocytes. Intriguingly, we found that UV light-inactivated HSV-1 (UV-HSV-1) is equally effective in promoting PBMC cytolysis of leukemic cells and is 1000- to 10 000-fold more potent at stimulating innate antileukemic responses than UV-inactivated cytomegalovirus, vesicular stomatitis virus, reovirus, or adenovirus. Mechanistically, UV-HSV-1 stimulates PBMC cytolysis of leukemic cells, partly via Toll-like receptor-2/protein kinase C/nuclear factor-κB signaling, and potently stimulates expression of CD69, degranulation, migration, and cytokine production in natural killer (NK) cells, suggesting that surface components of UV-HSV-1 directly activate NK cells. Importantly, UV-HSV-1 synergizes with interleukin-15 (IL-15) and IL-2 in inducing activation and cytolytic activity of NK cells. Additionally, UV-HSV-1 stimulates glycolysis and fatty acid oxidation-dependent oxygen consumption in NK cells, but only glycolysis is required for their enhanced antileukemic activity. Last, we demonstrate that T cell-depleted human PBMCs exposed to UV-HSV-1 provide a survival benefit in a murine xenograft model of human acute myeloid leukemia (AML). Taken together, our results support the preclinical development of UV-HSV-1 as an adjuvant, alone or in combination with IL-15, for allogeneic donor mononuclear cell infusions to treat AML.
Collapse
Affiliation(s)
- Ismael Samudio
- Programa de Investigacion e Innovacion en Leucemia Aguda y Cronica, Bogotá, Colombia; Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Hila Shaim
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Elyse Hofs
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Mor Ngom
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Luke Bu
- Brain Research Centre, University of British Columbia, Vancouver, Canada
| | - Guoyu Liu
- Brain Research Centre, University of British Columbia, Vancouver, Canada
| | - Jason T C Lee
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Suzan Imren
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Vivian Lam
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Grace F T Poon
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Maryam Ghaedi
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Fumio Takei
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Keith Humphries
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - William Jia
- Brain Research Centre, University of British Columbia, Vancouver, Canada
| | - Gerald Krystal
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| |
Collapse
|
41
|
Progress and problems with the use of suicide genes for targeted cancer therapy. Adv Drug Deliv Rev 2016; 99:113-128. [PMID: 26004498 DOI: 10.1016/j.addr.2015.05.009] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 02/19/2015] [Accepted: 05/14/2015] [Indexed: 12/16/2022]
Abstract
Among various gene therapy methods for cancer, suicide gene therapy attracts a special attention because it allows selective conversion of non-toxic compounds into cytotoxic drugs inside cancer cells. As a result, therapeutic index can be increased significantly by introducing high concentrations of cytotoxic molecules to the tumor environment while minimizing impact on normal tissues. Despite significant success at the preclinical level, no cancer suicide gene therapy protocol has delivered the desirable clinical significance yet. This review gives a critical look at the six main enzyme/prodrug systems that are used in suicide gene therapy of cancer and familiarizes readers with the state-of-the-art research and practices in this field. For each enzyme/prodrug system, the mechanisms of action, protein engineering strategies to enhance enzyme stability/affinity and chemical modification techniques to increase prodrug kinetics and potency are discussed. In each category, major clinical trials that have been performed in the past decade with each enzyme/prodrug system are discussed to highlight the progress to date. Finally, shortcomings are underlined and areas that need improvement in order to produce clinical significance are delineated.
Collapse
|
42
|
Whisenhunt TR, Rajneesh KF, Hackney JR, Markert JM. Extended disease-free interval of 6 years in a recurrent glioblastoma multiforme patient treated with G207 oncolytic viral therapy. Oncolytic Virother 2015; 4:33-8. [PMID: 27512668 PMCID: PMC4918376 DOI: 10.2147/ov.s62461] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a relentless primary central nervous system malignancy that remains resistant to conventional therapy despite major advances in clinical neurooncology. This report details the case of a patient who had failed conventional treatment for recurrent GBM and was ultimately treated with a genetically engineered herpes simplex virus (HSV) type 1 vector, G207. METHODS Case report detailing the outcomes of one patient enrolled into the gene therapy arm of the Neurovir G207 protocol whereby stereotactic injection of 120 µL G207 viral suspension containing 1×10(7) plaque-forming units (or active viral particles) was made into the enhancing region of the tumor. RESULTS In this patient, despite aggressive surgical resection, adjuvant radiotherapy and chemotherapy, tumor progression occurred. However, with G207 oncolytic therapy and brief exposures to second and third treatments, this patient had an extended survival time of 7.5 years and a 6-year apparent disease-free interval, an extraordinarily unusual finding in the pretemozolomide era. CONCLUSION With minimal adjunctive chemotherapy, including one course of temozolomide, one course of procarbazine, and four cycles of irinotecan, the patient survived over 7 years before the next recurrence. Addition of G207 to this patient's traditional therapy may have been the critical treatment producing her prolonged survival. This report demonstrates the potential for long-term response to a one-time treatment with oncolytic HSV and encourages continued research on oncolytic viral therapy for GBM.
Collapse
Affiliation(s)
- Thomas R Whisenhunt
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kiran F Rajneesh
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James R Hackney
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
43
|
Characterization of a replication-incompetent pseudorabies virus mutant lacking the sole immediate early gene IE180. mBio 2014; 5:e01850. [PMID: 25389174 PMCID: PMC4235210 DOI: 10.1128/mbio.01850-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The alphaherpesvirus pseudorabies virus (PRV) encodes a single immediate early gene called IE180. The IE180 protein is a potent transcriptional activator of viral genes involved in DNA replication and RNA transcription. A PRV mutant with both copies of IE180 deleted was constructed 20 years ago (S. Yamada and M. Shimizu, Virology 199:366–375, 1994, doi:10.1006/viro.1994.1134), but propagation of the mutant depended on complementing cell lines that expressed the toxic IE180 protein constitutively. Recently, Oyibo et al. constructed a novel set of PRV IE180 mutants and a stable cell line with inducible IE180 expression (H. Oyibo, P. Znamenskiy, H. V. Oviedo, L. W. Enquist, A. Zador, Front. Neuroanat. 8:86, 2014, doi:10.3389/fnana.2014.00086), which we characterized further here. These mutants failed to replicate new viral genomes, synthesize immediate early, early, or late viral proteins, and assemble infectious virions. The PRV IE180-null mutant did not form plaques in epithelial cell monolayers and could not spread from primary infected neurons to second-order neurons in culture. PRV IE180-null mutants lacked the property of superinfection exclusion. When PRV IE180-null mutants infected cells first, subsequent superinfecting viruses were not blocked in cell entry and formed replication compartments in epithelial cells, fibroblasts, and neurons. Cells infected with PRV IE180-null mutants survived as long as uninfected cells in culture while expressing a fluorescent reporter gene. Transcomplementation with IE180 in epithelial cells restored all mutant phenotypes to wild type. The conditional expression of PRV IE180 protein enables the propagation of replication-incompetent PRV IE180-null mutants and will facilitate construction of long-term single-cell-infecting PRV mutants for precise neural circuit tracing and high-capacity gene delivery vectors. Pseudorabies virus (PRV) is widely used for neural tracing in animal models. The virus replicates and spreads between synaptically connected neurons. Current tracing strains of PRV are cytotoxic and kill infected cells. Infected cells exclude superinfection with a second virus, limiting multiple virus infections in circuit tracing. By removing the only immediate early gene of PRV (called IE180), the mutant virus will not replicate or spread in epithelial cells, fibroblasts, or neurons. The wild-type phenotype can be restored by transcomplementation of infected cells with IE180. The PRV IE180-null mutant can express fluorescent reporters for weeks in cells with no toxicity; infected cells survive as long as uninfected cells. Infection with the mutant virus allows superinfection of the same cell with a second virus that can enter and replicate. The PRV IE180-null mutant will permit conditional long-term tracing in animals and is a high-capacity vector for gene delivery.
Collapse
|
44
|
Wang J, Xu L, Zeng W, Hu P, Zeng M, Rabkin SD, Liu R. Treatment of human hepatocellular carcinoma by the oncolytic herpes simplex virus G47delta. Cancer Cell Int 2014; 14:83. [PMID: 25360068 PMCID: PMC4213511 DOI: 10.1186/s12935-014-0083-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 08/05/2014] [Indexed: 12/31/2022] Open
Abstract
Background Oncolytic herpes simplex virus (HSV) can replicate in and kill cancer cells while sparing the adjacent normal tissue. Hepatocellular carcinoma (HCC) is amongst the most common and lethal cancers, especially in Third World countries. In this study, the cytotoxicity of a third-generation oncolytic HSV, G47Δ, was investigated in different human HCC cell lines and in an immortalized human hepatic cell line. Additionally, subcutaneous models of HCC were established to evaluate the in vivo anti-tumor efficacy of G47Δ. Methods The HepG2, HepB, SMMC-7721, BEL-7404, and BEL-7405 human HCC cell lines and the HL-7702 human hepatic immortalized cell lines were infected with G47Δ at different multiplicities of infection (MOIs). The viability of infected cells was determined, and the G47Δ replication was identified by X-gal staining for LacZ expression. Two subcutaneous (s.c.) HCC tumor models of HCC were also established in Balb/c nude mice, which were intratumorally(i.t.) treated with either G47Δ or mock virus. Tumor volume and mouse survival times were documented. Results More than 95% of the HepG2, Hep3B,and SMMC-7721 HCC cells were killed on by day 5 after infection with a MOI’s of 0.01. For the HL-7702 human hepatic immortalized cells, 100% of the cells were killed on by day 5 after infection with a MOI’s of 0.01. The BEL-7404 HCC cell line was less susceptible with about 70% cells were killed by day 5 after infection with a MOI’s of 0.01. Whereas the BEL-7405 HCC cells were the least susceptible, with only 30% of the cells were killed. Both the SMMC-7721 and BEL-7404 cells form aggressive sc tumor models. G47Δ replicates in the tumors, such that most of the tumors regressed after the G47Δ-treatment, and treated tumor-bearing mice survived much longer than the control animals. Conclusions G47Δ effectively kills human HCC cells and an immortalized hepatic cell line at low MOI. Intra-tumor injection of G47Δ can induce a therapeutic effect and prolong the survival of treated mice bearing SMMC-7721 and BEL-7404 subcutaneously (s.c.) tumors. Thus, G47Δ may be useful as a novel therapeutic agent for HCC.
Collapse
Affiliation(s)
- Jiani Wang
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, 510630 Guangzhou, China
| | - Lihua Xu
- Department of Oncology and Hematology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weigen Zeng
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, 510630 Guangzhou, China ; Department of Colorectal Surgery, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, 100021 Beijing, China
| | - Pan Hu
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, 510630 Guangzhou, China
| | - Musheng Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Samuel D Rabkin
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Renbin Liu
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, 510630 Guangzhou, China
| |
Collapse
|
45
|
Ning J, Wakimoto H. Oncolytic herpes simplex virus-based strategies: toward a breakthrough in glioblastoma therapy. Front Microbiol 2014; 5:303. [PMID: 24999342 PMCID: PMC4064532 DOI: 10.3389/fmicb.2014.00303] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/03/2014] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viruses (OV) are a class of antitumor agents that selectively kill tumor cells while sparing normal cells. Oncolytic herpes simplex virus (oHSV) has been investigated in clinical trials for patients with the malignant brain tumor glioblastoma for more than a decade. These clinical studies have shown the safety of oHSV administration to the human brain, however, therapeutic efficacy of oHSV as a single treatment remains unsatisfactory. Factors that could hamper the anti-glioblastoma efficacy of oHSV include: attenuated potency of oHSV due to deletion or mutation of viral genes involved in virulence, restricting viral replication and spread within the tumor; suboptimal oHSV delivery associated with intratumoral injection; virus infection-induced inflammatory and cellular immune responses which could inhibit oHSV replication and promote its clearance; lack of effective incorporation of oHSV into standard-of-care, and poor knowledge about the ability of oHSV to target glioblastoma stem cells (GSCs). In an attempt to address these issues, recent research efforts have been directed at: (1) design of new engineered viruses to enhance potency, (2) better understanding of the role of the cellular immunity elicited by oHSV infection of tumors, (3) combinatorial strategies with different antitumor agents with a mechanistic rationale, (4) “armed” viruses expressing therapeutic transgenes, (5) use of GSC-derived models in oHSV evaluation, and (6) combinations of these. In this review, we will describe the current status of oHSV clinical trials for glioblastoma, and discuss recent research advances and future directions toward successful oHSV-based therapy of glioblastoma.
Collapse
Affiliation(s)
- Jianfang Ning
- Department of Neurosurgery, Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School Boston, MA, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School Boston, MA, USA
| |
Collapse
|
46
|
Permissiveness of human cancer cells to oncolytic bovine herpesvirus 1 is mediated in part by KRAS activity. J Virol 2014; 88:6885-95. [PMID: 24696490 DOI: 10.1128/jvi.00849-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED Oncolytic viruses (OVs) are attractive avenues of cancer therapy due to the absence of toxic side effects often seen with current treatment modalities. Bovine herpesvirus 1 (BHV-1) is a species-specific virus that does not induce cytotoxicity in normal primary human cells but can infect and kill various human immortalized and transformed cell lines. To gain a better understanding of the oncolytic breadth of BHV-1, the NCI panel of established human tumor cell lines was screened for sensitivity to the virus. Overall, 72% of the panel is permissive to BHV-1 infection, with corresponding decreases in cellular viability. This sensitivity is in comparison to a sensitivity of only 32% for a herpes simplex virus 1 (HSV-1)-based oncolytic vector. Strikingly, while 35% of the panel supports minimal or no BHV-1 replication, significant decreases in cellular viability still occur. These data suggest that BHV-1 is an OV with tropism for multiple tumor types and is able to induce cytotoxicity independent of significant virus replication. In contrast to other species-specific OVs, cellular sensitivity to BHV-1 does not correlate with type I interferon (IFN) signaling; however, mutations in KRAS were found to correlate with high levels of virus replication. The knockdown or overexpression of KRAS in human tumor cell lines yields modest changes in viral titers; however, overexpression of KRAS in normal primary cells elicits permissivity to BHV-1 infection. Together, these data suggest that BHV-1 is a broad-spectrum OV with a distinct mechanism of tumor targeting. IMPORTANCE Cancer remains a significant health issue, and novel treatments are required, particularly for tumors that are refractory to conventional therapies. Oncolytic viruses are a novel platform given their ability to specifically target tumor cells while leaving healthy cells intact. For this strategy to be successful, a fundamental understanding of virus-host interactions is required. We previously identified bovine herpesvirus 1 as a novel oncolytic virus with many unique and clinically relevant features. Here, we show that BHV-1 can target a wide range of human cancer types, most potently lung cancer. In addition, we show that enhanced KRAS activity, a hallmark of many cancers, is one of the factors that increases BHV-1 oncolytic capacity. These findings hold potential for future treatments, particularly in the context of lung cancer, where KRAS mutations are a negative predictor of treatment efficacy.
Collapse
|
47
|
Conrad SJ, Essani K. Oncoselectivity in Oncolytic Viruses against Colorectal Cancer. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/jct.2014.513118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Increased Resistance of Breast, Prostate, and Embryonic Carcinoma Cells against Herpes Simplex Virus in Three-Dimensional Cultures. ISRN ONCOLOGY 2013; 2013:104913. [PMID: 24455304 PMCID: PMC3885282 DOI: 10.1155/2013/104913] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 10/07/2013] [Indexed: 11/25/2022]
Abstract
In previous studies we found that uveal melanoma cells grown in extracellular matrix (ECM)-containing three-dimensional (3D) cultures have increased resistance against herpes simplex virus type 1 (HSV-1)-mediated destruction relative to cells cultured without ECM. Using additional tumor cell types including MB-231 human breast cancer cells, PC-3 human prostate cancer cells, and P19 mouse embryonal carcinoma cells, we show here that tumor cell lines other than melanoma are also more resistant to HSV-1-mediated destruction in 3D cultures than cells grown in 2D. We also demonstrate here that one mechanism responsible for the increased resistance of tumor cells to HSV-1 infection in 3D cultures is an ECM-mediated inhibition of virus replication following virus entry into cells. These findings confirm and extend previous observations related to the role of the ECM in tumor resistance against HSV-1 and may lead to improved strategies of oncolytic virotherapy.
Collapse
|
49
|
Toxicology and Biodistribution Studies for MGH2.1, an Oncolytic Virus that Expresses Two Prodrug-activating Genes, in Combination with Prodrugs. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e113. [PMID: 23922029 PMCID: PMC3759737 DOI: 10.1038/mtna.2013.38] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 06/01/2013] [Indexed: 01/06/2023]
Abstract
MGH2.1 is a herpes simplex virus type 1 (HSV1) oncolytic virus that expresses two prodrug-activating transgenes: the cyclophosphamide (CPA)-activating cytochrome P4502B1 (CYP2B1) and the CPT11-activating secreted human intestinal carboxylesterase (shiCE). Toxicology and biodistribution of MGH2.1 in the presence/absence of prodrugs was evaluated in mice. MGH2.1 ± prodrugs was cytotoxic to human glioma cells, but not to normal cells. Pharmacokinetically, intracranial MGH2.1 did not significantly alter the metabolism of intraperitoneally (i.p.) administered prodrugs in mouse plasma, brain, or liver. MGH2.1 did not induce an acute inflammatory reaction. MGH2.1 DNA was detected in brains of mice inoculated with 108 pfus for up to 60 days. However, only one animal showed evidence of viral gene expression at this time. Expression of virally encoded genes was restricted to brain. Intracranial inoculation of MGH2.1 did not induce lethality at 108 pfus in the absence of prodrugs and at 106 pfus in the presence of prodrugs. This study provides safety and toxicology data justifying a possible clinical trial of intratumoral injection of MGH2.1 with peripheral administration of CPA and/or CPT11 prodrugs in humans with malignant gliomas.
Collapse
|
50
|
Current status of gene therapy for brain tumors. Transl Res 2013; 161:339-54. [PMID: 23246627 PMCID: PMC3733107 DOI: 10.1016/j.trsl.2012.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/14/2012] [Accepted: 11/16/2012] [Indexed: 01/06/2023]
Abstract
Glioblastoma (GBM) is the most common and deadliest primary brain tumor in adults, with current treatments having limited impact on disease progression. Therefore the development of alternative treatment options is greatly needed. Gene therapy is a treatment strategy that relies on the delivery of genetic material, usually transgenes or viruses, into cells for therapeutic purposes, and has been applied to GBM with increasing promise. We have included selectively replication-competent oncolytic viruses within this strategy, although the virus acts directly as a complex biologic anti-tumor agent rather than as a classic gene delivery vehicle. GBM is a good candidate for gene therapy because tumors remain locally within the brain and only rarely metastasize to other tissues; the majority of cells in the brain are post-mitotic, which allows for specific targeting of dividing tumor cells; and tumors can often be accessed neurosurgically for administration of therapy. Delivery vehicles used for brain tumors include nonreplicating viral vectors, normal adult stem/progenitor cells, and oncolytic viruses. The therapeutic transgenes or viruses are typically cytotoxic or express prodrug activating suicide genes to kill glioma cells, immunostimulatory to induce or amplify anti-tumor immune responses, and/or modify the tumor microenvironment such as blocking angiogenesis. This review describes current preclinical and clinical gene therapy strategies for the treatment of glioma.
Collapse
|