1
|
Wienen F, Nilson R, Allmendinger E, Peters S, Barth TF, Kochanek S, Krutzke L. An oncolytic HAdV-5 with reduced surface charge combines diminished toxicity and improved tumor targeting. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200909. [PMID: 39758252 PMCID: PMC11699628 DOI: 10.1016/j.omton.2024.200909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/12/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025]
Abstract
Human adenovirus type 5 (HAdV-5)-based oncolytic viruses hold significant promise for anti-cancer therapy. However, poor tumor-targeting and off-target organ transduction after systemic administration limit their therapeutic efficacy. In addition, the strong liver tropism of HAdV-5-based vectors poses the risk of hepatotoxicity. By genetic modification of the major capsid protein hexon we generated a HAdV-5-based oncolytic vector (HAdV-5-HexPos3) with reduced negative surface charge. Coxsackie and adenovirus receptor (CAR) binding-ablated (ΔCAR) HAdV-5-HexPos3_ΔCAR exhibited superior and CAR-independent transduction of various cancer cell lines in vitro, further enhanced in the presence of HAdV-5 naive murine plasma. Upon intravenous administration into tumor-bearing immunodeficient NSG mice, replication-deficient HAdV-5-HexPos3_ΔCAR vector particles showed significantly reduced off-target organ tropism in all tissues analyzed, including the liver. Moreover, we detected a significantly increased intratumoral vector load for HAdV-5-HexPos3_ΔCAR, leading to a 29-fold elevated tumor-to-liver ratio compared with a control vector with unmodified hexon. Intravenous injection of a conditionally replicating hexon-unmodified control vector induced severe hepatotoxicity in tumor-bearing NSG mice, while a conditionally replicating HAdV-5-HexPos3_ΔCAR vector was well tolerated and resulted in intratumoral vector presence for up to 56 days. HAdV-5-HexPos3_ΔCAR represents a promising vector platform for the generation of HAdV-5-based oncolytic viruses with reduced systemic toxicity and improved therapeutic efficacy.
Collapse
Affiliation(s)
- Frederik Wienen
- Department of Gene Therapy, Ulm University, 89081 Ulm, Germany
| | - Robin Nilson
- Department of Gene Therapy, Ulm University, 89081 Ulm, Germany
| | | | - Sarah Peters
- Department of Clinical Chemistry, Ulm University Medical Center, 89081 Ulm, Germany
| | - Thomas F.E. Barth
- Institute of Pathology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Stefan Kochanek
- Department of Gene Therapy, Ulm University, 89081 Ulm, Germany
| | - Lea Krutzke
- Department of Gene Therapy, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
2
|
Mellid-Carballal R, Gutierrez-Gutierrez S, Rivas C, Garcia-Fuentes M. Viral protein nanoparticles (Part 1): Pharmaceutical characteristics. Eur J Pharm Sci 2023; 187:106460. [PMID: 37156338 DOI: 10.1016/j.ejps.2023.106460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/21/2023] [Accepted: 05/06/2023] [Indexed: 05/10/2023]
Abstract
Viral protein nanoparticles fill the gap between viruses and synthetic nanoparticles. Combining advantageous properties of both systems, they have revolutionized pharmaceutical research. Virus-like particles are characterized by a structure identical to viruses but lacking genetic material. Another type of viral protein nanoparticles, virosomes, are similar to liposomes but include viral spike proteins. Both systems are effective and safe vaccine candidates capable of overcoming the disadvantages of both traditional and subunit vaccines. Besides, their particulate structure, biocompatibility, and biodegradability make them good candidates as vectors for drug and gene delivery, and for diagnostic applications. In this review, we analyze viral protein nanoparticles from a pharmaceutical perspective and examine current research focused on their development process, from production to administration. Advances in synthesis, modification and formulation of viral protein nanoparticles are critical so that large-scale production of viral protein nanoparticle products becomes viable and affordable, which ultimately will increase their market penetration in the future. We will discuss their expression systems, modification strategies, formulation, biopharmaceutical properties, and biocompatibility.
Collapse
Affiliation(s)
- Rocio Mellid-Carballal
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Sara Gutierrez-Gutierrez
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Carmen Rivas
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain; Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CNB)-CSIC, Spain
| | - Marcos Garcia-Fuentes
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain.
| |
Collapse
|
3
|
Recent Developments in NSG and NRG Humanized Mouse Models for Their Use in Viral and Immune Research. Viruses 2023; 15:v15020478. [PMID: 36851692 PMCID: PMC9962986 DOI: 10.3390/v15020478] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Humanized mouse models have been widely used in virology, immunology, and oncology in the last decade. With advances in the generation of knockout mouse strains, it is now possible to generate animals in which human immune cells or human tissue can be engrafted. These models have been used for the study of human infectious diseases, cancers, and autoimmune diseases. In recent years, there has been an increase in the use of humanized mice to model human-specific viral infections. A human immune system in these models is crucial to understand the pathogenesis observed in human patients, which allows for better treatment design and vaccine development. Recent advances in our knowledge about viral pathogenicity and immune response using NSG and NRG mice are reviewed in this paper.
Collapse
|
4
|
Naumenko VA, Stepanenko AA, Lipatova AV, Vishnevskiy DA, Chekhonin VP. Infection of non-cancer cells: A barrier or support for oncolytic virotherapy? MOLECULAR THERAPY - ONCOLYTICS 2022; 24:663-682. [PMID: 35284629 PMCID: PMC8898763 DOI: 10.1016/j.omto.2022.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oncolytic viruses are designed to specifically target cancer cells, sparing normal cells. Although numerous studies demonstrate the ability of oncolytic viruses to infect a wide range of non-tumor cells, the significance of this phenomenon for cancer virotherapy is poorly understood. To fill the gap, we summarize the data on infection of non-cancer targets by oncolytic viruses with a special focus on tumor microenvironment and secondary lymphoid tissues. The review aims to address two major questions: how do attenuated viruses manage to infect normal cells, and whether it is of importance for oncolytic virotherapy.
Collapse
Affiliation(s)
- Victor A. Naumenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Corresponding author Victor A. Naumenko, PhD, V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia.
| | - Aleksei A. Stepanenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Anastasiia V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Daniil A. Vishnevskiy
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
| | - Vladimir P. Chekhonin
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| |
Collapse
|
5
|
Rai P, Chuong C, LeRoith T, Smyth JW, Panov J, Levi M, Kehn-Hall K, Duggal NK, Lucarelli JW. Adenovirus transduction to express human ACE2 causes obesity-specific morbidity in mice, impeding studies on the effect of host nutritional status on SARS-CoV-2 pathogenesis. Virology 2021; 563:98-106. [PMID: 34509029 PMCID: PMC8414371 DOI: 10.1016/j.virol.2021.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has paralyzed the global economy and resulted in millions of deaths globally. People with co-morbidities like obesity, diabetes and hypertension are at an increased risk for severe COVID-19 illness. This is of overwhelming concern because 42% of Americans are obese, 30% are pre-diabetic and 9.4% have clinical diabetes. Here, we investigated the effect of obesity on disease severity following SARS-CoV-2 infection using a well-established mouse model of diet-induced obesity. Diet-induced obese and lean control C57BL/6 N mice, transduced for ACE2 expression using replication-defective adenovirus, were infected with SARS-CoV-2, and monitored for lung pathology, viral titers, and cytokine expression. No significant differences in tissue pathology or viral replication was observed between AdV transduced lean and obese groups, infected with SARS-CoV-2, but certain cytokines were expressed more significantly in infected obese mice compared to the lean ones. Notably, significant weight loss was observed in obese mice treated with the adenovirus vector, independent of SARS-CoV-2 infection, suggesting an obesity-dependent morbidity induced by the vector. These data indicate that the adenovirus-transduced mouse model of SARS-CoV-2 infection, as described here and elsewhere, may be inappropriate for nutrition studies.
Collapse
Affiliation(s)
- Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Christina Chuong
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - James W Smyth
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA, USA; Department of Biological Sciences, College of Science, Virginia Polytechnic State Institute and State University, Blacksburg, VA, USA; Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Julia Panov
- Tauber Bioinformatics Research Center, Haifa 3498838, Israel; Sagol Department of Neurobiology, University of Haifa, Haifa 3498838, Israel
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Kylene Kehn-Hall
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Nisha K Duggal
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - James-Weger Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA.
| |
Collapse
|
6
|
Vupputuri S, Tayebi L, Hikkaduwa Koralege RS, Nigatu A, Mozafari M, Mishra A, Liu L, Ramsey JD. Polyethylene glycol–modified DOTAP:cholesterol/adenovirus hybrid vectors have improved transduction efficiency and reduced immunogenicity. JOURNAL OF NANOPARTICLE RESEARCH 2021; 23:37. [DOI: 10.1007/s11051-020-05134-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 12/26/2020] [Indexed: 03/07/2025]
|
7
|
Shiri A, Sarvari J, Firoozi Ghahestani S, Gholijani N, Tamaddon AM, Rastegari M, Moattari A, Hosseini SY. The Inflammatory and Fibrotic Patterns of Hepatic Stellate Cells Following Coagulation Factors (VII or X)-Shielded Adenovirus Infection. Curr Microbiol 2021; 78:718-726. [PMID: 33410956 DOI: 10.1007/s00284-020-02297-5] [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: 03/07/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022]
Abstract
The role of coagulation factors on the inflammatory effect of adenovirus (Ad) is an unresolved question that was considered herein. Adenovirus-36(Ad36) and adenovector-5-GFP(Ad5-GFP) were prepared; then, they were loaded with VII or FX factors. The size/charge parameters and transduction efficiency were evaluated using fluorescent microscopy and Zetasizer, respectively. The Ad36-coagulation factor complexes were added on the stellate cells, LX-2. Thereafter, the expression levels of inflammatory and fibrotic genes including PKR, IL-1β, TNF-α, TIMP-1, collagen, and TGF-β were measured by qPCR and ELISA assays. The loading of FVII or FX factors not only increased the size/charge of Ad5-GFP but also enhanced the transduction rate up to 60% and 75%, respectively, compared to the controls (45%). The PKR expression analysis showed an upregulation following treatment with all Ad36 forms (P = 0.0152). The IL-1β and TNF-α cytokines analyses demonstrated that the Ad36-FVII complex elicited the highest inflammatory response (P = 0.05). Similarly, the fibrosis-related expression analysis revealed a more inductive role of FVII when loaded on Ad36, compared to the FX factor. The findings suggested that adenovirus elicited the innate inflammatory and activation state in the hepatic stellate cell. In addition, adenovirus shielded by FVII exhibited more innate inflammation as well as activation of the stellate cells than the FX-loaded virus.
Collapse
Affiliation(s)
- Alireza Shiri
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jamal Sarvari
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,GastroenteroHepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Firoozi Ghahestani
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasser Gholijani
- Autoimmunity Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Pharmaceutics Department, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahroo Rastegari
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afagh Moattari
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
8
|
Xu D, Yao J, Zhang Y, Xiao N, Peng P, Li Z, Pan Z, Yao Z. The Effect of PEI-Mediated E1A on the Radiosensitivity of Hepatic Carcinoma Cells. Asian Pac J Cancer Prev 2020; 21:911-917. [PMID: 32334450 PMCID: PMC7445989 DOI: 10.31557/apjcp.2020.21.4.911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Indexed: 11/26/2022] Open
Abstract
Objective: The study was undertaken to investigate the effects of polyethyleneimine (PEI)-mediated adenovirus 5 early region 1A (E1A) on radiosensitivity of human hepatic carcinoma cell in vitro and to disclosure the underlying mechanism. Materials and Methods: Human hepatic carcinoma SMMC-7721 cell line was transfected with E1A gene using PEI vector. Untransfected cells (SMMC-7721 group), cells transfected with blank-vector (SMMC-7721-vect group), and cells transfected with E1A gene (SMMC-7721-E1A group) were treated with 6 MV X-ray irradiation at doses of 0, 1, 2, 4, 8 and Gy, respectively. Radiosensitivity was determined by MTT assay and quantified by calculating the cell survival rate. Cell-cycle distribution and apotosis rate were monitored by flow cytometry. Results: The survival rate of SMMC-7721-E1A was significantly lower than that of SMMC-7721 cell. Apoptosis rate of SMMC-7721-E1A group was significantly higher than that of SMMC-7721group (P<0.01).The ratio of S stage in cell cycle of SMMC-7721-E1A was significantly lower than that in SMMC-7721 cell. The ratio of G2/M stage in cell cycle of SMMC-7721-E1A was significantly higher than that in SMMC-7721 cell (P<0.01). Conclusion: PEI could transfect E1A gene into hepatic carcinoma cells PEI-mediated E1A could effectively enhance radiosensitivity of hepatic carcinoma cells which may be related to its effects on apoptosis promoting leading to S phase suppression and G2/M phase arrest.
Collapse
Affiliation(s)
- Danghui Xu
- Department of Radiology,Affiliated Hospital of Nanjing University of Chinese Medicine,Jiangsu Provincial Hospital of Traditional Chinese Medicine,Nanjing, Jiangsu Province ,China
| | - Jianxin Yao
- Department of Medical Imaging, Nanjing Vocational Health College, Nanjing, Jiangsu Province, China
| | - Yiwen Zhang
- Department of Nursing, The Affiliated Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Nan Xiao
- Department of Medical Imaging, Nanjing Vocational Health College, Nanjing, Jiangsu Province, China
| | - Peng Peng
- Department of Nursing, Nanjing Health College of Jiangsu Union Technical Institute, Nanjing, Jiangsu Province, China
| | - Zhanfeng Li
- Department of Medical Imaging, Nanjing Vocational Health College, Nanjing, Jiangsu Province, China
| | - Zhiyao Pan
- Department of Basic Medical Science, Zhejiang University Medical College, Hangzhou, Zhejiang Province, China
| | - Zhifeng Yao
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China.,Department of Radiotherapy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
9
|
Machitani M, Sakurai F, Wakabayashi K, Nakatani K, Tachibana M, Kato N, Fujiwara T, Mizuguchi H. Suppression of Oncolytic Adenovirus-Mediated Hepatotoxicity by Liver-Specific Inhibition of NF-κB. MOLECULAR THERAPY-ONCOLYTICS 2017; 7:76-85. [PMID: 29202008 PMCID: PMC5704103 DOI: 10.1016/j.omto.2017.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/22/2017] [Indexed: 11/28/2022]
Abstract
Telomerase-specific replication-competent adenoviruses (Ads), i.e., TRADs, which possess an E1 gene expression cassette driven by the human telomerase reverse transcriptase promoter, are promising agents for cancer treatment. However, even though oncolytic Ads, including TRAD, are intratumorally administered, they are disseminated from the tumor to systemic circulation, causing concern about oncolytic Ad-mediated hepatotoxicity (due mainly to leaky expression of Ad genes in liver). We reported that inhibition of nuclear factor-κB (NF-κB) leads to the suppression of replication-incompetent Ad vector-mediated hepatotoxicity via reduction of the leaky expression of Ad genes in liver. Here, to develop a TRAD with an improved safety profile, we designed a TRAD that carries a liver-specific promoter-driven dominant-negative IκBα (DNIκBα) expression cassette (TRAD-DNIκBα). Compared with a conventional TRAD, TRAD-DNIκBα showed hepatocyte-specific inhibition of NF-κB signaling and significantly reduced Ad gene expression and replication in the normal human hepatocyte cell line. TRAD-induced hepatotoxicity was largely suppressed in mice following intravenous administration of TRAD-DNIκBα. However, the replication profiles and oncolytic activities of TRAD-DNIκBα were comparable with those of the conventional TRAD in human non-hepatic tumor cells. These results indicate that oncolytic Ads containing the liver-specific DNIκBα expression cassette have improved safety profiles without inhibiting oncolytic activities.
Collapse
Affiliation(s)
- Mitsuhiro Machitani
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Laboratory of Regulatory Sciences for Oligonucleotide Therapeutics, Clinical Drug Development Unit, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keisaku Wakabayashi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kosuke Nakatani
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masashi Tachibana
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Nobuyuki Kato
- Department of Tumor Virology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.,Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito, Asagi, Ibaraki, Osaka 567-0085, Japan.,iPS Cell-Based Research Project on Hepatic Toxicity and Metabolism, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
10
|
Zhang L, Steele MB, Jenks N, Grell J, Suksanpaisan L, Naik S, Federspiel MJ, Lacy MQ, Russell SJ, Peng KW. Safety Studies in Tumor and Non-Tumor-Bearing Mice in Support of Clinical Trials Using Oncolytic VSV-IFNβ-NIS. HUM GENE THER CL DEV 2017; 27:111-22. [PMID: 27532609 DOI: 10.1089/humc.2016.061] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Oncolytic VSV-IFNβ-NIS is selectively destructive to tumors. Here, we present the IND enabling preclinical rodent studies in support of clinical testing of vesicular stomatitis virus (VSV) as a systemic therapy. Efficacy studies showed dose-dependent tumor regression in C57BL/KaLwRij mice bearing syngeneic 5TGM1 plasmacytomas after systemic VSV administration. In contrast, the virus was effective at all doses tested against human KAS6/1 xenografts in SCID mice. Intravenous administration of VSV-mIFNβ-NIS is well tolerated in C57BL/6 mice up to 5 × 10(10) TCID50 (50% tissue culture infective dose)/kg with no neurovirulence, no cytokine storm, and no abnormalities in tissues. Dose-limiting toxicities included elevated transaminases, thrombocytopenia, and lymphopenia. Inactivated viral particles did not cause hepatic toxicity. Intravenously administered VSV was preferentially sequestered by macrophages in the spleen and liver. Quantitative RT-PCR analysis for total viral RNA on days 2, 7, 21, and 58 showed highest VSV RNA in day 2 samples; highest in spleen, liver, lung, lymph node, kidney, gonad, and bone marrow. No infectious virus was recovered from tissues at any time point. The no observable adverse event level and maximum tolerated dose of VSV-mIFNβ-NIS in C57BL/6 mice are 10(10) TCID50/kg and 5 × 10(10) TCID50/kg, respectively. Clinical translation of VSV-IFNβ-NIS is underway in companion dogs with cancer and in human patients with relapsed hematological malignancies and endometrial cancer.
Collapse
Affiliation(s)
- Lianwen Zhang
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota
| | - Michael B Steele
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,2 Toxicology and Pharmacology Laboratory, Mayo Clinic , Rochester, Minnesota
| | - Nathan Jenks
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,2 Toxicology and Pharmacology Laboratory, Mayo Clinic , Rochester, Minnesota
| | - Jacquelyn Grell
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,2 Toxicology and Pharmacology Laboratory, Mayo Clinic , Rochester, Minnesota
| | | | - Shruthi Naik
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota
| | - Mark J Federspiel
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,4 Viral Vector Production Laboratory, Mayo Clinic , Rochester, Minnesota
| | - Martha Q Lacy
- 5 Division of Hematology, Mayo Clinic , Rochester, Minnesota
| | - Stephen J Russell
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,5 Division of Hematology, Mayo Clinic , Rochester, Minnesota
| | - Kah-Whye Peng
- 1 Department of Molecular Medicine, Mayo Clinic , Rochester, Minnesota.,2 Toxicology and Pharmacology Laboratory, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
11
|
Liu Q, Zhou S, Fan C, Huang W, Li Q, Liu S, Wu X, Li B, Wang Y. Biodistribution and residence time of adenovector serotype 5 in normal and immunodeficient mice and rats detected with bioluminescent imaging. Sci Rep 2017; 7:3597. [PMID: 28620164 PMCID: PMC5472566 DOI: 10.1038/s41598-017-03852-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 05/08/2017] [Indexed: 12/03/2022] Open
Abstract
As concerns increase about adenovirus type 5 (Ad5) being a safe gene transfer vector, it is important to evaluate its distribution, residence time, and possible toxicity in immunodeficient populations. To characterize the potential risk associated with different Ad5 vector delivery modes, we used immunocompetent and immunodeficient Rag2 -/- animals to establish mouse and rat models that could be monitored with bioluminescent imaging following intramuscular or intravascular infection with an engineered replication-incompetent Ad5 virus carrying the firefly luciferase gene (Ad5-Fluc). The Ad5 vector was less well-tolerated by Rag2 -/- animals than by wildtype ones, with delayed residence time, wider virus dissemination, less weight gain, and relatively severe pathological changes. In intravascularly Ad5-Fluc-infected Rag2 -/- mice, systemic virus dissemination extended from the abdomen to the limbs and head on day 9 post-infection. Additionally, significant increases in plasma TNF-α and IFN-γ, which may be important factors in the heightened immunopathology in the liver and brain, were detected in the Rag2 -/- mice 30 days after intravascular delivery. The Ad5 vector was better tolerated after intramuscular delivery than after intravascular delivery. Ad5-Fluc/Rag2 -/- mice and rats can be used as reliable models of an immunodeficient population in which to evaluate the safety of Ad5-vectored vaccines or gene therapy products.
Collapse
Affiliation(s)
- Qiang Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Shuya Zhou
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Changfa Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Qianqian Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Susu Liu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Xi Wu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Baowen Li
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, 100050, China.
| |
Collapse
|
12
|
Fajardo CA, Guedan S, Rojas LA, Moreno R, Arias-Badia M, de Sostoa J, June CH, Alemany R. Oncolytic Adenoviral Delivery of an EGFR-Targeting T-cell Engager Improves Antitumor Efficacy. Cancer Res 2017; 77:2052-2063. [PMID: 28143835 DOI: 10.1158/0008-5472.can-16-1708] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/11/2017] [Accepted: 01/24/2017] [Indexed: 11/16/2022]
Abstract
Antiviral immune responses present a major hurdle to the efficacious use of oncolytic adenoviruses as cancer treatments. Despite the existence of a highly immunosuppressive tumor environment, adenovirus-infected cells can nonetheless be efficiently cleared by infiltrating cytotoxic T lymphocytes (CTL) without compromising tumor burden. In this study, we tested the hypothesis that tumor-infiltrating T cells could be more effectively activated and redirected by oncolytic adenoviruses that were armed with bispecific T-cell-engager (BiTE) antibodies. The oncolytic adenovirus ICOVIR-15K was engineered to express an EGFR-targeting BiTE (cBiTE) antibody under the control of the major late promoter, leading to generation of ICOVIR-15K-cBiTE, which retained its oncolytic properties in vitro cBiTE expression and secretion was detected in supernatants from ICOVIR-15K-cBiTE-infected cells, and the secreted BiTEs bound specifically to both CD3+ and EGFR+ cells. In cell coculture assays, ICOVIR-15K-cBiTE-mediated oncolysis resulted in robust T-cell activation, proliferation, and bystander cell-mediated cytotoxicity. Notably, intratumoral injection of this cBiTE-expressing adenovirus increased the persistence and accumulation of tumor-infiltrating T cells in vivo, compared with the parental virus lacking such effects. Moreover, in two distinct tumor xenograft models, combined delivery of ICOVIR-15K-cBiTE with peripheral blood mononuclear cells or T cells enhanced the antitumor efficacy achieved by the parental counterpart. Overall, our results show how arming oncolytic adenoviruses with BiTE can overcome key limitations in oncolytic virotherapy. Cancer Res; 77(8); 2052-63. ©2017 AACR.
Collapse
Affiliation(s)
| | - Sonia Guedan
- Abramson Cancer Center and the Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Luis Alfonso Rojas
- ProCure Program, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Spain
| | - Rafael Moreno
- ProCure Program, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Spain
| | - Marcel Arias-Badia
- ProCure Program, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Spain
| | - Jana de Sostoa
- ProCure Program, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Spain
| | - Carl H June
- Abramson Cancer Center and the Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ramon Alemany
- ProCure Program, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
13
|
Rodríguez E, Ip WH, Kolbe V, Hartmann K, Pilnitz-Stolze G, Tekin N, Gómez-Medina S, Muñoz-Fontela C, Krasemann S, Dobner T. Humanized Mice Reproduce Acute and Persistent Human Adenovirus Infection. J Infect Dis 2016; 215:70-79. [PMID: 28077585 DOI: 10.1093/infdis/jiw499] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/13/2016] [Indexed: 11/12/2022] Open
Abstract
Severe human adenovirus (HAdV) infections are an increasing threat for immunosuppressed individuals, particularly those who have received stem cell transplants. It has been previously hypothesized that severe infections might be due to reactivation of a persistent infection, but this hypothesis has been difficult to test owing to the lack of a permissive in vivo model of HAdV infection. Here we established a humanized mouse model that reproduces features of acute and persistent HAdV infection. In this model, acute infection correlated with high mortality, weight loss, liver pathology, and expression of viral proteins in several organs. In contrast, persistent infection was asymptomatic and led to establishment of HAdV-specific adaptive immunity and expression of early viral genes exclusively in the bone marrow. These findings validate the use of humanized mice to study acute and persistent HAdV infection and strongly suggest the presence of cellular reservoirs in the bone marrow.
Collapse
Affiliation(s)
- Estefanía Rodríguez
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg.,German Center for Infection Research, Braunschweig, (DZIF), Partner Site Hamburg, Germany
| | - Wing Hang Ip
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg.,German Center for Infection Research, Braunschweig, (DZIF), Partner Site Hamburg, Germany
| | - Viktoria Kolbe
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg.,German Center for Infection Research, Braunschweig, (DZIF), Partner Site Hamburg, Germany
| | - Kristin Hartmann
- Institute for Neuropathology, University Medical Center Hamburg-Eppendorf
| | - Gundula Pilnitz-Stolze
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg.,German Center for Infection Research, Braunschweig, (DZIF), Partner Site Hamburg, Germany
| | - Nilgün Tekin
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg.,German Center for Infection Research, Braunschweig, (DZIF), Partner Site Hamburg, Germany
| | - Sergio Gómez-Medina
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg.,German Center for Infection Research, Braunschweig, (DZIF), Partner Site Hamburg, Germany
| | - César Muñoz-Fontela
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg.,German Center for Infection Research, Braunschweig, (DZIF), Partner Site Hamburg, Germany
| | - Susanne Krasemann
- Institute for Neuropathology, University Medical Center Hamburg-Eppendorf
| | - Thomas Dobner
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg.,German Center for Infection Research, Braunschweig, (DZIF), Partner Site Hamburg, Germany
| |
Collapse
|
14
|
Bofill-De Ros X, Villanueva E, Fillat C. Late-phase miRNA-controlled oncolytic adenovirus for selective killing of cancer cells. Oncotarget 2016; 6:6179-90. [PMID: 25714032 PMCID: PMC4467430 DOI: 10.18632/oncotarget.3350] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/12/2015] [Indexed: 01/13/2023] Open
Abstract
Tissue-specific detargeting by miRNAs has been demonstrated to be a potent strategy to restrict adenoviral replication to cancer cells. These studies have generated adenoviruses with miRNA target sites placed in the 3′UTR of early gene products. In this work, we have studied the feasibility of providing tissue-specific selectivity to replication-competent adenoviruses through the regulation of the late structural protein fiber (L5 gene). We have engineered a 3′UTR containing eight miR-148a binding sites downstream the L5 coding sequence (Ad-L5-8miR148aT). We present in vitro and in vivo evidences of Ad-L5-8miR148aT miRNA-dependent regulation. In vitro data show that at 72 hours post-infection miR-148a-regulation impaired fiber expression leading to a 70% reduction of viral release. The application of seven consecutive rounds of infection in miR-148a cells resulted in 10.000-fold reduction of viral genomes released. In vivo, liver production of infective viral particles was highly impaired, similarly to that triggered by an adenovirus with miRNA target sites regulating the early E1A gene. Noticeably, mice treated with Ad-L5-8miR148aT showed an attenuation of adenoviral-induced hepatotoxicity but retained full lytic activity in cancer cells and exhibited robust antitumoral responses in patient-derived xenografts. Thus, miRNA-control of late proteins constitutes a novel strategy to provide selectivity to adenoviruses.
Collapse
Affiliation(s)
- Xavier Bofill-De Ros
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Eneko Villanueva
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Cristina Fillat
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| |
Collapse
|
15
|
Browne A, Tookman LA, Ingemarsdotter CK, Bouwman RD, Pirlo K, Wang Y, McNeish IA, Lockley M. Pharmacological Inhibition of β3 Integrin Reduces the Inflammatory Toxicities Caused by Oncolytic Adenovirus without Compromising Anticancer Activity. Cancer Res 2015; 75:2811-21. [PMID: 25977332 DOI: 10.1158/0008-5472.can-14-3761] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 04/07/2015] [Indexed: 01/28/2023]
Abstract
Adenoviruses have been clinically tested as anticancer therapies but their utility has been severely limited by rapid, systemic cytokine release and consequent inflammatory toxicity. Here, we describe a new approach to tackling these dangerous side effects. Using human ovarian cancer cell lines as well as malignant epithelial cells harvested from the ascites of women with ovarian cancer, we show that tumor cells do not produce cytokines in the first 24 hours following in vitro infection with the oncolytic adenovirus dl922-947. In contrast, dl922-947 does induce inflammatory cytokines at early time points following intraperitoneal delivery in mice with human ovarian cancer intraperitoneal xenografts. In these animals, cytokines originate predominantly in murine tissues, especially in macrophage-rich organs such as the spleen. We use a nonreplicating adenovirus to confirm that early cytokine production is independent of adenoviral replication. Using β3 integrin knockout mice injected intraperitoneally with dl922-947 and β3 null murine peritoneal macrophages, we confirm a role for macrophage cell surface β3 integrin in this dl922-947-induced inflammation. We present new evidence that co-administration of a cyclic RGD-mimetic-specific inhibitor of β3 integrin significantly attenuates the cytokine release and inflammatory hepatic toxicity induced by dl922-947 in an intraperitoneal murine model of ovarian cancer. Importantly, we find no evidence that β3 inhibition compromises viral infectivity and oncolysis in vitro or anticancer efficacy in vivo. By enabling safe, systemic delivery of replicating adenoviruses, this novel approach could have a major impact on the future development of these effective anticancer agents.
Collapse
Affiliation(s)
- Ashley Browne
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Laura A Tookman
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Carin K Ingemarsdotter
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Russell D Bouwman
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Katrina Pirlo
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Yaohe Wang
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Iain A McNeish
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom. Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Michelle Lockley
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom.
| |
Collapse
|
16
|
Xu W, Zhang Z, Yang Y, Hu Z, Wang CH, Morgan M, Wu Y, Hutten R, Xiao X, Stock S, Guise T, Prabhakar BS, Brendler C, Seth P. Ad5/48 hexon oncolytic virus expressing sTGFβRIIFc produces reduced hepatic and systemic toxicities and inhibits prostate cancer bone metastases. Mol Ther 2014; 22:1504-1517. [PMID: 24791939 DOI: 10.1038/mt.2014.80] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 04/24/2014] [Indexed: 01/17/2023] Open
Abstract
We are interested in developing oncolytic adenoviruses for the treatment of prostate cancer (PCa) bone metastases. A key limitation of Adenovirus 5 (Ad5) is that upon systemic administration, it produces major liver and systemic toxicities. To address this issue, a chimaeric Ad5/48 adenovirus mHAd.sTβRFc was created. Seven hypervariable regions of Ad5 hexon present in Ad5-based Ad.sTβRFc expressing soluble transforming growth factor beta receptor II-Fc fusion protein (sTGβRIIFc), were replaced by those of Ad48. mHAd.sTβRFc, like Ad.sTβRFc, was replication competent in the human PCa cells, and produced high levels of sTGβRIIFc expression. Compared to Ad.sTβRFc, the systemic delivery of mHAd.sTβRFc in nude mice resulted in much reduced systemic toxicity, and reduced liver sequestration. Ad.sTβRFc produced significant liver necrosis, and increases in alanine transaminase, aspartate transaminase, lactate dehydrogenase, tumor necrosis factor-α, and interleukin-6 levels, while mHAd.sTβRFc produced much reduced responses of these markers. Intravenous delivery of Ad.sTβRFc or mHAd.sTβRFc (5 × 10(10) viral particles/mouse) in nude mice bearing PC-3-luc PCa bone metastases produced inhibition of bone metastases. Moreover, a larger dose of the mHAd.sTβRFc (4 × 10(11) viral particles /mouse) was also effective in inhibiting bone metastases. Thus, mHAd.sTβRFc could be developed for the treatment of PCa bone metastases.
Collapse
Affiliation(s)
- Weidong Xu
- Gene Therapy Program, Department of Medicine, NorthShore Research Institute, Evanston, Illinois, USA
| | - Zhenwei Zhang
- Gene Therapy Program, Department of Medicine, NorthShore Research Institute, Evanston, Illinois, USA
| | - Yuefeng Yang
- Gene Therapy Program, Department of Medicine, NorthShore Research Institute, Evanston, Illinois, USA
| | - Zebin Hu
- Gene Therapy Program, Department of Medicine, NorthShore Research Institute, Evanston, Illinois, USA
| | - Chi-Hsiung Wang
- Gene Therapy Program, Department of Surgery, NorthShore Research Institute, Evanston, Illinois, USA
| | - Melanie Morgan
- Gene Therapy Program, Department of Pathology, NorthShore Research Institute, Evanston, Illinois, USA
| | - Ying Wu
- Image Processing Lab, Center for Advanced Imaging, Department of Radiology, NorthShore Research Institute, Evanston, Illinois, USA
| | - Ryan Hutten
- Image Processing Lab, Center for Advanced Imaging, Department of Radiology, NorthShore Research Institute, Evanston, Illinois, USA
| | - Xianghui Xiao
- Advanced Photon Source, Argonne National Laboratory, Argonne, Illinois, USA
| | - Stuart Stock
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, Chicago, Illinois, USA
| | - Theresa Guise
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois, Chicago, Illinois, USA
| | - Charles Brendler
- Gene Therapy Program, Department of Surgery, NorthShore Research Institute, Evanston, Illinois, USA
| | - Prem Seth
- Gene Therapy Program, Department of Medicine, NorthShore Research Institute, Evanston, Illinois, USA.
| |
Collapse
|
17
|
Hamada K, Shirakawa T, Terao S, Gotoh A, Tani K, Huang W. Biosafety studies of carrier cells infected with a replication-competent adenovirus introduced by IAI.3B promoter. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14019. [PMID: 26015963 PMCID: PMC4362384 DOI: 10.1038/mtm.2014.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/15/2014] [Indexed: 01/03/2023]
Abstract
The use of carrier cells infected with oncolytic viruses in cancer gene therapy is an attractive method because it can overcome viral immunogenicity and induce tumor immunity and significant antitumor activity. To enable human clinical trials of this treatment, acute and chronic toxicity tests must first be performed to ensure safety. IAI.3B promoter, oncolytic adenovirus AdE3-IAI.3B introduced by IAI.3B promoter, and A549 carrier cells infected with AdE3-IAI.3B were highly active in cancer cells but not in normal cells. Freeze-thawing increased the antitumor effect of A549 carrier cells by promoting the translocation of oncolytic adenovirus particles from the nucleus to the cytoplasm following the rupture of the nuclear membranes. No deaths or abnormal blood test data resulted from acute toxicity tests conducted in nude mice after a single dose. In chronic toxicity tests in rabbits, there were no serious side effects after eight doses of 1.25 × 107 cells/kg or less for 4 weeks; a significant immune response is known to elicit increased numbers of antiadenovirus antibodies and enlarge the spleen. From these results, it could be concluded that cancer gene therapy of recurrent solid tumors using carrier cells can be safely trialed in humans.
Collapse
Affiliation(s)
- Katsuyuki Hamada
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University , Shitsukawa, Toon, Ehime Japan
| | - Toshiro Shirakawa
- Division of Infectious Disease Control, International Center for Medical Research and Treatment, Faculty of Medicine, Kobe University Graduate School of Medicine , Kusunoki, Chuo-ku, Kobe, Hyogo, Japan
| | - Shuji Terao
- Laboratory of Cell and Gene Therapy, Institute of Advanced Medical Sciences, Hyogo College of Medicine , Mukogawa, Nishinomiya, Hyogo, Japan
| | - Akinobu Gotoh
- Laboratory of Cell and Gene Therapy, Institute of Advanced Medical Sciences, Hyogo College of Medicine , Mukogawa, Nishinomiya, Hyogo, Japan
| | - Kenzaburo Tani
- Department of Advanced Molecular and Cell Therapy, Kyushu University Hospital, Kyushu University , Maidashi, Higashi-ku, Fukuoka, Japan
| | - Wenlin Huang
- Department of Molecular Virology, Cancer Center, Sun Yat-sen University , Guangdong, China
| |
Collapse
|
18
|
José A, Sobrevals L, Miguel Camacho-Sánchez J, Huch M, Andreu N, Ayuso E, Navarro P, Alemany R, Fillat C. Intraductal delivery of adenoviruses targets pancreatic tumors in transgenic Ela-myc mice and orthotopic xenografts. Oncotarget 2013; 4:94-105. [PMID: 23328228 PMCID: PMC3702210 DOI: 10.18632/oncotarget.795] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Gene-based anticancer therapies delivered by adenoviruses are limited by the poor viral distribution into the tumor. In the current work we have explored the feasibility of targeting pancreatic tumors through a loco-regional route. We have taken advantage of the ductal network in the pancreas to retrogradelly inject adenoviruses through the common bile duct in two different mouse models of pancreatic carcinogenesis: The transgenic Ela-myc mice that develop mixed neoplasms displaying both acinar-like and duct-like neoplastic cells affecting the whole pancreas; and mice bearing PANC-1 and BxPC-3 orthotopic xenografts that constitute a model of localized human neoplastic tumors. We studied tumor targeting and the anticancer effects of newly thymidine kinase-engineered adenoviruses both in vitro and in vivo, and conducted comparative studies between intraductal or intravenous administration. Our data indicate that the intraductal delivery of adenovirus efficiently targets pancreatic tumors in the two mouse models. The in vivo application of AduPARTKT plus ganciclovir (GCV) treatment induced tumor regression in Ela-myc mice. Moreover, the intraductal injection of ICOVIR15-TKT oncolytic adenoviruses significantly improved mean survival of mice bearing PANC-1 and BxPC-3 pancreatic xenografts from 30 to 52 days and from 20 to 68 days respectively (p less than 0.0001) when combined with GCV. Of notice, both AduPARTKT and ICOVIR15-TKT antitumoral responses were stronger by ductal viral application than intravenously, in line with the 38-fold increase in pancreas transduction observed upon ductal administration. In summary our data show that cytotoxic adenoviruses retrogradelly injected to the pancreas can be a feasible approach to treat localized pancreatic tumors.
Collapse
Affiliation(s)
- Anabel José
- Institut d'Investigacions Biomèdiques August Pi i Sunyer IDIBAPS, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Wang F, Wang Z, Tian H, Qi M, Zhai Z, Li S, Li R, Zhang H, Wang W, Fu S, Lu J, Rodriguez R, Guo Y, Zhou L. Biodistribution and safety assessment of bladder cancer specific recombinant oncolytic adenovirus in subcutaneous xenografts tumor model in nude mice. Curr Gene Ther 2012; 12:67-76. [PMID: 22384806 DOI: 10.2174/156652312800099599] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 01/27/2012] [Accepted: 01/27/2012] [Indexed: 11/22/2022]
Abstract
BACKGROUND The previous works about safety evaluation for constructed bladder tissue specific adenovirus are poorly documented. Thus, we investigated the biodistribution and body toxicity of bladder specific oncolytic adenovirus Ad-PSCAE-UPII-E1A (APU-E1A) and Ad-PSCAE-UPII-E1A-AR (APU-E1A-AR), providing meaningful information prior to embarking on human clinical trials. MATERIALS AND METHOD Conditionally replicate recombinant adenovirus (CRADs) APU-E1A, APU-EIA-AR were constructed with bladder tissue specific UroplakinII(UPII) promoter to induce the expression of Ad5E1A gene and E1A-AR fusing gene, and PSCAE was inserted at upstream of promoter to enhance the function of promoter. Based on the cytopathic and anti-tumor effect of bladder cancer, these CRADs were intratumorally injected into subcutaneous xenografts tumor in nude mice. We then determined the toxicity through general health and behavioral assessment, hepatic and hematological toxicity evaluation, macroscopic and microscopic postmortem analyses. The spread of the transgene E1A of adenovirus was detected with RT-PCR and Western blot. Virus replication and distribution were examined with APU-LUC administration and Luciferase Assay. RESULTS General assessment and body weight of the animals did not reveal any alteration in general behavior. The hematological alterations of groups which were injected with 5x10(8) pfu or higher dose (5x10(9) pfu) of APU-E1A and APU-E1A-AR showed no difference in comparison with PBS group, and only slight increased transaminases in contrast to PBS group at 5x10(9) pfu of APU-E1A and APU-E1A-AR were observed. E1A transgene did not disseminate to organs outside of xenograft tumor. Virus replication was not detected in other organs beside tumor according to Luciferase Assay. CONCLUSIONS Our study showed that recombinant adenovirus APU-E1A-AR and APU-E1A appear safe with 5x10(7) pfu and 5x10(8) pfu intratumorally injection in mice, without any discernable effects on general health and behavior.
Collapse
Affiliation(s)
- Fang Wang
- School of Life Sciences in Lanzhou University, Lanzhou, Gansu Province, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Oncolytic adenovirus expressing soluble TGFβ receptor II-Fc-mediated inhibition of established bone metastases: a safe and effective systemic therapeutic approach for breast cancer. Mol Ther 2011; 19:1609-18. [PMID: 21712815 DOI: 10.1038/mt.2011.114] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In recent years, oncolytic adenoviruses have shown some promise as a novel class of antitumor agents. However, their utility in targeting bone metastases is relatively less studied. We have examined whether the systemic therapy of oncolytic adenoviruses expressing the soluble form of transforming growth factor-β (TGFβ) receptor II fused with human immunoglobulin G1 can be developed for the treatment of established breast cancer bone metastases. MDA-MB-231-luc2 human breast cancer cells were injected in the left heart ventricle of nude mice to establish bone metastasis. Mice with hind limb tumors were administered (on days 8 and 11) oncolytic adenoviruses-Ad.sTβRFc or mhTERTAd.sTβRFc. Skeletal tumor growth was monitored weekly by bioluminescence imaging (BLI) and radiography. At the termination time on day 28, hind limb bones were analyzed for tumor burden, synchrotron micro-computed tomography, and osteoclast activation. Intravenous delivery of Ad.sTβRFc and mhTERTAd.sTβRFc induced significant inhibition of tumor growth, reduction of tumor burden, osteoclast activation, and increased animals' survival. Oncolytic adenoviruses were safer than dl309, a wild-type virus. A slight elevation of liver enzyme activity was observed after Ad.sTβRFc administration; this subsided with time. Based on these studies, we believe that Ad.sTβRFc and mhTERTAd.sTβRFc can be developed as a safe and effective approach for the treatment of established bone metastasis.
Collapse
|
21
|
Salako MA, Kulbe H, Ingemarsdotter CK, Pirlo KJ, Williams SL, Lockley M, Balkwill FR, McNeish IA. Inhibition of the inflammatory cytokine TNF-α increases adenovirus activity in ovarian cancer via modulation of cIAP1/2 expression. Mol Ther 2011; 19:490-9. [PMID: 21081903 PMCID: PMC3048177 DOI: 10.1038/mt.2010.247] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 10/18/2010] [Indexed: 01/08/2023] Open
Abstract
Oncolytic adenoviruses show promise as a cancer treatment. However, they generate acute inflammatory responses with production of cytokines, including tumor necrosis factor-α (TNF-α). We investigated whether inhibition of TNF-α augments efficacy of the E1A CR2-deleted adenovirus dl922-947 in ovarian cancer. dl922-947 induced transcription of TNF-α and its downstream signaling targets interleukin-6 and -8 (IL-6 and IL-8) in ovarian cancer cells. In vitro, RNAi-mediated knockdown of TNF-α reduced production of multiple inflammatory cytokines after infection and increased ovarian cancer cell sensitivity to virus cytotoxicity, as did treatment with the anti-TNF-α antibody infliximab. In vivo, stable knockdown of TNF-α in IGROV-1 xenografts increased the anticancer activity of dl922-947. In addition, inhibition of TNF-α using monoclonal antibodies also improved dl922-947 efficacy. This increased efficacy resulted from suppression of cellular inhibitor of apoptosis-1 and -2 (cIAP1 and cIAP2) transcription in malignant cells and a consequent increase in caspase-mediated apoptosis. These findings suggest that TNF-α acts as a survival factor in adenovirus-infected cells. Combining TNF-α inhibition with oncolytic adenoviruses could improve antitumor activity in clinical trials.
Collapse
Affiliation(s)
- Michael A Salako
- Centres for Molecular Oncology and Imaging, Institute of Cancer, Barts and The London School of Medicine and Dentistry, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Gregory SM, Nazir SA, Metcalf JP. Implications of the innate immune response to adenovirus and adenoviral vectors. Future Virol 2011; 6:357-374. [PMID: 21738557 PMCID: PMC3129286 DOI: 10.2217/fvl.11.6] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adenovirus (AdV) is a common cause of respiratory illness in both children and adults. Respiratory symptoms can range from those of the common cold to severe pneumonia. Infection can also cause significant disease in the immunocompromised and among immunocompetent subjects in close quarters. Fortunately, infection with AdV in the normal host is generally mild. This is one reason why its initial use as a gene-therapy vector appeared to be so promising. Unfortunately, both innate and adaptive responses to the virus have limited the development of AdV vectors as a tool of gene therapy by increasing toxicity and limiting duration of transgene expression. This article will focus on the innate immune response to infection with wild-type AdV and exposure to AdV gene-therapy vectors. As much of the known information relates to the pulmonary inflammatory response, this organ system will be emphasized. This article will also discuss how that understanding has led to the creation of new vectors for use in gene therapy.
Collapse
Affiliation(s)
- Seth M Gregory
- Division of Pulmonary & Critical Care Medicine of the Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Shoab A Nazir
- Division of Pulmonary & Critical Care Medicine of the Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jordan P Metcalf
- Division of Pulmonary & Critical Care Medicine of the Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104-5097, USA
| |
Collapse
|
23
|
Cawood R, Wong SL, Di Y, Baban DF, Seymour LW. MicroRNA controlled adenovirus mediates anti-cancer efficacy without affecting endogenous microRNA activity. PLoS One 2011; 6:e16152. [PMID: 21264344 PMCID: PMC3018484 DOI: 10.1371/journal.pone.0016152] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 12/13/2010] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs are small non-coding RNA molecules that regulate mRNA translation and stability by binding to complementary sequences usually within the 3' un-translated region (UTR). We have previously shown that the hepatic toxicity caused by wild-type Adenovirus 5 (Ad5WT) in mice can be prevented by incorporating 4 binding sites for the liver-specific microRNA, mir122, into the 3' UTR of E1A mRNA. This virus, termed Ad5mir122, is a promising virotherapy candidate and causes no obvious liver pathology. Herein we show that Ad5mir122 maintains wild-type lytic activity in cancer cells not expressing mir122 and assess any effects of possible mir122 depletion in host cells. Repeat administration of 2×10(10) viral particles of Admir122 to HepG2 tumour bearing mice showed significant anti-cancer efficacy. RT-QPCR showed that E1A mRNA was down-regulated 29-fold in liver when compared to Ad5WT. Western blot for E1A confirmed that all protein variants were knocked down. RT-QPCR for mature mir122 in infected livers showed that quantity of mir122 remained unaffected. Genome wide mRNA microarray profiling of infected livers showed that although the transcript level of >3900 different mRNAs changed more than 2-fold following Ad5WT infection, less than 600 were changed by Ad5mir122. These were then filtered to select mRNAs that were only altered by Ad5mir122 and the remaining 21 mRNAs were compared to predicted mir122 targets. No mir122 target mRNAs were affected by Ad5 mir122. These results demonstrate that the exploitation of microRNA regulation to control virus replication does not necessarily affect the level of the microRNA or the endogenous mRNA targets.
Collapse
Affiliation(s)
- Ryan Cawood
- Medicinal Virology Research Group, Department of Clinical Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Suet-Ling Wong
- Medicinal Virology Research Group, Department of Clinical Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Ying Di
- Medicinal Virology Research Group, Department of Clinical Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Dilair F. Baban
- Genomics Group, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Leonard W. Seymour
- Medicinal Virology Research Group, Department of Clinical Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
24
|
Steinmetz NF. Viral nanoparticles as platforms for next-generation therapeutics and imaging devices. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2010; 6:634-41. [PMID: 20433947 PMCID: PMC2948632 DOI: 10.1016/j.nano.2010.04.005] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Revised: 03/25/2010] [Accepted: 04/07/2010] [Indexed: 12/12/2022]
Abstract
Nanomaterials have been developed for potential applications in biomedicine, such as tissue-specific imaging and drug delivery. There are many different platforms under development, each with advantages and disadvantages, but viral nanoparticles (VNPs) are particularly attractive because they are naturally occurring nanomaterials, and as such they are both biocompatible and biodegradable. VNPs can be designed and engineered using both genetic and chemical protocols. The use of VNPs has evolved rapidly since their introduction 20 years ago, encompassing numerous chemistries and modification strategies that allow the functionalization of VNPs with imaging reagents, targeting ligands, and therapeutic molecules. This review discusses recent advances in the design of "smart" targeted VNPs for therapeutic and imaging applications. FROM THE CLINICAL EDITOR This review focuses on viral nanoparticles, which are considered attractive naturally occurring nanomaterials due to their inherent biocompatibility and biodegradability. These can be used as imaging reagents, targeting ligands and therapeutic molecules.
Collapse
Affiliation(s)
- Nicole F Steinmetz
- Department of Molecular Biology and Center of Integrative Molecular Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
25
|
Minimal RB-responsive E1A promoter modification to attain potency, selectivity, and transgene-arming capacity in oncolytic adenoviruses. Mol Ther 2010; 18:1960-71. [PMID: 20808288 DOI: 10.1038/mt.2010.173] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oncolytic adenoviruses are promising anticancer agents due to their ability to self-amplify at the tumor mass. However, tumor stroma imposes barriers difficult to overcome by these agents. Transgene expression is a valuable strategy to counteract these limitations and to enhance antitumor activity. For this purpose, the genetic backbone in which the transgene is inserted should be optimized to render transgene expression compatible with the adenovirus replication cycle and to keep genome size within the encapsidation size limit. In order to design a potent and selective oncolytic adenovirus that keeps intact all the viral functions with minimal increase in genome size, we inserted palindromic E2F-binding sites into the endogenous E1A promoter. The insertion of these sites controlling E1A-Δ24 results in a low systemic toxicity profile in mice. Importantly, the E2F-binding sites also increased the cytotoxicity and the systemic antitumor activity relative to wild-type adenovirus in all cancer models tested. The low toxicity and the increased potency results in improved antitumor efficacy after systemic injection and increased survival of mice carrying tumors. Furthermore, the constrained genome size of this backbone allows an efficient and potent expression of transgenes, indicating that this virus holds promise for overcoming the limitations of oncolytic adenoviral therapy.
Collapse
|
26
|
Fang L, Pu YY, Hu XC, Sun LJ, Luo HM, Pan SK, Gu JZ, Cao XR, Su CQ. Antiangiogenesis gene armed tumor-targeting adenovirus yields multiple antitumor activities in human HCC xenografts in nude mice. Hepatol Res 2010; 40:216-28. [PMID: 19788685 DOI: 10.1111/j.1872-034x.2009.00580.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIM Gene therapy represents a promising therapeutic strategy for hepatocellular carcinoma (HCC). To improve the ratio of killing efficacy on tumor cells to side-effect on normal cells, we constructed an oncolytic adenovirus vector, AdSu-hE, expressing the human endostatin (hE) gene, in which the chimeric promoter of human epidermal growth factor receptor 2 enhancer and human telomerase reverse transcriptase promoter was used to control the adenoviral E1a gene. METHODS Tumor-selective replication of adenovirus AdSu-hE and its concomitant expression of endostatin were measured by 50% tissue culture infective dose method, fluorescent protein expression, Western blot and enzyme linked immunosorbent assay in cancer and normal cell lines. The antitumor efficacy was observed in nude mice bearing human HCCs. RESULTS The oncolytic adenovirus AdSu-hE replicated restrictedly in telomerase-positive cancer cells and resulted in oncolysis, but did not replicate in normal cell lines. Along with virus replication, AdSu-hE mediated 5-fold increased expression of endostatin in tumor cells compared with that in normal cells. Moreover, AdSu-hE expressed more endostatin in cancer cells than the non-replicative adenovirus vector Ad-hE. In vivo administration of the oncolytic adenovirus AdSu-hE into HCC-bearing nude mice produced a significant tumor reduction by synergistic effects of virus oncolysis and endostatin antiangiogenesis. CONCLUSION The oncolytic virus with antiangiogenesis gene driven by the chimeric promoter has an improved killing efficacy on tumor cells, and may be useful for cancer gene therapy.
Collapse
Affiliation(s)
- Lin Fang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
A modified E2F-1 promoter improves the efficacy to toxicity ratio of oncolytic adenoviruses. Gene Ther 2009; 16:1441-51. [DOI: 10.1038/gt.2009.103] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
28
|
Young A, McNeish IA. Oncolytic adenoviral gene therapy in ovarian cancer: why we are not wasting our time. Future Oncol 2009; 5:339-57. [PMID: 19374541 DOI: 10.2217/fon.09.11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Preclinical gene-therapy studies in the past 15 years have repeatedly raised hopes that we were about to enter a brave new era. However, many clinical trials have disappointed. For tumor types with poor response rates to first-line conventional cytotoxic chemotherapy and/or high rates of chemorefractory disease, there remain very few treatment options. In this article we review gene therapy within the context of ovarian cancer. We examine why clinical data have been discouraging and discuss how the lessons learned from earlier trials are being applied to current research.
Collapse
Affiliation(s)
- Am Young
- Centre for Molecular Oncology & Imaging, Institute of Cancer, Barts & The London School of Medicine & Dentistry, Charterhouse Square, London EC1M 6BQ, UK
| | | |
Collapse
|
29
|
Abstract
Advances in genetics, proteomics and cellular and molecular biology are being integrated and translated to develop effective methods for the prevention and control of cancer. One such combined effort is to create multifunctional nanodevices that will specifically recognize tumors and thus enable early diagnosis and provide targeted treatment of this disease. Viral particles are being considered for this purpose since they are inherently nanostructures with well-defined geometry and uniformity, ideal for displaying molecules in a precise spatial distribution at the nanoscale level and subject to greater structural control. Viruses are presumably the most efficient nanocontainer for cellular delivery as they have naturally evolved mechanisms for binding to and entering cells. Virus-based systems typically require genetic or chemical modification of their surfaces to achieve tumor-specific interactions. Interestingly, canine parvovirus (CPV) has a natural affinity for transferrin receptors (TfRs) (both of canine and human origin) and this property could be harnessed as TfRs are overexpressed by a variety of human tumor cells. Since TfR recognition relies on the CPV capsid protein, we envisioned the use of virus or its shells as tumor targeting agents. We observed that derivatization of CPV virus-like particles (VLPs) with dye molecules did not impair particle binding to TfRs or internalization into human tumor cells. Thus CPV-based VLPs with a natural tropism for TfRs hold great promise in the development of novel nanomaterial for delivery of a therapeutic and/or genetic cargo.
Collapse
Affiliation(s)
- P Singh
- Division of Hematology and Oncology, Department of Medicine, Building 23, Room 436A, UCI Medical Center, 101 City Drive South, Orange, CA 92868, USA.
| |
Collapse
|
30
|
Destito G, Schneemann A, Manchester M. Biomedical Nanotechnology Using Virus-Based Nanoparticles. Curr Top Microbiol Immunol 2009; 327:95-122. [DOI: 10.1007/978-3-540-69379-6_5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
31
|
Doloff JC, Waxman DJ, Jounaidi Y. Human telomerase reverse transcriptase promoter-driven oncolytic adenovirus with E1B-19 kDa and E1B-55 kDa gene deletions. Hum Gene Ther 2008; 19:1383-400. [PMID: 18771358 PMCID: PMC2715448 DOI: 10.1089/hum.2008.056] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Accepted: 09/04/2008] [Indexed: 12/14/2022] Open
Abstract
We constructed an oncolytic adenovirus, Adeno-hTERT-E1A, with deletions of the viral E1B, E3A, and E3B regions and insertion of a human telomerase reverse transcriptase (hTERT) promoter-driven early viral 1A (E1A) cassette that confers high transcriptional activity in multiple human tumor cell lines. The oncolytic potential of Adeno-hTERT-E1A was characterized in comparison with that of the E1B-55 kDa- and E3B-region-deleted oncolytic adenovirus ONYX-015. Tumor cells infected with Adeno-hTERT-E1A expressed dramatically higher levels of E1A oncoprotein, underwent enhanced lysis, and displayed an earlier and higher apoptotic index than cells infected with ONYX-015. Despite the increase in virus-induced apoptotic death, Adeno-hTERT-E1A replicated and produced functional progeny leading to viral spread, but with reduced efficiency compared with ONYX-015, in particular in A549 cells. Virus-induced E1A expression, host cell apoptosis, viral hexon protein production, and DNA synthesis were markedly reduced in primary human hepatocytes after infection with Adeno-hTERT-E1A as compared with ONYX-015. The strong oncolytic activity of Adeno-hTERT-E1A in tumor cell culture translated into superior antitumor activity in vivo in an MDA-MB-231 solid tumor xenograft model. Adeno-hTERT-E1A thus has strong therapeutic potential and an improved safety profile compared with ONYX-015, which may lead to reduced toxicity in the clinic.
Collapse
Affiliation(s)
- Joshua C Doloff
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, MA 02215, USA
| | | | | |
Collapse
|
32
|
Liu TC, Hwang TH, Bell JC, Kirn DH. Development of targeted oncolytic virotherapeutics through translational research. Expert Opin Biol Ther 2008; 8:1381-91. [PMID: 18694356 DOI: 10.1517/14712598.8.9.1381] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Oncolytic virotherapeutics is a promising platform for cancer treatment but the product class has yet been successful. The key to success is integration of bidirectional translational research to rapidly address issues encountered in the laboratory and the clinics. OBJECTIVE We highlight the hurdles identified for the targeted oncolytic virotherapy approach, specifically those identified in clinical trials with wild-type viruses and first-generation targeted agents. We also analyze the translational research and development that has been applied to overcome these hurdles, including virus engineering and design improvements for next-generation virotherapeutics. RESULTS/CONCLUSION The iterative loop between the clinic and the lab can function as a major driving force to optimize products from this platform.
Collapse
Affiliation(s)
- Ta-Chiang Liu
- Jennerex Biotherapeutics, One Market Street, Spear Tower, Suite 2260, San Francisco, CA 94105, USA
| | | | | | | |
Collapse
|
33
|
Doloff JC, Waxman DJ, Jounaidi Y. hTERT-promoter driven oncolytic adenovirus with E1B-19 kDa and E1B-55 kDa gene deletions. Hum Gene Ther 2008. [DOI: 10.1089/hgt.2008.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
34
|
Singh P, Prasuhn D, Yeh RM, Destito G, Rae CS, Osborn K, Finn MG, Manchester M. Bio-distribution, toxicity and pathology of cowpea mosaic virus nanoparticles in vivo. J Control Release 2007; 120:41-50. [PMID: 17512998 PMCID: PMC2849971 DOI: 10.1016/j.jconrel.2007.04.003] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2006] [Revised: 03/29/2007] [Accepted: 04/04/2007] [Indexed: 12/16/2022]
Abstract
Virus-based nanoparticles (VNPs) from a variety of sources are being developed for biomedical and nanotechnology applications that include tissue targeting and drug delivery. However, the fate of most of those particles in vivo has not been investigated. Cowpea mosaic virus (CPMV), a plant comovirus, has been found to be amenable to the attachment of a variety of molecules to its coat protein, as well as to modification of the coat protein sequence by genetic means. We report here the results of studies of the bio-distribution, toxicology, and pathology of CPMV in mice. Plasma clearance and tissue biodistribution were measured using CPMV particles derivatized with lanthanide metal complexes. CPMV particles were cleared rapidly from plasma, falling to undetectable levels within 20 min. By 30 min the majority of the injected VNPs were trapped in the liver and to a lesser extent the spleen with undetectable amounts in other tissues. At doses of 1 mg, 10 mg and 100 mg per kg body weight, no toxicity was noted and the mice appeared to be normal. Hematology was essentially normal, although with the highest dose examined, the mice were somewhat leukopenic with relative decreases in both neutrophils and lymphocytes. Histological examination of the spleen showed cellular infiltration, which upon flow cytometry analyses revealed elevated B lymphocytes on the first day following virus administration that subsequently subsided. Microscopic evaluation of various other tissues revealed a lack of apparent tissue degeneration or necrosis. Overall, CPMV appears to be a safe and non-toxic platform for in vivo biomedical applications.
Collapse
Affiliation(s)
- Pratik Singh
- Center for Integrative Molecular Biosciences, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- Department of Cell Biology, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Duane Prasuhn
- Department of Chemistry and The Skaggs Institute for Chemical Biology, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Robert M. Yeh
- Department of Chemistry and The Skaggs Institute for Chemical Biology, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Giuseppe Destito
- Center for Integrative Molecular Biosciences, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- Department of Cell Biology, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi Magna Graecia di Catanzaro Viale Europa, Campus Universitario di Germaneto 88100, Catanzaro, ITALY
| | - Chris S. Rae
- Center for Integrative Molecular Biosciences, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- Department of Cell Biology, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Kent Osborn
- Department of Animal Resources, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - M. G. Finn
- Center for Integrative Molecular Biosciences, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- Department of Chemistry and The Skaggs Institute for Chemical Biology, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Marianne Manchester
- Center for Integrative Molecular Biosciences, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- Department of Cell Biology, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
35
|
Cascallo M, Alonso MM, Rojas JJ, Perez-Gimenez A, Fueyo J, Alemany R. Systemic toxicity-efficacy profile of ICOVIR-5, a potent and selective oncolytic adenovirus based on the pRB pathway. Mol Ther 2007; 15:1607-15. [PMID: 17579575 DOI: 10.1038/sj.mt.6300239] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
E2F acts as a transcriptional repressor when bound to unphosphorylated RB during the G(1) or G(0) phase. Upon phosphorylation, E2F is released from the E2F-RB complexes to activate transcription. Tumor cells are characterized by an increase in the level of "free" E2F as a consequence of the absence or hyperphosphorylation of RB. The E2F-1 promoter is a well-characterized E2F-responsive promoter, and it can be used to control adenovirus E1a gene expression as a strategy to achieve tumor-selective expression and replication of an adenovirus. ICOVIR-5 (Ad-DM-E2F-K-Delta24RGD) is an optimized oncolytic adenovirus that combines E1a transcriptional control by an insulated form of the E2F promoter with the Delta24 mutation of E1a to improve the therapeutic index of AdDelta24RGD. ICOVIR-5 also contains the Kozak sequence at the E1a start codon, which is important to restore E1a expression and viral replication to AdwtRGD levels in tumor cells. The unique combination of genetic elements in ICOVIR-5 allows the selectivity for cells with a deregulated E2F-RB pathway to be increased and potent anti-tumoral activity to be maintained. Dose-response toxicological and efficacy studies after a single systemic administration in pre-clinical models in mice are presented to demonstrate that this virus holds promise for treatment of disseminated cancer.
Collapse
Affiliation(s)
- Manel Cascallo
- Translational Research Laboratory, Institut d'Investigació Biomèdica de Bellvitge-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
36
|
Koizumi N, Yamaguchi T, Kawabata K, Sakurai F, Sasaki T, Watanabe Y, Hayakawa T, Mizuguchi H. Fiber-modified adenovirus vectors decrease liver toxicity through reduced IL-6 production. THE JOURNAL OF IMMUNOLOGY 2007; 178:1767-73. [PMID: 17237426 DOI: 10.4049/jimmunol.178.3.1767] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adenovirus (Ad) vectors are one of the most commonly used viral vectors in gene therapy clinical trials. However, they elicit a robust innate immune response and inflammatory responses. Improvement of the therapeutic index of Ad vector gene therapy requires elucidation of the mechanism of Ad vector-induced inflammation and cytokine/chemokine production as well as development of the safer vector. In the present study, we found that the fiber-modified Ad vector containing poly-lysine peptides in the fiber knob showed much lower serum IL-6 and aspartate aminotransferase levels (as a maker of liver toxicity) than the conventional Ad vector after i.v. administration, although the modified Ad vector showed higher transgene production in the liver than the conventional Ad vector. RT-PCR analysis showed that spleen, not liver, is the major site of cytokine, chemokine, and IFN expression. Splenic CD11c(+) cells were found to secret cytokines. The tissue distribution of Ad vector DNA showed that spleen distribution was much reduced in this modified Ad vector, reflecting reduced IL-6 levels in serum. Liver toxicity by the conventional Ad vector was reduced by anti-IL-6R Ab, suggesting that IL-6 signaling is involved in liver toxicity and that decreased liver toxicity of the modified Ad vector was due in part to the reduced IL-6 production. This study contributes to an understanding of the biological mechanism in innate immune host responses and liver toxicity toward systemically administered Ad vectors and will help in designing safer gene therapy methods that can reduce robust innate immunity and inflammatory responses.
Collapse
Affiliation(s)
- Naoya Koizumi
- Laboratory of Gene Transfer and Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Shashkova EV, Spencer JF, Wold WSM, Doronin K. Targeting Interferon-α Increases Antitumor Efficacy and Reduces Hepatotoxicity of E1A-mutated Spread-enhanced Oncolytic Adenovirus. Mol Ther 2007; 15:598-607. [PMID: 17191072 DOI: 10.1038/sj.mt.6300064] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Accepted: 10/30/2006] [Indexed: 11/09/2022] Open
Abstract
Novel approaches are needed to improve the antitumor potency and to increase the cancer specificity of oncolytic adenoviruses (Ad). We hypothesized that the combination of interferon-alpha (IFN-alpha) expression with a specific mutation in the e1a gene of Ad could target vector replication to genetic defects in the IFN-alpha pathway resulting in both improved antitumor efficacy and reduced toxicity. The conditionally replicative Ad vector KD3-IFN carries the dl1101/1107 mutation in the e1a gene that eliminates binding of E1A proteins to p300/CBP and pRb. KD3-IFN expresses human IFN-alpha in concurrence with vector replication and overexpresses the adenovirus death protein (ADP; E3-11.6K). The antitumor activity of KD3-IFN was significantly higher than that of a control vector in established human hepatocellular carcinoma tumors in immunodeficient mice and in hamster kidney cancer tumors in immunocompetent Syrian hamsters. The dl1101/1107 mutation rendered Ad replication sensitive to the antiviral effect of IFN-alpha in normal as opposed to cancer cells. These results translated to reduced vector toxicity upon systemic administration to C57BL/6 mice. The combination of Ad oncolysis, ADP overexpression, and IFN-alpha-mediated immunotherapy represents a three-pronged approach for increasing the anticancer efficacy of replicative Ads. Exploiting the dl1101/1107 mutation provides a mechanism for additional selectivity of IFN-alpha-expressing replication-competent Ads.
Collapse
|
38
|
Majem M, Cascallo M, Bayo-Puxan N, Mesia R, Germa JR, Alemany R. Control of E1A under an E2F-1 promoter insulated with the myotonic dystrophy locus insulator reduces the toxicity of oncolytic adenovirus Ad-Δ24RGD. Cancer Gene Ther 2006; 13:696-705. [PMID: 16498429 DOI: 10.1038/sj.cgt.7700940] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We previously described Ad-Delta24RGD as an enhanced-infectivity oncolytic adenovirus that targets tumors with an impaired RB pathway. The common alteration of this pathway in cancer eliminates the interaction of pRB with E2F and releases free E2F to activate E2F-responsive promoters, including the E2F-1 promoter. To improve the selectivity towards RB pathway-defective tumors and reduce the toxicity of Ad-Delta24RGD we aimed to control E1A-Delta24 expression under the E2F-1 promoter. A polyA signal was inserted upstream of the E2F-1 promoter to stop transcription initiated at the adenovirus ITR and packaging signal. The human myotonic dystropy locus insulator (DM-1) was also located between the E1a enhancers and the E2F-1 promoter to further insulate the promoter. The Ad-Delta24RGD derivative containing these insulation sequences expressed less E1a-Delta24 in normal cells and resulted less toxic while maintaining the potent oncolytic activity of the parental virus. These results demonstrate that the human DM-1 inslulator can function in an adenovirus context to maintain heterologous promoter selectivity. The new oncolytic adenovirus presented here may represent a valuable therapeutic option for a broad range of tumors with a deregulated E2F/pRB pathway.
Collapse
Affiliation(s)
- M Majem
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
39
|
Lockley M, Fernandez M, Wang Y, Li NF, Conroy S, Lemoine N, McNeish I. Activity of the adenoviral E1A deletion mutant dl922-947 in ovarian cancer: comparison with E1A wild-type viruses, bioluminescence monitoring, and intraperitoneal delivery in icodextrin. Cancer Res 2006; 66:989-98. [PMID: 16424034 DOI: 10.1158/0008-5472.can-05-2691] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The adenoviral mutant dl922-947 has potent activity in a variety of tumors. We investigated the efficacy of dl922-947 in ovarian carcinoma; compared its activity to wild-type adenovirus, dl309, and dl1520; and investigated the use of icodextrin to enhance activity in vivo. We also assessed the utility of luciferase bioluminescence imaging to quantify the response of human ovarian carcinoma xenografts to dl922-947. Ovarian carcinoma cell lines were transfected in vitro with dl922-947, adenovirus 5 wild-type (Ad5 WT), dl309, and dl1520 and monitored for S-phase induction, viral protein expression, replication, and overall survival. In vivo, the efficacy of dl922-947 when delivered in PBS or icodextrin to female nude mice bearing IGROV1 xenografts was determined. In vitro, dl922-947 induced lysis with greater efficacy than Ad5 WT, dl309, or dl1520 in all ovarian carcinoma cell lines tested, which was associated with earlier expression of viral proteins and S-phase induction. The lytic effect in immortalized ovarian surface epithelial cells confirmed that cellular retinoblastoma pathway status is a strong determinant of dl922-947 activity. In vivo, i.p. delivery of dl922-947 (5 x 10(9) particles daily x 5) increased median survival from 20 to 96 days (P < 0.0001) and delivery in icodextrin-enhanced survival further. However, delayed hepatic toxicity was evident in some dl922-947-treated mice, which was not dependent upon viral replication within tumor cells or the liver. dl922-947 has potency in ovarian carcinoma and i.p. delivery in icodextrin may enhance this activity. Immunocompetent models of ovarian carcinoma are required for further evaluation of hepatotoxicity.
Collapse
Affiliation(s)
- Michelle Lockley
- Cancer Research UK Molecular Oncology Unit, Barts and the London School of Medicine, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Lenaerts L, Verbeken E, De Clercq E, Naesens L. Mouse adenovirus type 1 infection in SCID mice: an experimental model for antiviral therapy of systemic adenovirus infections. Antimicrob Agents Chemother 2005; 49:4689-99. [PMID: 16251313 PMCID: PMC1280150 DOI: 10.1128/aac.49.11.4689-4699.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2005] [Revised: 07/15/2005] [Accepted: 08/16/2005] [Indexed: 11/20/2022] Open
Abstract
The importance of human adenovirus infections in immunocompromised patients urges for new and adequate antiadenovirus compounds. Since human adenoviruses are species specific, animal models for systemic adenovirus infections rely on a nonhuman adenovirus. We established mouse adenovirus type 1 (MAV-1) infection of BALB/c SCID mice as a model for the evaluation of antiadenovirus therapy. In vitro studies with mouse embryonic fibroblasts pointed to the acyclic nucleoside phosphonate cidofovir and the N-7-substituted acyclic derivative 2-amino-7-(1,3-dihydroxy-2-propoxymethyl)purine (S-2242) as markedly active compounds against MAV-1. SCID mice, infected intranasally with MAV-1, developed a fatal disseminated infection after approximately 19 days, characterized by hemorrhagic enteritis. Several techniques were optimized to monitor viral, immunological, and pathological aspects of MAV-1 infection. Real-time PCR quantification of viral DNA revealed that after replication in the lungs, virus disseminated to several organs, including the brain, liver, spleen, intestine, heart, and kidneys (resulting in viruria). Immunohistochemical staining showed that MAV-1 was localized in the endothelial cells of the affected organs. Using reverse transcription-PCR, tissue levels of proinflammatory cytokines (i.e., interleukin-1beta and tumor necrosis factor alpha) were found to be markedly increased. The MAV-1/SCID model appears to be an appropriate model for in vivo evaluation of antiadenovirus agents. Treatment with cidofovir or S-2242 at a dose of 100 mg per kg of body weight resulted in a significant delay in MAV-1-related death, although these antivirals were unable to completely suppress virus replication despite continued drug treatment. These findings suggest that complete virus clearance during antiviral therapy for disseminated adenovirus infection may require an efficient adaptive immune response from the host.
Collapse
Affiliation(s)
- L Lenaerts
- Rega Institute for Medical Research, Division of Virology and Chemotherapy, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | | | | | | |
Collapse
|
42
|
Abstract
Adenovirus is a common infectious pathogen in both children and adults. It is a significant cause of morbidity in immunocompetent people living in crowded living conditions and of mortality in immunocompromised hosts. It has more recently become a popular vehicle for gene therapy applications. The host response to wild-type infection and gene therapy vector exposure involves both the innate and adaptive immune systems. The initial innate immune response is associated with the severe acute manifestations of adenovirus infection and also plays a significant role in acute toxicity owing to adenovirus vector exposure. This review discusses the innate immune response primarily during wild-type adenovirus infection because this serves as the basis for understanding the response during both natural infection and exposure to adenovirus vectors.
Collapse
Affiliation(s)
- Shoab A Nazir
- Oklahoma University Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | |
Collapse
|
43
|
Shayakhmetov DM, Li ZY, Ni S, Lieber A. Interference with the IL-1-signaling pathway improves the toxicity profile of systemically applied adenovirus vectors. THE JOURNAL OF IMMUNOLOGY 2005; 174:7310-9. [PMID: 15905578 DOI: 10.4049/jimmunol.174.11.7310] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The safety of gene therapy vectors is a major concern when novel viral or nonviral therapeutics are proposed for applications in humans. Adenovirus (Ad) vectors have been extensively used as efficient gene delivery vehicles in vitro over the last two decades. However, upon i.v. application, they elicit robust innate and inflammatory responses that may be fatal for the host. To date, the primary cytokines and chemokines involved in the initiation of these host responses remain illusive. In this study, we demonstrate that IL-1 is a major mediator involved in the initiation of immediate host responses toward i.v. applied Ad vectors. Using mice in which IL-1 signaling was genetically eliminated (IL-1RI-KO), or wild-type animals for which signaling was blocked by anti-IL-1 Abs, we found that i.v. applied Ad vectors elicited dramatically reduced acute inflammatory responses when compared with control animals. Importantly, the efficiency of Ad gene transfer in vivo was not significantly affected by interfering with IL-1 signaling. Using an in situ hybridization technique, we found that hepatocytes and Kupffer cells trigger IL-1 transcription in liver tissue after i.v. Ad vector administration. We also found that expression of the MIP-2 chemokine gene (which is responsible for recruitment of neutrophils to the liver) depends on IL-1 activation. Our data indicate that immediate innate and inflammatory host responses toward i.v. applied Ad vectors can be pharmacologically controlled through interference with IL-1 signaling pathways.
Collapse
Affiliation(s)
- Dmitry M Shayakhmetov
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, 98195-7720, USA
| | | | | | | |
Collapse
|