1
|
Fayyaz A, Haqqi A, Khan R, Irfan M, Khan K, Reiner Ž, Sharifi-Rad J, Calina D. Revolutionizing cancer treatment: the rise of personalized immunotherapies. Discov Oncol 2024; 15:756. [PMID: 39692978 DOI: 10.1007/s12672-024-01638-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Interest in biological therapy for cancer has surged due to its precise targeting of cancer cells and minimized impact on surrounding healthy tissues. This review discusses various biological cancer therapies, highlighting advanced alternatives over conventional chemotherapy alone. It explores DNA and RNA-based vaccines, T-cell modifications, adoptive cell transfer, CAR T cell therapy, angiogenesis inhibitors, and the combination of immunotherapy with chemotherapy, offering a holistic view of the potential in cancer treatment. Additionally, it discusses the role of nanotechnology in increasing the efficacy of cancer-targeting drugs, as well as cytokine and immunoconjugate therapies for bolstering immune system effectiveness against neoplastic cells. The potential of gene potential for precise targeting of cancer-linked genes and the application of oncolytic viruses against virus-associated cancers are also discussed. The review identifies significant advancements in the targeted treatment of cancer by biological methods. It acknowledges the challenges, including drug resistance and the need for high specificity in certain therapies, while also highlighting the effectiveness of cancer vaccines, modified T-cells, and oncolytic viruses. Biological therapies are a promising frontier in cancer treatment, offering the potential for more personalized and effective therapeutic strategies. Despite existing challenges, ongoing research and clinical trials are fundamental for overcoming current limitations and enhancing the efficacy of biological therapies in cancer care.
Collapse
Affiliation(s)
- Amna Fayyaz
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Aleena Haqqi
- School of Medical Laboratory Technology, Faculty of Allied Health Sciences, Minhaj University Lahore (MUL), Lahore, 54000, Pakistan
| | - Rashid Khan
- Department of Pharmacy, Punjab University College of Pharmacy University of Punjab Lahore, Lahore, 54000, Pakistan
| | - Muhammad Irfan
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Khushbukhat Khan
- Cancer Clinical Research Unit, Trials360, Lahore, 54000, Pakistan.
| | - Željko Reiner
- Department for Metabolic Diseases, University Hospital Center Zagreb, Zagreb, Croatia
- Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Centro de Estudios Tecnológicos, Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
2
|
Semenova AV, Sivolobova GF, Grazhdantseva AA, Agafonov AP, Kochneva GV. Reporter Transgenes for Monitoring the Antitumor Efficacy of Recombinant Oncolytic Viruses. Acta Naturae 2022; 14:46-56. [PMID: 36348722 PMCID: PMC9611865 DOI: 10.32607/actanaturae.11719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022] Open
Abstract
Accurate measurement of tumor size and margins is crucial for successful oncotherapy. In the last decade, non-invasive imaging modalities, including optical imaging using non-radioactive substrates, deep-tissue imaging with radioactive substrates, and magnetic resonance imaging have been developed. Reporter genes play the most important role among visualization tools; their expression in tumors and metastases makes it possible to track changes in the tumor growth and gauge therapy effectiveness. Oncolytic viruses are often chosen as a vector for delivering reporter genes into tumor cells, since oncolytic viruses are tumor-specific, meaning that they infect and lyse tumor cells without damaging normal cells. The choice of reporter transgenes for genetic modification of oncolytic viruses depends on the study objectives and imaging methods used. Optical imaging techniques are suitable for in vitro studies and small animal models, while deep-tissue imaging techniques are used to evaluate virotherapy in large animals and humans. For optical imaging, transgenes of fluorescent proteins, luciferases, and tyrosinases are used; for deep-tissue imaging, the most promising transgene is the sodium/iodide symporter (NIS), which ensures an accumulation of radioactive isotopes in virus-infected tumor cells. Currently, NIS is the only reporter transgene that has been shown to be effective in monitoring tumor virotherapy not only in preclinical but also in clinical studies.
Collapse
Affiliation(s)
- A. V. Semenova
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| | - G. F. Sivolobova
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| | - A. A. Grazhdantseva
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| | - A. P. Agafonov
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| | - G. V. Kochneva
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| |
Collapse
|
3
|
Shao S, Yang X, Zhang YN, Wang XJ, Li K, Zhao YL, Mou XZ, Hu PY. Oncolytic Virotherapy in Peritoneal Metastasis Gastric Cancer: The Challenges and Achievements. Front Mol Biosci 2022; 9:835300. [PMID: 35295845 PMCID: PMC8918680 DOI: 10.3389/fmolb.2022.835300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer (GC) is the fourth most common cancer and the second leading cause of cancer death globally. Although the mortality rate in some parts of the world, such as East Asia, is still high, new treatments and lifestyle changes have effectively reduced deaths from this type of cancer. One of the main challenges of this type of cancer is its late diagnosis and poor prognosis. GC patients are usually diagnosed in the advanced stages of the disease, which is often associated with peritoneal metastasis (PM) and significantly reduces survival. This type of metastasis in patients with GC poses a serious challenge due to limitations in common therapies such as surgery and tumor resection, as well as failure to respond to systemic chemotherapy. To solve this problem, researchers have used virotherapy such as reovirus-based anticancer therapy in patients with GC along with PM who are resistant to current chemotherapies because this therapeutic approach is able to overcome immune suppression by activating dendritic cells (DCs) and eventually lead to the intrinsic activity of antitumor effector T cells. This review summarizes the immunopathogenesis of peritoneal metastasis of gastric cancer (PMGC) and the details for using virotherapy as an effective anticancer treatment approach, as well as its challenges and opportunities.
Collapse
Affiliation(s)
- Su Shao
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, China
| | - Xue Yang
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
- Clinical Research Institute, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
| | - You-Ni Zhang
- Department of Traumatology, Tiantai People’s Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People’s Hospital), Taizhou, China
| | - Xue-Jun Wang
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, China
| | - Ke Li
- Guangdong Techpool Bio-pharma Co., Ltd., Guangzhou, China
| | - Ya-Long Zhao
- Guangdong Techpool Bio-pharma Co., Ltd., Guangzhou, China
| | - Xiao-Zhou Mou
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
- Clinical Research Institute, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
- *Correspondence: Xiao-Zhou Mou, ; Pei-Yang Hu,
| | - Pei-Yang Hu
- Department of Traumatology, Tiantai People’s Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People’s Hospital), Taizhou, China
- *Correspondence: Xiao-Zhou Mou, ; Pei-Yang Hu,
| |
Collapse
|
4
|
Biological Therapies in the Treatment of Cancer-Update and New Directions. Int J Mol Sci 2021; 22:ijms222111694. [PMID: 34769123 PMCID: PMC8583892 DOI: 10.3390/ijms222111694] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
Biological therapies have changed the face of oncology by targeting cancerous cells while reducing the effect on normal tissue. This publication focuses mainly on new therapies that have contributed to the advances in treatment of certain malignancies. Immunotherapy, which has repeatedly proven to be a breakthrough therapy in melanoma, as well as B-ALL therapy with CAR T cells, are of great merit in this progress. These therapies are currently being developed by modifying bispecific antibodies and CAR T cells to improve their efficiency and bioavailability. Work on improving the therapy with oncolytic viruses is also progressing, and efforts are being made to improve the immunogenicity and stability of cancer vaccines. Combining various biological therapies, immunotherapy with oncolytic viruses or cancer vaccines is gaining importance in cancer therapy. New therapeutic targets are intensively sought among neoantigens, which are not immunocompromised, or antigens associated with tumor stroma cells. An example is fibroblast activation protein α (FAPα), the overexpression of which is observed in the case of tumor progression. Universal therapeutic targets are also sought, such as the neurotrophic receptor tyrosine kinase (NTRK) gene fusion, a key genetic driver present in many types of cancer. This review also raises the problem of the tumor microenvironment. Stromal cells can protect tumor cells from chemotherapy and contribute to relapse and progression. This publication also addresses the problem of cancer stem cells resistance to treatment and presents attempts to avoid this phenomenon. This review focuses on the most important strategies used to improve the selectivity of biological therapies.
Collapse
|
5
|
Concilio SC, Russell SJ, Peng KW. A brief review of reporter gene imaging in oncolytic virotherapy and gene therapy. Mol Ther Oncolytics 2021; 21:98-109. [PMID: 33981826 PMCID: PMC8065251 DOI: 10.1016/j.omto.2021.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reporter gene imaging (RGI) can accelerate development timelines for gene and viral therapies by facilitating rapid and noninvasive in vivo studies to determine the biodistribution, magnitude, and durability of viral gene expression and/or virus infection. Functional molecular imaging systems used for this purpose can be divided broadly into deep-tissue and optical modalities. Deep-tissue modalities, which can be used in animals of any size as well as in human subjects, encompass single photon emission computed tomography (SPECT), positron emission tomography (PET), and functional/molecular magnetic resonance imaging (f/mMRI). Optical modalities encompass fluorescence, bioluminescence, Cerenkov luminescence, and photoacoustic imaging and are suitable only for small animal imaging. Here we discuss the mechanisms of action and relative merits of currently available reporter gene systems, highlighting the strengths and weaknesses of deep tissue versus optical imaging systems and the hardware/reagents that are used for data capture and processing. In light of recent technological advances, falling costs of imaging instruments, better availability of novel radioactive and optical tracers, and a growing realization that RGI can give invaluable insights across the entire in vivo translational spectrum, the approach is becoming increasingly essential to facilitate the competitive development of new virus- and gene-based drugs.
Collapse
Affiliation(s)
| | | | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
6
|
Zheng M, Huang J, Tong A, Yang H. Oncolytic Viruses for Cancer Therapy: Barriers and Recent Advances. MOLECULAR THERAPY-ONCOLYTICS 2019; 15:234-247. [PMID: 31872046 PMCID: PMC6911943 DOI: 10.1016/j.omto.2019.10.007] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oncolytic viruses (OVs) are powerful new therapeutic agents in cancer therapy. With the first OV (talimogene laherparepvec [T-vec]) obtaining US Food and Drug Administration approval, interest in OVs has been boosted greatly. Nevertheless, despite extensive research, oncolytic virotherapy has shown limited efficacy against solid tumors. Recent advances in viral retargeting, genetic editing, viral delivery platforms, tracking strategies, OV-based gene therapy, and combination strategies have the potential to broaden the applications of oncolytic virotherapy in oncology. In this review, we present several insights into the limitations and challenges of oncolytic virotherapy, describe the strategies mentioned above, provide a summary of recent preclinical and clinical trials in the field of oncolytic virotherapy, and highlight the need to optimize current strategies to improve clinical outcomes.
Collapse
Affiliation(s)
- Meijun Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Jianhan Huang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Aiping Tong
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Hui Yang
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| |
Collapse
|
7
|
Wu ZJ, Tang FR, Ma ZW, Peng XC, Xiang Y, Zhang Y, Kang J, Ji J, Liu XQ, Wang XW, Xin HW, Ren BX. Oncolytic Viruses for Tumor Precision Imaging and Radiotherapy. Hum Gene Ther 2018; 29:204-222. [PMID: 29179583 DOI: 10.1089/hum.2017.189] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In 2003 in China, Peng et al. invented the recombinant adenovirus expressing p53 (Gendicine) for clinical tumor virotherapy. This was the first clinically approved gene therapy and tumor virotherapy drug in the world. An oncolytic herpes simplex virus expressing granulocyte-macrophage colony-stimulating factor (Talimogene laherparepvec) was approved for melanoma treatment in the United States in 2015. Since then, oncolytic viruses have been attracting more and more attention in the field of oncology, and may become novel significant modalities of tumor precision imaging and radiotherapy after further improvement. Oncolytic viruses carrying reporter genes can replicate and express genes of interest selectively in tumor cells, thus improving in vivo noninvasive precision molecular imaging and radiotherapy. Here, the latest developments and molecular mechanisms of tumor imaging and radiotherapy using oncolytic viruses are reviewed, and perspectives are given for further research. Various types of tumors are discussed, and special attention is paid to gastrointestinal tumors.
Collapse
Affiliation(s)
- Zi J Wu
- 1 Laboratory of Oncology, Center for Molecular Medicine, Yangtze University , Jingzhou, China .,2 Department of Medical Imaging, School of Medicine, Yangtze University , Jingzhou, China .,3 The Second School of Clinical Medicine, Yangtze University , Jingzhou, China
| | - Feng R Tang
- 4 Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore , Create Tower, Singapore
| | - Zhao-Wu Ma
- 1 Laboratory of Oncology, Center for Molecular Medicine, Yangtze University , Jingzhou, China
| | - Xiao-Chun Peng
- 1 Laboratory of Oncology, Center for Molecular Medicine, Yangtze University , Jingzhou, China
| | - Ying Xiang
- 1 Laboratory of Oncology, Center for Molecular Medicine, Yangtze University , Jingzhou, China
| | - Yanling Zhang
- 5 Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Guangzhou, China .,6 School of Biotechnology, Southern Medical University , Guangzhou, China
| | - Jingbo Kang
- 7 The Navy General Hospital Tumor Diagnosis and Treatment Center , Beijing, China
| | - Jiafu Ji
- 8 Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute , Beijing, China
| | - Xiao Q Liu
- 1 Laboratory of Oncology, Center for Molecular Medicine, Yangtze University , Jingzhou, China .,2 Department of Medical Imaging, School of Medicine, Yangtze University , Jingzhou, China .,3 The Second School of Clinical Medicine, Yangtze University , Jingzhou, China
| | - Xian-Wang Wang
- 1 Laboratory of Oncology, Center for Molecular Medicine, Yangtze University , Jingzhou, China
| | - Hong-Wu Xin
- 1 Laboratory of Oncology, Center for Molecular Medicine, Yangtze University , Jingzhou, China
| | - Bo X Ren
- 2 Department of Medical Imaging, School of Medicine, Yangtze University , Jingzhou, China .,3 The Second School of Clinical Medicine, Yangtze University , Jingzhou, China
| |
Collapse
|
8
|
Yokoda R, Nagalo BM, Vernon B, Oklu R, Albadawi H, DeLeon TT, Zhou Y, Egan JB, Duda DG, Borad MJ. Oncolytic virus delivery: from nano-pharmacodynamics to enhanced oncolytic effect. Oncolytic Virother 2017; 6:39-49. [PMID: 29184854 PMCID: PMC5687448 DOI: 10.2147/ov.s145262] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
With the advancement of a growing number of oncolytic viruses (OVs) to clinical development, drug delivery is becoming an important barrier to overcome for optimal therapeutic benefits. Host immunity, tumor microenvironment and abnormal vascularity contribute to inefficient vector delivery. A number of novel approaches for enhanced OV delivery are under evaluation, including use of nanoparticles, immunomodulatory agents and complex viral–particle ligands along with manipulations of the tumor microenvironment. This field of OV delivery has quickly evolved to bioengineering of complex nanoparticles that could be deposited within the tumor using minimal invasive image-guided delivery. Some of the strategies include ultrasound (US)-mediated cavitation-enhanced extravasation, magnetic viral complexes delivery, image-guided infusions with focused US and targeting photodynamic virotherapy. In addition, strategies that modulate tumor microenvironment to decrease extracellular matrix deposition and increase viral propagation are being used to improve tumor penetration by OVs. Some involve modification of the viral genome to enhance their tumoral penetration potential. Here, we highlight the barriers to oncolytic viral delivery, and discuss the challenges to improving it and the perspectives of establishing new modes of active delivery to achieve enhanced oncolytic effects.
Collapse
Affiliation(s)
- Raquel Yokoda
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Bolni M Nagalo
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Brent Vernon
- Department of Biomedical Engineering, Arizona State University, Tempe
| | - Rahmi Oklu
- Division of Vascular and Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, AZ
| | - Hassan Albadawi
- Division of Vascular and Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, AZ
| | - Thomas T DeLeon
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Yumei Zhou
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Jan B Egan
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Dan G Duda
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Mitesh J Borad
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| |
Collapse
|
9
|
Haddad D. Genetically Engineered Vaccinia Viruses As Agents for Cancer Treatment, Imaging, and Transgene Delivery. Front Oncol 2017; 7:96. [PMID: 28589082 PMCID: PMC5440573 DOI: 10.3389/fonc.2017.00096] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 04/27/2017] [Indexed: 01/08/2023] Open
Abstract
Despite advances in technology, the formidable challenge of treating cancer, especially if advanced, still remains with no significant improvement in survival rates, even with the most common forms of cancer. Oncolytic viral therapies have shown great promise for the treatment of various cancers, with the possible advantages of stronger treatment efficacy compared to conventional therapy due to higher tumor selectivity, and less toxicity. They are able to preferentially and selectively propagate in cancer cells, consequently destroying tumor tissue mainly via cell lysis, while leaving non-cancerous tissues unharmed. Several wild-type and genetically engineered vaccinia virus (VACV) strains have been tested in both preclinical and clinical trials with promising results. Greater understanding and advancements in molecular biology have enabled the generation of genetically engineered oncolytic viruses for safer and more efficacious treatment, including arming VACVs with cytokines and immunostimulatory molecules, anti-angiogenic agents, and enzyme prodrug therapy, in addition to combining VACVs with conventional external and systemic radiotherapy, chemotherapy, immunotherapy, and other virus strains. Furthermore, novel oncolytic vaccinia virus strains have been generated that express reporter genes for the tracking and imaging of viral therapy and monitoring of therapeutic response. Further study is needed to unlock VACVs’ full potential as part of the future of cancer therapy.
Collapse
Affiliation(s)
- Dana Haddad
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
10
|
Tsoneva D, Stritzker J, Bedenk K, Zhang Q, Frentzen A, Cappello J, Fischer U, Szalay AA. Drug-Encoded Biomarkers for Monitoring Biological Therapies. PLoS One 2015; 10:e0137573. [PMID: 26348361 PMCID: PMC4562523 DOI: 10.1371/journal.pone.0137573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 08/18/2015] [Indexed: 11/19/2022] Open
Abstract
Blood tests are necessary, easy-to-perform and low-cost alternatives for monitoring of oncolytic virotherapy and other biological therapies in translational research. Here we assessed three candidate proteins with the potential to be used as biomarkers in biological fluids: two glucuronidases from E. coli (GusA) and Staphylococcus sp. RLH1 (GusPlus), and the luciferase from Gaussia princeps (GLuc). The three genes encoding these proteins were inserted individually into vaccinia virus GLV-1h68 genome under the control of an identical promoter. The three resulting recombinant viruses were used to infect tumor cells in cultures and human tumor xenografts in nude mice. In contrast to the actively secreted GLuc, the cytoplasmic glucuronidases GusA and GusPlus were released into the supernatants only as a result of virus-mediated oncolysis. GusPlus resulted in the most sensitive detection of enzyme activity under controlled assay conditions in samples containing as little as 1 pg/ml of GusPlus, followed by GusA (25 pg/ml) and GLuc (≥375 pg/ml). Unexpectedly, even though GusA had a lower specific activity compared to GusPlus, the substrate conversion in the serum of tumor-bearing mice injected with the GusA-encoding virus strains was substantially higher than that of GusPlus. This was attributed to a 3.2 fold and 16.2 fold longer half-life of GusA in the blood stream compared to GusPlus and GLuc respectively, thus a more sensitive monitor of virus replication than the other two enzymes. Due to the good correlation between enzymatic activity of expressed marker gene and virus titer, we conclude that the amount of the biomarker protein in the body fluid semiquantitatively represents the amount of virus in the infected tumors which was confirmed by low light imaging. We found GusA to be the most reliable biomarker for monitoring oncolytic virotherapy among the three tested markers.
Collapse
Affiliation(s)
- Desislava Tsoneva
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | - Jochen Stritzker
- Genelux Corporation, San Diego Science Center, San Diego, CA, United States of America
- Biocenter, University of Würzburg, Würzburg, Germany
- * E-mail: (JS); (AAS)
| | - Kristina Bedenk
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | - Qian Zhang
- Genelux Corporation, San Diego Science Center, San Diego, CA, United States of America
| | - Alexa Frentzen
- Genelux Corporation, San Diego Science Center, San Diego, CA, United States of America
| | - Joseph Cappello
- Genelux Corporation, San Diego Science Center, San Diego, CA, United States of America
| | - Utz Fischer
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
- Department of Radiation Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA, United States of America
| | - Aladar A. Szalay
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
- Department of Radiation Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA, United States of America
- * E-mail: (JS); (AAS)
| |
Collapse
|
11
|
Ady JW, Johnsen C, Mojica K, Heffner J, Love D, Pugalenthi A, Belin LJ, Chen NG, Yu YA, Szalay AA, Fong Y. Oncolytic gene therapy with recombinant vaccinia strain GLV-2b372 efficiently kills hepatocellular carcinoma. Surgery 2015; 158:331-8. [PMID: 26049609 DOI: 10.1016/j.surg.2015.03.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) commonly presents at a late stage when surgery is no longer a curative option. As such, novel therapies for advanced HCC are needed. Oncolytic viruses are a viable option for cancer therapy owing to their ability to specifically infect, replicate within, and kill cancer cells. In this study, we have investigated the ability of GLV-2b372, a novel light-emitting recombinant vaccinia virus derived from a wild-type Lister strain, to kill HCC. METHODS Four human HCC cell lines were assayed in vitro for infectivity and cytotoxicity. Viral replication was quantified via standard viral plaque assays. Flank HCC xenografts generated in athymic nude mice were treated with intratumoral GLV-2b372 to assess for tumor growth inhibition and viral biodistribution. RESULTS Infectivity occurred in a time- and concentration-dependent manner with 70% cell death in all cell lines by day 5. All cell lines supported efficient viral replication. At 25 days after infection, flank tumor volumes decreased by 50% whereas controls increased by 400%. Tumor tissue demonstrated substantial GLV-2b372 infection at 24 hours, 48 hours, and 2 weeks. CONCLUSION We demonstrate that GLV-2b372 efficiently kills human HCC in vitro and in vivo and is a viable treatment option for patients with HCC.
Collapse
Affiliation(s)
- Justin W Ady
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Clark Johnsen
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Kelly Mojica
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Jacqueline Heffner
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Damon Love
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Amudhan Pugalenthi
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Laurence J Belin
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Nanhai G Chen
- Genelux Corporation, San Diego Science Center, San Diego, CA; Department of Radiation Medicine and Applied Sciences, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA
| | - Yong A Yu
- Genelux Corporation, San Diego Science Center, San Diego, CA
| | - Aladar A Szalay
- Genelux Corporation, San Diego Science Center, San Diego, CA; Department of Radiation Medicine and Applied Sciences, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA; Department of Biochemistry, Rudolph Virchow Center for Experimental Biomedicine, and Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Yuman Fong
- Department of Surgery, City of Hope Medical Center, Duarte, CA.
| |
Collapse
|
12
|
Molecular imaging of oncolytic viral therapy. MOLECULAR THERAPY-ONCOLYTICS 2015; 1:14007. [PMID: 27119098 PMCID: PMC4782985 DOI: 10.1038/mto.2014.7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 03/09/2014] [Indexed: 01/25/2023]
Abstract
Oncolytic viruses have made their mark on the cancer world as a potential therapeutic option, with the possible advantages of reduced side effects and strengthened treatment efficacy due to higher tumor selectivity. Results have been so promising, that oncolytic viral treatments have now been approved for clinical trials in several countries. However, clinical studies may benefit from the ability to noninvasively and serially identify sites of viral targeting via molecular imaging in order to provide safety, efficacy, and toxicity information. Furthermore, molecular imaging of oncolytic viral therapy may provide a more sensitive and specific diagnostic technique to detect tumor origin and, more importantly, presence of metastases. Several strategies have been investigated for molecular imaging of viral replication broadly categorized into optical and deep tissue imaging, utilizing several reporter genes encoding for fluorescence proteins, conditional enzymes, and membrane protein and transporters. Various imaging methods facilitate molecular imaging, including computer tomography, magnetic resonance imaging, positron emission tomography, single photon emission CT, gamma-scintigraphy, and photoacoustic imaging. In addition, several molecular probes are used for medical imaging, which act as targeting moieties or signaling agents. This review will explore the preclinical and clinical use of in vivo molecular imaging of replication-competent oncolytic viral therapy.
Collapse
|
13
|
Gholami S, Marano A, Chen NG, Aguilar RJ, Frentzen A, Chen CH, Lou E, Fujisawa S, Eveno C, Belin L, Zanzonico P, Szalay A, Fong Y. A novel vaccinia virus with dual oncolytic and anti-angiogenic therapeutic effects against triple-negative breast cancer. Breast Cancer Res Treat 2014; 148:489-99. [PMID: 25391896 DOI: 10.1007/s10549-014-3180-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/23/2014] [Indexed: 12/31/2022]
Abstract
Vascular endothelial growth factor (VEGF) expression is higher in triple-negative breast cancers (TNBC) compared to other subtypes and is reported to predict incidence of distant metastases and shorter overall survival. We investigated the therapeutic impact of a vaccinia virus (VACV) GLV-1h164 (derived from its parent virus GLV-1h100), encoding a single-chain antibody (scAb) against VEGF (GLAF-2) in an orthotopic TNBC murine model. GLV-1h164 was tested against multiple TNBC cell lines. Viral infectivity, cytotoxicity, and replication were determined. Mammary fat pad tumors were generated in athymic nude mice using MDA-MB-231 cells. Xenografts were treated with GLV-1h164, GLV-1h100, or PBS and followed for tumor growth. Viral infectivity was time- and concentration-dependent. GLV-1h164 killed TNBC cell lines in a dose-dependent fashion with greater than 90% cytotoxicity within 4 days at a multiplicity of infection of 5.0. In vitro, cytotoxicity of GLV-1h164 was identical to GLV-1h100. GLV-1h164 replicated efficiently in all cell lines with an over 400-fold increase in copy numbers from the initial viral dose within 4 days. In vivo, mean tumor volumes after 2 weeks of treatment were 73, 191, and 422 mm(3) (GLV-1h164, GLV-1h100, and PBS, respectively) (p < 0.05). Both in vivo Doppler ultrasonography and immuno-staining showed decreased neo-angiogenesis in GLV-1h164-treated tumors compared to both GLV-1h100 and PBS controls (p < 0.05). This is the first study to demonstrate efficient combination of oncolytic and anti-angiogenic activity of a novel VACV on TNBC xenografts. Our results suggest that GLV-1h164 is a promising therapeutic agent that warrants testing for patients with TNBC.
Collapse
Affiliation(s)
- Sepideh Gholami
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Stritzker J, Huppertz S, Zhang Q, Geissinger U, Härtl B, Gentschev I, Szalay AA. Inducible gene expression in tumors colonized by modified oncolytic vaccinia virus strains. J Virol 2014; 88:11556-67. [PMID: 25056902 PMCID: PMC4178832 DOI: 10.1128/jvi.00681-14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/12/2014] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED Exogenous gene induction of therapeutic, diagnostic, and safety mechanisms could be a considerable improvement in oncolytic virotherapy. Here, we introduced a doxycycline-inducible promoter system (comprised of a tetracycline repressor, several promoter constructs, and a tet operator sequence) into oncolytic recombinant vaccinia viruses (rVACV), which were further characterized in detail. Experiments in cell cultures as well as in tumor-bearing mice were analyzed to determine the role of the inducible-system components. To accomplish this, we took advantage of the optical reporter construct, which resulted in the production of click-beetle luciferase as well as a red fluorescent protein. The results indicated that each of the system components could be used to optimize the induction rates and had an influence on the background expression levels. Depending on the given gene to be induced in rVACV-colonized tumors of patients, we discuss the doxycycline-inducible promoter system adjustment and further optimization. IMPORTANCE Oncolytic virotherapy of cancer can greatly benefit from the expression of heterologous genes. It is reasonable that some of those heterologous gene products could have detrimental effects either on the cancer patient or on the oncolytic virus itself if they are expressed at the wrong time or if the expression levels are too high. Therefore, exogenous control of gene expression levels by administration of a nontoxic inducer will have positive effects on the safety as well as the therapeutic outcome of oncolytic virotherapy. In addition, it paves the way for the introduction of new therapeutic genes into the genome of oncolytic viruses that could not have been tested otherwise.
Collapse
Affiliation(s)
- Jochen Stritzker
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany Genelux Corporation, San Diego Science Center, San Diego, California, USA
| | - Sascha Huppertz
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | - Qian Zhang
- Genelux Corporation, San Diego Science Center, San Diego, California, USA Department of Radiation Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
| | - Ulrike Geissinger
- Genelux Corporation, San Diego Science Center, San Diego, California, USA
| | - Barbara Härtl
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany Genelux GmbH, Bernried, Germany
| | - Ivaylo Gentschev
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany Genelux Corporation, San Diego Science Center, San Diego, California, USA
| | - Aladar A Szalay
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany Department of Radiation Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
15
|
Abstract
Current standard treatments of cancer can prolong survival of many cancer patients but usually do not effectively cure the disease. Oncolytic virotherapy is an emerging therapeutic for the treatment of cancer that exploits replication-competent viruses to selectively infect and destroy cancerous cells while sparing normal cells and tissues. Clinical and/or preclinical studies on oncolytic viruses have revealed that the candidate viruses being tested in trials are remarkably safe and offer potential for treating many classes of currently incurable cancers. Among these candidates are vaccinia and myxoma viruses, which belong to the family Poxviridae and possess promising oncolytic features. This article describes poxviruses that are being developed for oncolytic virotherapy and summarizes the outcomes of both clinical and preclinical studies. Additionally, studies demonstrating superior efficacy when poxvirus oncolytic virotherapy is combined with conventional therapies are described.
Collapse
Affiliation(s)
- Winnie M. Chan
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
16
|
Hofmann E, Weibel S, Szalay AA. Combination treatment with oncolytic Vaccinia virus and cyclophosphamide results in synergistic antitumor effects in human lung adenocarcinoma bearing mice. J Transl Med 2014; 12:197. [PMID: 25030093 PMCID: PMC4105246 DOI: 10.1186/1479-5876-12-197] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 07/10/2014] [Indexed: 12/27/2022] Open
Abstract
Background The capacity of the recombinant Vaccinia virus GLV-1h68 as a single agent to efficiently treat different human or canine cancers has been shown in several preclinical studies. Currently, its human safety and efficacy are investigated in phase I/II clinical trials. In this study we set out to evaluate the oncolytic activity of GLV-1h68 in the human lung adenocarcinoma cell line PC14PE6-RFP in cell cultures and analyzed the antitumor potency of a combined treatment strategy consisting of GLV-1h68 and cyclophosphamide (CPA) in a mouse model of PC14PE6-RFP lung adenocarcinoma. Methods PC14PE6-RFP cells were treated in cell culture with GLV-1h68. Viral replication and cell survival were determined by plaque assays and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, respectively. Subcutaneously implanted PC14PE6-RFP xenografts were treated by systemic injection of GLV-1h68, CPA or a combination of both. Tumor growth and viral biodistribution were monitored and immune-related antigen profiling of tumor lysates was performed. Results GLV-1h68 efficiently infected, replicated in and lysed human PC14PE6-RFP cells in cell cultures. PC14PE6-RFP tumors were efficiently colonized by GLV-1h68 leading to much delayed tumor growth in PC14PE6-RFP tumor-bearing nude mice. Combination treatment with GLV-1h68 and CPA significantly improved the antitumor efficacy of GLV-1h68 and led to an increased viral distribution within the tumors. Pro-inflammatory cytokines and chemokines were distinctly elevated in tumors of GLV-1h68-treated mice. Factors expressed by endothelial cells or present in the blood were decreased after combination treatment. A complete loss in the hemorrhagic phenotype of the PC14PE6-RFP tumors and a decrease in the number of blood vessels after combination treatment could be observed. Conclusions CPA and GLV-1h68 have synergistic antitumor effects on PC14PE6-RFP xenografts. We strongly suppose that in the PC14PE6-RFP model the enhanced tumor growth inhibition achieved by combining GLV-1h68 with CPA is due to an effect on the vasculature rather than an immunosuppressive action of CPA. These results provide evidence to support further preclinical studies of combining GLV-1h68 and CPA in other highly angiogenic tumor models. Moreover, data presented here demonstrate that CPA can be combined successfully with GLV-1h68 based oncolytic virus therapy and therefore might be promising as combination therapy in human clinical trials.
Collapse
Affiliation(s)
| | | | - Aladar A Szalay
- Department of Biochemistry, Biocenter, University of Wuerzburg, D-97074 Wuerzburg, Germany.
| |
Collapse
|
17
|
Jun KH, Gholami S, Song TJ, Au J, Haddad D, Carson J, Chen CH, Mojica K, Zanzonico P, Chen NG, Zhang Q, Szalay A, Fong Y. A novel oncolytic viral therapy and imaging technique for gastric cancer using a genetically engineered vaccinia virus carrying the human sodium iodide symporter. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:2. [PMID: 24383569 PMCID: PMC3883485 DOI: 10.1186/1756-9966-33-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 12/11/2013] [Indexed: 11/10/2022]
Abstract
Background Gastric cancers have poor overall survival despite recent advancements in early detection methods, endoscopic resection techniques, and chemotherapy treatments. Vaccinia viral therapy has had promising therapeutic potential for various cancers and has a great safety profile. We investigated the therapeutic efficacy of a novel genetically-engineered vaccinia virus carrying the human sodium iodide symporter (hNIS) gene, GLV-1 h153, on gastric cancers and its potential utility for imaging with 99mTc pertechnetate scintigraphy and 124I positron emission tomography (PET). Methods GLV-1 h153 was tested against five human gastric cancer cell lines using cytotoxicity and standard viral plaque assays. In vivo, subcutaneous flank tumors were generated in nude mice with human gastric cancer cells, MKN-74. Tumors were subsequently injected with either GLV-1 h153 or PBS and followed for tumor growth. 99mTc pertechnetate scintigraphy and 124I microPET imaging were performed. Results GFP expression, a surrogate for viral infectivity, confirmed viral infection by 24 hours. At a multiplicity of infection (MOI) of 1, GLV-1 h153 achieved > 90% cytotoxicity in MNK-74, OCUM-2MD3, and AGS over 9 days, and >70% cytotoxicity in MNK- 45 and TMK-1. In vivo, GLV-1 h153 was effective in treating xenografts (p < 0.001) after 2 weeks of treatment. GLV-1 h153-infected tumors were readily imaged by 99mTc pertechnetate scintigraphy and 124I microPET imaging 2 days after treatment. Conclusions GLV-1 h153 is an effective oncolytic virus expressing the hNIS protein that can efficiently regress gastric tumors and allow deep-tissue imaging. These data encourages its continued investigation in clinical settings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yuman Fong
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065 USA.
| |
Collapse
|
18
|
Ruiz-Hernández E, Hess M, Melen GJ, Theek B, Talelli M, Shi Y, Ozbakir B, Teunissen EA, Ramírez M, Moeckel D, Kiessling F, Storm G, Scheeren HW, Hennink WE, Szalay AA, Stritzker J, Lammers T. PEG-pHPMAm-based polymeric micelles loaded with doxorubicin-prodrugs in combination antitumor therapy with oncolytic vaccinia viruses. Polym Chem 2014:1674-1681. [PMID: 24518685 DOI: 10.1039/c3py01097j] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
An enzymatically activatable prodrug of doxorubicin was covalently coupled, using click-chemistry, to the hydrophobic core of poly(ethylene glycol)-b-poly[N-(2-hydroxypropyl)-methacrylamide-lactate] micelles. The release and cytotoxic activity of the prodrug was evaluated in vitro in A549 non-small-cell lung cancer cells after adding β-glucuronidase, an enzyme which is present intracellularly in lysosomes and extracellularly in necrotic areas of tumor lesions. The prodrug-containing micelles alone and in combination with standard and β-glucuronidase-producing oncolytic vaccinia viruses were also evaluated in vivo, in mice bearing A549 xenograft tumors. When combined with the oncolytic viruses, the micelles completely blocked tumor growth. Moreover, a significantly better antitumor efficacy as compared to virus treatment alone was observed when β-glucuronidase virus treated tumor-bearing mice received the prodrug-containing micelles. These findings show that combining tumor-targeted drug delivery systems with oncolytic vaccinia viruses holds potential for improving anticancer therapy.
Collapse
Affiliation(s)
- Eduardo Ruiz-Hernández
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Michael Hess
- Department of Biochemistry, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Gustavo J Melen
- Department of Hematooncology & Stem Cell Transplantation Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Benjamin Theek
- Department of Experimental Molecular Imaging, University Clinic and Helmholtz Center for Biomedical Engineering, Aachen, Germany
| | - Marina Talelli
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Department of Inorganic and Bioinorganic Chemistry, Facultad de Farmacia, Universidad Complutense de Madrid, Spain
| | - Yang Shi
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Burcin Ozbakir
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Erik A Teunissen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Manuel Ramírez
- Department of Hematooncology & Stem Cell Transplantation Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Diana Moeckel
- Department of Experimental Molecular Imaging, University Clinic and Helmholtz Center for Biomedical Engineering, Aachen, Germany
| | - Fabian Kiessling
- Department of Experimental Molecular Imaging, University Clinic and Helmholtz Center for Biomedical Engineering, Aachen, Germany
| | - Gert Storm
- Department of Controlled Drug Delivery, Targeted Therapeutics Section, University of Twente and MIRA Institute for Biomedical Engineering and Technical Medicine, Enschede, The Netherlands
| | - Hans W Scheeren
- Department of Organic Chemistry, Radboud Univ Nijmegen, Heyendaalse weg 135, 6525 AJ Nijmegen, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Aladar A Szalay
- Department of Biochemistry, Biocenter, University of Würzburg, 97074 Würzburg, Germany.,Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA.,Department of Radiation Oncology, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jochen Stritzker
- Department of Biochemistry, Biocenter, University of Würzburg, 97074 Würzburg, Germany.,Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA
| | - Twan Lammers
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Department of Experimental Molecular Imaging, University Clinic and Helmholtz Center for Biomedical Engineering, Aachen, Germany.,Department of Controlled Drug Delivery, Targeted Therapeutics Section, University of Twente and MIRA Institute for Biomedical Engineering and Technical Medicine, Enschede, The Netherlands
| |
Collapse
|
19
|
Buckel L, Advani SJ, Frentzen A, Zhang Q, Yu YA, Chen NG, Ehrig K, Stritzker J, Mundt AJ, Szalay AA. Combination of fractionated irradiation with anti-VEGF expressing vaccinia virus therapy enhances tumor control by simultaneous radiosensitization of tumor associated endothelium. Int J Cancer 2013; 133:2989-99. [PMID: 23729266 DOI: 10.1002/ijc.28296] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 04/29/2013] [Indexed: 02/04/2023]
Abstract
Oncolytic viruses are currently in clinical trials for a variety of tumors, including high grade gliomas. A characteristic feature of high grade gliomas is their high vascularity and treatment approaches targeting tumor endothelium are under investigation, including bevacizumab. The aim of this study was to improve oncolytic viral therapy by combining it with ionizing radiation and to radiosensitize tumor vasculature through a viral encoded anti-angiogenic payload. Here, we show how vaccinia virus-mediated expression of a single-chain antibody targeting VEGF resulted in radiosensitization of the tumor-associated vasculature. Cell culture experiments demonstrated that purified vaccinia virus encoded antibody targeting VEGF reversed VEGF-induced radioresistance specifically in endothelial cells but not tumor cells. In a subcutaneous model of U-87 glioma, systemically administered oncolytic vaccinia virus expressing anti-VEGF antibody (GLV-1h164) in combination with fractionated irradiation resulted in enhanced tumor growth inhibition when compared to nonanti-VEGF expressing oncolytic virus (GLV-1h68) and irradiation. Irradiation of tumor xenografts resulted in an increase in VACV replication of both GLV-1h68 and GLV-1h164. However, GLV-1h164 in combination with irradiation resulted in a drastic decrease in intratumoral VEGF levels and tumor vessel numbers in comparison to GLV-1h68 and irradiation. These findings demonstrate the incorporation of an oncolytic virus expressing an anti-VEGF antibody (GLV-1h164) into a fractionated radiation scheme to target tumor cells by enhanced VACV replication in irradiated tumors as well as to radiosensitize tumor endothelium which results in enhanced efficacy of combination therapy of human glioma xenografts.
Collapse
Affiliation(s)
- Lisa Buckel
- Department of Biochemistry, Rudolph Virchow Center for Experimental Biomedicine and Institute for Molecular Infection Biology, University of Würzburg, D-97074, Würzburg, Germany; Genelux Corporation, San Diego Science Center, San Diego, CA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Growth inhibition of different human colorectal cancer xenografts after a single intravenous injection of oncolytic vaccinia virus GLV-1h68. J Transl Med 2013; 11:79. [PMID: 23531320 PMCID: PMC3621142 DOI: 10.1186/1479-5876-11-79] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 03/20/2013] [Indexed: 12/18/2022] Open
Abstract
Background Despite availability of efficient treatment regimens for early stage colorectal cancer, treatment regimens for late stage colorectal cancer are generally not effective and thus need improvement. Oncolytic virotherapy using replication-competent vaccinia virus (VACV) strains is a promising new strategy for therapy of a variety of human cancers. Methods Oncolytic efficacy of replication-competent vaccinia virus GLV-1h68 was analyzed in both, cell cultures and subcutaneous xenograft tumor models. Results In this study we demonstrated for the first time that the replication-competent recombinant VACV GLV-1h68 efficiently infected, replicated in, and subsequently lysed various human colorectal cancer lines (Colo 205, HCT-15, HCT-116, HT-29, and SW-620) derived from patients at all four stages of disease. Additionally, in tumor xenograft models in athymic nude mice, a single injection of intravenously administered GLV-1h68 significantly inhibited tumor growth of two different human colorectal cell line tumors (Duke’s type A-stage HCT-116 and Duke’s type C-stage SW-620), significantly improving survival compared to untreated mice. Expression of the viral marker gene ruc-gfp allowed for real-time analysis of the virus infection in cell cultures and in mice. GLV-1h68 treatment was well-tolerated in all animals and viral replication was confined to the tumor. GLV-1h68 treatment elicited a significant up-regulation of murine immune-related antigens like IFN-γ, IP-10, MCP-1, MCP-3, MCP-5, RANTES and TNF-γ and a greater infiltration of macrophages and NK cells in tumors as compared to untreated controls. Conclusion The anti-tumor activity observed against colorectal cancer cells in these studies was a result of direct viral oncolysis by GLV-1h68 and inflammation-mediated innate immune responses. The therapeutic effects occurred in tumors regardless of the stage of disease from which the cells were derived. Thus, the recombinant vaccinia virus GLV-1h68 has the potential to treat colorectal cancers independently of the stage of progression.
Collapse
|
21
|
Evaluation of Norepinephrine Transporter Expression and Metaiodobenzylguanidine Avidity in Neuroblastoma: A Report from the Children's Oncology Group. INTERNATIONAL JOURNAL OF MOLECULAR IMAGING 2012; 2012:250834. [PMID: 23050139 PMCID: PMC3463166 DOI: 10.1155/2012/250834] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 08/23/2012] [Indexed: 11/18/2022]
Abstract
Purpose. (123)I-metaiodobenzylguanidine (MIBG) is used for the diagnostic evaluation of neuroblastoma. We evaluated the relationship between norepinephrine transporter (NET) expression and clinical MIBG uptake. Methods. Quantitative reverse transcription PCR (N = 82) and immunohistochemistry (IHC; N = 61) were performed for neuroblastoma NET mRNA and protein expression and correlated with MIBG avidity on diagnostic scans. The correlation of NET expression with clinical features was also performed. Results. Median NET mRNA expression level for the 19 MIBG avid patients was 12.9% (range 1.6-73.7%) versus 5.9% (range 0.6-110.0%) for the 8 nonavid patients (P = 0.31). Median percent NET protein expression was 50% (range 0-100%) in MIBG avid patients compared to 10% (range 0-80%) in nonavid patients (P = 0.027). MYCN amplified tumors had lower NET protein expression compared to nonamplified tumors (10% versus 50%; P = 0.0002). Conclusions. NET protein expression in neuroblastoma correlates with MIBG avidity. MYCN amplified tumors have lower NET protein expression.
Collapse
|
22
|
Haddad D, Chen CH, Carlin S, Silberhumer G, Chen NG, Zhang Q, Longo V, Carpenter SG, Mittra A, Carson J, Au J, Gonen M, Zanzonico PB, Szalay AA, Fong Y. Imaging characteristics, tissue distribution, and spread of a novel oncolytic vaccinia virus carrying the human sodium iodide symporter. PLoS One 2012; 7:e41647. [PMID: 22912675 PMCID: PMC3422353 DOI: 10.1371/journal.pone.0041647] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 06/27/2012] [Indexed: 11/18/2022] Open
Abstract
Introduction Oncolytic viruses show promise for treating cancer. However, to assess therapy and potential toxicity, a noninvasive imaging modality is needed. This study aims to determine the in vivo biodistribution, and imaging and timing characteristics of a vaccinia virus, GLV-1h153, encoding the human sodium iodide symporter (hNIS. Methods GLV-1h153 was modified from GLV-1h68 to encode the hNIS gene. Timing of cellular uptake of radioiodide 131I in human pancreatic carcinoma cells PANC-1 was assessed using radiouptake assays. Viral biodistribution was determined in nude mice bearing PANC-1 xenografts, and infection in tumors confirmed histologically and optically via Green Fluorescent Protein (GFP) and bioluminescence. Timing characteristics of enhanced radiouptake in xenografts were assessed via 124I-positron emission tomography (PET). Detection of systemic administration of virus was investigated with both 124I-PET and 99m-technecium gamma-scintigraphy. Results GLV-1h153 successfully facilitated time-dependent intracellular uptake of 131I in PANC-1 cells with a maximum uptake at 24 hours postinfection (P<0.05). In vivo, biodistribution profiles revealed persistence of virus in tumors 5 weeks postinjection at 109 plaque-forming unit (PFU)/gm tissue, with the virus mainly cleared from all other major organs. Tumor infection by GLV-1h153 was confirmed via optical imaging and histology. GLV-1h153 facilitated imaging virus replication in tumors via PET even at 8 hours post radiotracer injection, with a mean %ID/gm of 3.82±0.46 (P<0.05) 2 days after intratumoral administration of virus, confirmed via tissue radiouptake assays. One week post systemic administration, GLV-1h153-infected tumors were detected via 124I-PET and 99m-technecium-scintigraphy. Conclusion GLV-1h153 is a promising oncolytic agent against pancreatic cancer with a promising biosafety profile. GLV-1h153 facilitated time-dependent hNIS-specific radiouptake in pancreatic cancer cells, facilitating detection by PET with both intratumoral and systemic administration. Therefore, GLV-1h153 is a promising candidate for the noninvasive imaging of virotherapy and warrants further study into longterm monitoring of virotherapy and potential radiocombination therapies with this treatment and imaging modality.
Collapse
Affiliation(s)
- Dana Haddad
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Bavaria, Germany
| | - Chun-Hao Chen
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Sean Carlin
- Radiopharmaceutical Chemistry Service, Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Gerd Silberhumer
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Nanhai G. Chen
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Radiation Oncology, University of California, San Diego, California, United States of America
| | - Qian Zhang
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
| | - Valerie Longo
- Departments of Medical Physics and Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Susanne G. Carpenter
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Arjun Mittra
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Joshua Carson
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Joyce Au
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Pat B. Zanzonico
- Departments of Medical Physics and Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Aladar A. Szalay
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Bavaria, Germany
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Radiation Oncology, University of California, San Diego, California, United States of America
- * E-mail: (AAS); (YF)
| | - Yuman Fong
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * E-mail: (AAS); (YF)
| |
Collapse
|
23
|
Wang H, Chen NG, Minev BR, Szalay AA. Oncolytic vaccinia virus GLV-1h68 strain shows enhanced replication in human breast cancer stem-like cells in comparison to breast cancer cells. J Transl Med 2012; 10:167. [PMID: 22901246 PMCID: PMC3478222 DOI: 10.1186/1479-5876-10-167] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Accepted: 08/02/2012] [Indexed: 12/18/2022] Open
Abstract
Background Recent data suggest that cancer stem cells (CSCs) play an important role in cancer, as these cells possess enhanced tumor-forming capabilities and are responsible for relapses after apparently curative therapies have been undertaken. Hence, novel cancer therapies will be needed to test for both tumor regression and CSC targeting. The use of oncolytic vaccinia virus (VACV) represents an attractive anti-tumor approach and is currently under evaluation in clinical trials. The purpose of this study was to demonstrate whether VACV does kill CSCs that are resistant to irradiation and chemotherapy. Methods Cancer stem-like cells were identified and separated from the human breast cancer cell line GI-101A by virtue of increased aldehyde dehydrogenase 1 (ALDH1) activity as assessed by the ALDEFLUOR assay and cancer stem cell-like features such as chemo-resistance, irradiation-resistance and tumor-initiating were confirmed in cell culture and in animal models. VACV treatments were applied to both ALDEFLUOR-positive cells in cell culture and in xenograft tumors derived from these cells. Moreover, we identified and isolated CD44+CD24+ESA+ cells from GI-101A upon an epithelial-mesenchymal transition (EMT). These cells were similarly characterized both in cell culture and in animal models. Results We demonstrated for the first time that the oncolytic VACV GLV-1h68 strain replicated more efficiently in cells with higher ALDH1 activity that possessed stem cell-like features than in cells with lower ALDH1 activity. GLV-1h68 selectively colonized and eventually eradicated xenograft tumors originating from cells with higher ALDH1 activity. Furthermore, GLV-1h68 also showed preferential replication in CD44+CD24+ESA+ cells derived from GI-101A upon an EMT induction as well as in xenograft tumors originating from these cells that were more tumorigenic than CD44+CD24-ESA+ cells. Conclusions Taken together, our findings indicate that GLV-1h68 efficiently replicates and kills cancer stem-like cells. Thus, GLV-1h68 may become a promising agent for eradicating both primary and metastatic tumors, especially tumors harboring cancer stem-like cells that are resistant to chemo and/or radiotherapy and may be responsible for recurrence of tumors.
Collapse
Affiliation(s)
- Huiqiang Wang
- Institute of Biochemistry, Biocenter, University of Würzburg, Am hubland, D-97074, Würzburg, Germany
| | | | | | | |
Collapse
|
24
|
Real-time imaging of tumors using replication-competent light-emitting microorganisms. Methods Mol Biol 2012. [PMID: 22700410 DOI: 10.1007/978-1-61779-797-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Early detection of cancer and metastases is pivotal to the success of subsequent treatment intervention. In recent years, the use of live microorganisms, such as viruses and bacteria, has gained substantial research and clinical interest in both detection and therapy of cancer. Many of these live microorganisms have shown remarkable tumor-specific replication following systemic delivery. With the aid of modern molecular technologies, modified live microorganisms can be engineered to carry additional diagnostic and therapeutic capabilities. We have shown that when armed with light-emitting protein genes, such as genes for luciferase and green fluorescent protein, the entry and specific amplification of systemically-delivered vaccinia virus and bacteria in tumors can be visualized in real time using a low-light imager, or using macro- and micro-fluorescence microscopes. Therefore, through optical imaging, the location of tumors and metastases could be revealed by these light-emitting microorganisms. The tumor-colonization capability has been demonstrated in both immuno-competent as well as immuno-compromised rodent models with syngeneic and allogeneic tumors. Based on their "tumor-finding" nature, bacteria and viruses could be further designed as "vehicles" to carry multiple genes for detection and therapy of cancer.
Collapse
|
25
|
Hess M, Stritzker J, Härtl B, Sturm JB, Gentschev I, Szalay AA. Bacterial glucuronidase as general marker for oncolytic virotherapy or other biological therapies. J Transl Med 2011; 9:172. [PMID: 21989091 PMCID: PMC3207905 DOI: 10.1186/1479-5876-9-172] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 10/11/2011] [Indexed: 11/23/2022] Open
Abstract
Background Oncolytic viral tumor therapy is an emerging field in the fight against cancer with rising numbers of clinical trials and the first clinically approved product (Adenovirus for the treatment of Head and Neck Cancer in China) in this field. Yet, until recently no general (bio)marker or reporter gene was described that could be used to evaluate successful tumor colonization and/or transgene expression in other biological therapies. Methods Here, a bacterial glucuronidase (GusA) encoded by biological therapeutics (e.g. oncolytic viruses) was used as reporter system. Results Using fluorogenic probes that were specifically activated by glucuronidase we could show 1) preferential activation in tumors, 2) renal excretion of the activated fluorescent compounds and 3) reproducible detection of GusA in the serum of oncolytic vaccinia virus treated, tumor bearing mice in several tumor models. Time course studies revealed that reliable differentiation between tumor bearing and healthy mice can be done as early as 9 days post injection of the virus. Regarding the sensitivity of the newly developed assay system, we could show that a single infected tumor cell could be reliably detected in this assay. Conclusion GusA therefore has the potential to be used as a general marker in the preclinical and clinical evaluation of (novel) biological therapies as well as being useful for the detection of rare cells such as circulating tumor cells.
Collapse
Affiliation(s)
- Michael Hess
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Chen NG, Yu YA, Zhang Q, Szalay AA. Replication efficiency of oncolytic vaccinia virus in cell cultures prognosticates the virulence and antitumor efficacy in mice. J Transl Med 2011; 9:164. [PMID: 21951588 PMCID: PMC3192684 DOI: 10.1186/1479-5876-9-164] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 09/27/2011] [Indexed: 01/09/2023] Open
Abstract
Background We have shown that insertion of the three vaccinia virus (VACV) promoter-driven foreign gene expression cassettes encoding Renilla luciferase-Aequorea GFP fusion protein, β-galactosidase, and β-glucuronidase into the F14.5L, J2R, and A56R loci of the VACV LIVP genome, respectively, results in a highly attenuated mutant strain GLV-1h68. This strain shows tumor-specific replication and is capable of eradicating tumors with little or no virulence in mice. This study aimed to distinguish the contribution of added VACV promoter-driven transcriptional units as inserts from the effects of insertional inactivation of three viral genes, and to determine the correlation between replication efficiency of oncolytic vaccinia virus in cell cultures and the virulence and antitumor efficacy in mice Methods A series of recombinant VACV strains was generated by replacing one, two, or all three of the expression cassettes in GLV-1h68 with short non-coding DNA sequences. The replication efficiency and tumor cell killing capacity of these newly generated VACV strains were compared with those of the parent virus GLV-1h68 in cell cultures. The virus replication efficiency in tumors and antitumor efficacy as well as the virulence were evaluated in nu/nu (nude) mice bearing human breast tumor xenografts. Results we found that virus replication efficiency increased with removal of each of the expression cassettes. The increase in virus replication efficiency was proportionate to the strength of removed VACV promoters linked to foreign genes. The replication efficiency of the new VACV strains paralleled their cytotoxicity in cell cultures. The increased replication efficiency in tumor xenografts resulted in enhanced antitumor efficacy in nude mice. Similarly, the enhanced virus replication efficiency was indicative of increased virulence in nude mice. Conclusions These data demonstrated that insertion of VACV promoter-driven transcriptional units into the viral genome for the purpose of insertional mutagenesis did modulate the efficiency of virus replication together with antitumor efficacy as well as virulence. Replication efficiency of oncolytic VACV in cell cultures can predict the virulence and therapeutic efficacy in nude mice. These findings may be essential for rational design of safe and potent VACV strains for vaccination and virotherapy of cancer in humans and animals.
Collapse
Affiliation(s)
- Nanhai G Chen
- Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA
| | | | | | | |
Collapse
|
27
|
Gentschev I, Müller M, Adelfinger M, Weibel S, Grummt F, Zimmermann M, Bitzer M, Heisig M, Zhang Q, Yu YA, Chen NG, Stritzker J, Lauer UM, Szalay AA. Efficient colonization and therapy of human hepatocellular carcinoma (HCC) using the oncolytic vaccinia virus strain GLV-1h68. PLoS One 2011; 6:e22069. [PMID: 21779374 PMCID: PMC3133637 DOI: 10.1371/journal.pone.0022069] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 06/16/2011] [Indexed: 01/08/2023] Open
Abstract
Virotherapy using oncolytic vaccinia virus strains is one of the most promising new strategies for cancer therapy. In this study, we analyzed for the first time the therapeutic efficacy of the oncolytic vaccinia virus GLV-1h68 in two human hepatocellular carcinoma cell lines HuH7 and PLC/PRF/5 (PLC) in cell culture and in tumor xenograft models. By viral proliferation assays and cell survival tests, we demonstrated that GLV-1h68 efficiently colonized, replicated in, and did lyse these cancer cells in culture. Experiments with HuH7 and PLC xenografts have revealed that a single intravenous injection (i.v.) of mice with GLV-1h68 resulted in a significant reduction of primary tumor sizes compared to uninjected controls. In addition, replication of GLV-1h68 in tumor cells led to strong inflammatory and oncolytic effects resulting in intense infiltration of MHC class II-positive cells like neutrophils, macrophages, B cells and dendritic cells and in up-regulation of 13 pro-inflammatory cytokines. Furthermore, GLV-1h68 infection of PLC tumors inhibited the formation of hemorrhagic structures which occur naturally in PLC tumors. Interestingly, we found a strongly reduced vascular density in infected PLC tumors only, but not in the non-hemorrhagic HuH7 tumor model. These data demonstrate that the GLV-1h68 vaccinia virus may have an enormous potential for treatment of human hepatocellular carcinoma in man.
Collapse
Affiliation(s)
- Ivaylo Gentschev
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Meike Müller
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Marion Adelfinger
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Stephanie Weibel
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Friedrich Grummt
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Martina Zimmermann
- Department of Gastroenterology and Hepatology, Medical University Hospital, Tuebingen, Germany
| | - Michael Bitzer
- Department of Gastroenterology and Hepatology, Medical University Hospital, Tuebingen, Germany
| | - Martin Heisig
- Department of Internal Medicine, School of Medicine,Yale University, New Haven, Connecticut, United States of America
| | - Qian Zhang
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Radiation Oncology, Rebecca and John Moores Comprehensive Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Yong A. Yu
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Radiation Oncology, Rebecca and John Moores Comprehensive Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Nanhai G. Chen
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Radiation Oncology, Rebecca and John Moores Comprehensive Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Jochen Stritzker
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Ulrich M. Lauer
- Department of Gastroenterology and Hepatology, Medical University Hospital, Tuebingen, Germany
| | - Aladar A. Szalay
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
- Rudolf Virchow Center for Experimental Biomedicine, University of Wuerzburg, Wuerzburg, Germany
- Institute for Molecular Infection Biology, University of Wuerzburg, Wuerzburg, Germany
- Department of Radiation Oncology, Rebecca and John Moores Comprehensive Cancer Center, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
28
|
Haddad D, Chen NG, Zhang Q, Chen CH, Yu YA, Gonzalez L, Carpenter SG, Carson J, Au J, Mittra A, Gonen M, Zanzonico PB, Fong Y, Szalay AA. Insertion of the human sodium iodide symporter to facilitate deep tissue imaging does not alter oncolytic or replication capability of a novel vaccinia virus. J Transl Med 2011; 9:36. [PMID: 21453532 PMCID: PMC3080806 DOI: 10.1186/1479-5876-9-36] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 03/31/2011] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Oncolytic viruses show promise for treating cancer. However, to assess therapeutic efficacy and potential toxicity, a noninvasive imaging modality is needed. This study aimed to determine if insertion of the human sodium iodide symporter (hNIS) cDNA as a marker for non-invasive imaging of virotherapy alters the replication and oncolytic capability of a novel vaccinia virus, GLV-1h153. METHODS GLV-1h153 was modified from parental vaccinia virus GLV-1h68 to carry hNIS via homologous recombination. GLV-1h153 was tested against human pancreatic cancer cell line PANC-1 for replication via viral plaque assays and flow cytometry. Expression and transportation of hNIS in infected cells was evaluated using Westernblot and immunofluorescence. Intracellular uptake of radioiodide was assessed using radiouptake assays. Viral cytotoxicity and tumor regression of treated PANC-1tumor xenografts in nude mice was also determined. Finally, tumor radiouptake in xenografts was assessed via positron emission tomography (PET) utilizing carrier-free 124I radiotracer. RESULTS GLV-1h153 infected, replicated within, and killed PANC-1 cells as efficiently as GLV-1h68. GLV-1h153 provided dose-dependent levels of hNIS expression in infected cells. Immunofluorescence detected transport of the protein to the cell membrane prior to cell lysis, enhancing hNIS-specific radiouptake (P < 0.001). In vivo, GLV-1h153 was as safe and effective as GLV-1h68 in regressing pancreatic cancer xenografts (P < 0.001). Finally, intratumoral injection of GLV-1h153 facilitated imaging of virus replication in tumors via 124I-PET. CONCLUSION Insertion of the hNIS gene does not hinder replication or oncolytic capability of GLV-1h153, rendering this novel virus a promising new candidate for the noninvasive imaging and tracking of oncolytic viral therapy.
Collapse
Affiliation(s)
- Dana Haddad
- Department of Biochemistry, University of Wuerzburg, Wuerzburg D-97074, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Guse K, Cerullo V, Hemminki A. Oncolytic vaccinia virus for the treatment of cancer. Expert Opin Biol Ther 2011; 11:595-608. [DOI: 10.1517/14712598.2011.558838] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
30
|
In vitro and in vivo studies of adenovirus-mediated human norepinephrine transporter gene transduction to hepatocellular carcinoma. Cancer Gene Ther 2010; 18:196-205. [PMID: 21072068 DOI: 10.1038/cgt.2010.70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The clinical value of (131)I-MIBG for targeted imaging and targeted radiotherapy is limited to neural crest-derived tumors expressing human norepinephrine transporters (hNET) protein. To extend (131)I-MIBG-targeted therapy to other nonexpressed hNET tumors, this study investigated the hNET expression in vitro and in vivo in HepG2 hepatoma mediated by recombinant adenovirus encoding the hNET gene (Ad-hNET). For this purpose, the HepG2 cells showed a 4.87-fold increase in (125)I-MIBG uptake after infection with Ad-hNET, and the uptake of (125)I-MIBG could be specifically inhibited by maprotiline. Immunohistological analysis, in vivo biological study and (131)I-MIBG scintigraphic imaging also revealed the high expression of hNET protein in hepatoma. This in vitro and in vivo studies demonstrate the feasibility of hNET gene transfer, meditated by adenovirus vector, could extend to tumors other than those derived from the neural crest, which provides a sound foundation for further investigation of hepatocellular carcinoma-targeted radiotherapy mediated by adenovirus transfection with hNET gene.
Collapse
|
31
|
Abstract
Vaccinia virus (VACV) is arguably the most successful live biotherapeutic agent because of its critical role in the eradication of smallpox, one of the most deadly diseases in human history. VACV has been exploited as an oncolytic therapeutic agent for cancer since 1922. This virus selectively infects and destroys tumor cells, while sparing normal cells, both in cell cultures and in animal models. A certain degree of therapeutic efficacy also has been demonstrated in patients with different types of cancer. In recent years, several strategies have been successfully developed to further improve its tumor selectivity and antitumor efficacy. Oncolytic VACVs carrying imaging genes represent a new treatment strategy that combines tumor site-specific therapeutics with diagnostics (theranostics).
Collapse
Affiliation(s)
- Nanhai G Chen
- Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA; Genelux Corporation, San Diego Science Center, 3030 Bunker Hill Street, Suite 310, San Diego, CA 92109, USA
| | - Aladar A Szalay
- Rudolf Virchow Center for Experimental Biomedicine, Institute for Biochemistry & Institute for Molecular Infection Biology, University of Würzburg, Am Hubland, D-97074 Würzburg, Germany
- Department of Radiation Oncology, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
| |
Collapse
|
32
|
Bray M, Di Mascio M, de Kok-Mercado F, Mollura DJ, Jagoda E. Radiolabeled antiviral drugs and antibodies as virus-specific imaging probes. Antiviral Res 2010; 88:129-142. [PMID: 20709111 PMCID: PMC7125728 DOI: 10.1016/j.antiviral.2010.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Accepted: 08/09/2010] [Indexed: 12/04/2022]
Abstract
A number of small-molecule drugs inhibit viral replication by binding directly to virion structural proteins or to the active site of a viral enzyme, or are chemically modified by a viral enzyme before inhibiting a downstream process. Similarly, antibodies used to prevent or treat viral infections attach to epitopes on virions or on viral proteins expressed on the surface of infected cells. Such drugs and antibodies can therefore be thought of as probes for the detection of viral infections, suggesting that they might be used as radiolabeled tracers to visualize sites of viral replication by single-photon emission computed tomography (SPECT) or positron emission tomography (PET) imaging. A current example of this approach is the PET imaging of herpes simplex virus infections, in which the viral thymidine kinase phosphorylates radiolabeled thymidine analogues, trapping them within infected cells. One of many possible future applications might be the use of a radiolabeled hepatitis C protease inhibitor to image infection in animals or humans and provide a quantitative measure of viral burden. This article reviews the basic features of radionuclide imaging and the characteristics of ideal tracer molecules, and discusses how antiviral drugs and antibodies could be evaluated for their suitability as virus-specific imaging probes. The use of labeled drugs as low-dose tracers would provide an alternative application for compounds that have failed to advance to clinical use because of insufficient in vivo potency, an unsuitable pharmacokinetic profile or hepato- or nephrotoxicity.
Collapse
Affiliation(s)
- Mike Bray
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, MD 21702, United States
| | - Michele Di Mascio
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Fabian de Kok-Mercado
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, MD 21702, United States
| | - Daniel J Mollura
- Center for Infectious Disease Imaging, Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States
| | - Elaine Jagoda
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
33
|
Regression of human prostate tumors and metastases in nude mice following treatment with the recombinant oncolytic vaccinia virus GLV-1h68. J Biomed Biotechnol 2010; 2010:489759. [PMID: 20379368 PMCID: PMC2850154 DOI: 10.1155/2010/489759] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 10/14/2009] [Accepted: 01/13/2010] [Indexed: 01/04/2023] Open
Abstract
Virotherapy using oncolytic vaccinia virus strains is one of the most promising new strategies for cancer therapy. In the current study, we analyzed the therapeutic efficacy of the oncolytic vaccinia virus GLV-1h68 against two human prostate cancer cell lines DU-145 and PC-3 in cell culture and in tumor xenograft models. By viral proliferation assays and cell survival tests, we demonstrated that GLV-1h68 was able to infect, replicate in, and lyse these prostate cancer cells in culture. In DU-145 and PC-3 tumor xenograft models, a single intravenous injection with GLV-1h68 resulted in a significant reduction of primary tumor size. In addition, the GLV-1h68-infection led to strong inflammatory and oncolytic effects resulting in drastic reduction of regional lymph nodes with PC-3 metastases. Our data documented that the GLV-1h68 virus has a great potential for treatment of human prostate carcinoma.
Collapse
|
34
|
Yu Z, Li S, Brader P, Chen N, Yu YA, Zhang Q, Szalay AA, Fong Y, Wong RJ. Oncolytic vaccinia therapy of squamous cell carcinoma. Mol Cancer 2009; 8:45. [PMID: 19580655 PMCID: PMC2714037 DOI: 10.1186/1476-4598-8-45] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Accepted: 07/06/2009] [Indexed: 11/18/2022] Open
Abstract
Background Novel therapies are necessary to improve outcomes for patients with squamous cell carcinomas (SCC) of the head and neck. Historically, vaccinia virus was administered widely to humans as a vaccine and led to the eradication of smallpox. We examined the therapeutic effects of an attenuated, replication-competent vaccinia virus (GLV-1h68) as an oncolytic agent against a panel of six human head and neck SCC cell lines. Results All six cell lines supported viral transgene expression (β-galactosidase, green fluorescent protein, and luciferase) as early as 6 hours after viral exposure. Efficient transgene expression and viral replication (>150-fold titer increase over 72 hrs) were observed in four of the cell lines. At a multiplicity of infection (MOI) of 1, GLV-1h68 was highly cytotoxic to the four cell lines, resulting in ≥ 90% cytotoxicity over 6 days, and the remaining two cell lines exhibited >45% cytotoxicity. Even at a very low MOI of 0.01, three cell lines still demonstrated >60% cell death over 6 days. A single injection of GLV-1h68 (5 × 106 pfu) intratumorally into MSKQLL2 xenografts in mice exhibited localized intratumoral luciferase activity peaking at days 2–4, with gradual resolution over 10 days and no evidence of spread to normal organs. Treated animals exhibited near-complete tumor regression over a 24-day period without any observed toxicity, while control animals demonstrated rapid tumor progression. Conclusion These results demonstrate significant oncolytic efficacy by an attenuated vaccinia virus for infecting and lysing head and neck SCC both in vitro and in vivo, and support its continued investigation in future clinical trials.
Collapse
Affiliation(s)
- Zhenkun Yu
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|