1
|
Liao C, Engelberts P, van Dijk M, Timmermans A, Martens JWM, Neubert E, Danen EH. CD3xHER2 bsAb-Mediated Activation of Resting T-cells at HER2 Positive Tumor Clusters Is Sufficient to Trigger Bystander Eradication of Distant HER2 Negative Clusters Through IFNγ and TNFα. Eur J Immunol 2025; 55:e202451589. [PMID: 40178291 PMCID: PMC11967296 DOI: 10.1002/eji.202451589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
Bispecific antibodies (bsAbs) bridging CD3 on T-cells to tumor-associated antigens (TAA) on tumor cells can direct T-cell immunity to solid tumors. "Bystander killing", where T-cell targeting of TAA-positive tumor cells also leads to the eradication of TAA-negative cells, may overcome TAA heterogeneity. While bystander activity of activated, engineered T-cells has been shown to be robust and wide-reaching, spatiotemporal aspects of bsAb-mediated bystander activity are unresolved. Here, we developed a model where breast cancer tumoroids varying in HER2 expression were printed in to extracellular matrix (ECM) scaffolds. We generated (1) mixed tumors containing different ratios of HER2+ and HER2- tumor cells, and (2) HER2+ and HER2- tumoroids spaced at different distances within the ECM scaffold. Subsequently, tumors were exposed to peripheral blood-derived T-cells in the presence of CD3xHER2 bsAbs. We find that CD3xHER2 bsAb-mediated interaction of resting, nonactivated T-cells with HER2+ tumor cells is sufficient (1) to eliminate 50% HER2- cells in mixed tumor areas, and (2) to eradicate distant HER2- tumor areas. Such bystander killing involves paracrine IFNγ and TNFα activity but does not require T-cell accumulation in HER2- areas. These findings indicate that bystander eradication of TAA-negative cells can significantly contribute to bsAb therapy for solid tumors.
Collapse
Affiliation(s)
- Chen‐Yi Liao
- Leiden Academic Centre for Drug ResearchLeiden UniversityLeidenthe Netherlands
| | | | - Michiel van Dijk
- Leiden Academic Centre for Drug ResearchLeiden UniversityLeidenthe Netherlands
| | - Annemieke Timmermans
- Department of Medical OncologyErasmus MC Cancer InstituteErasmus University Medical CenterRotterdamthe Netherlands
| | - John W. M. Martens
- Department of Medical OncologyErasmus MC Cancer InstituteErasmus University Medical CenterRotterdamthe Netherlands
| | - Elsa Neubert
- Leiden Academic Centre for Drug ResearchLeiden UniversityLeidenthe Netherlands
| | - Erik H.J. Danen
- Leiden Academic Centre for Drug ResearchLeiden UniversityLeidenthe Netherlands
| |
Collapse
|
2
|
Dong C, Tan D, Sun H, Li Z, Zhang L, Zheng Y, Liu S, Zhang Y, He Q. Interleukin-12 Delivery Strategies and Advances in Tumor Immunotherapy. Curr Issues Mol Biol 2024; 46:11548-11579. [PMID: 39451566 PMCID: PMC11506767 DOI: 10.3390/cimb46100686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Interleukin-12 (IL-12) is considered to be a promising cytokine for enhancing an antitumor immune response; however, recombinant IL-12 has shown significant toxicity and limited efficacy in early clinical trials. Recently, many strategies for delivering IL-12 to tumor tissues have been developed, such as modifying IL-12, utilizing viral vectors, non-viral vectors, and cellular vectors. Previous studies have found that the fusion of IL-12 with extracellular matrix proteins, collagen, and immune factors is a way to enhance its therapeutic potential. In addition, studies have demonstrated that viral vectors are a good platform, and a variety of viruses such as oncolytic viruses, adenoviruses, and poxviruses have been used to deliver IL-12-with testing previously conducted in various cancer models. The local expression of IL-12 in tumors based on viral delivery avoids systemic toxicity while inducing effective antitumor immunity and acting synergistically with other therapies without compromising safety. In addition, lipid nanoparticles are currently considered to be the most mature drug delivery system. Moreover, cells are also considered to be drug carriers because they can effectively deliver therapeutic substances to tumors. In this article, we will systematically discuss the anti-tumor effects of IL-12 on its own or in combination with other therapies based on different delivery strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qing He
- State Key Laboratory of Drug Regulatory Sciences, National Institutes for Food and Drug Control, Beijing 102629, China; (C.D.); (D.T.); (H.S.); (Z.L.); (L.Z.); (Y.Z.); (S.L.); (Y.Z.)
| |
Collapse
|
3
|
Zarezadeh Mehrabadi A, Roozbahani F, Ranjbar R, Farzanehpour M, Shahriary A, Dorostkar R, Esmaeili Gouvarchin Ghaleh H. Overview of the pre-clinical and clinical studies about the use of CAR-T cell therapy of cancer combined with oncolytic viruses. World J Surg Oncol 2022; 20:16. [PMID: 35027068 PMCID: PMC8756705 DOI: 10.1186/s12957-021-02486-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/24/2021] [Indexed: 12/14/2022] Open
Abstract
Background Cancer is one of the critical issues of the global health system with a high mortality rate even with the available therapies, so using novel therapeutic approaches to reduce the mortality rate and increase the quality of life is sensed more than ever. Main body CAR-T cell therapy and oncolytic viruses are innovative cancer therapeutic approaches with fewer complications than common treatments such as chemotherapy and radiotherapy and significantly improve the quality of life. Oncolytic viruses can selectively proliferate in the cancer cells and destroy them. The specificity of oncolytic viruses potentially maintains the normal cells and tissues intact. T-cells are genetically manipulated and armed against the specific antigens of the tumor cells in CAR-T cell therapy. Eventually, they are returned to the body and act against the tumor cells. Nowadays, virology and oncology researchers intend to improve the efficacy of immunotherapy by utilizing CAR-T cells in combination with oncolytic viruses. Conclusion Using CAR-T cells along with oncolytic viruses can enhance the efficacy of CAR-T cell therapy in destroying the solid tumors, increasing the permeability of the tumor cells for T-cells, reducing the disturbing effects of the immune system, and increasing the success chance in the treatment of this hazardous disease. In recent years, significant progress has been achieved in using oncolytic viruses alone and in combination with other therapeutic approaches such as CAR-T cell therapy in pre-clinical and clinical investigations. This principle necessitates a deeper consideration of these treatment strategies. This review intends to curtly investigate each of these therapeutic methods, lonely and in combination form. We will also point to the pre-clinical and clinical studies about the use of CAR-T cell therapy combined with oncolytic viruses.
Collapse
Affiliation(s)
- Ali Zarezadeh Mehrabadi
- Immunology Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Roozbahani
- Department of Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Ranjbar
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahdieh Farzanehpour
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ruhollah Dorostkar
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
4
|
Wang Z, Zhang X. Adenovirus vector-attributed hepatotoxicity blocks clinical application in gene therapy. Cytotherapy 2021; 23:1045-1052. [PMID: 34548241 DOI: 10.1016/j.jcyt.2021.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 02/07/2023]
Abstract
Adenoviruses (Ads), common self-limiting pathogens in humans and animals, usually cause conjunctivitis, mild upper respiratory tract infection or gastroenteritis in humans and hepatotoxicity syndrome in chickens and dogs, posing great threats to public health and livestock husbandry. Artificially modified Ads, which wipe out virulence-determining genes, are the most frequently used viral vectors in gene therapy, and some Ad vector (AdV)-related medicines and vaccines have been licensed and applied. Inherent liver tropism enables AdVs to specifically deliver drugs/genes to the liver; however, AdVs are closely associated with acute hepatotoxicity in immunocompromised individuals, and the side effects of AdVs, which stimulate a strong inflammatory reaction in the liver and cause acute hepatotoxicity, have largely limited clinical application. Therefore, this review systematically elucidates the intimate relationship between AdVs and hepatotoxicity in terms of virus and host and precisely illustrates the accumulated understanding in this field over the past decades. This review demonstrates the liver tropism of AdVs and molecular mechanism of AdV-induced hepatotoxicity and looks at the studies on AdV-mediated animal hepatotoxicity, which will undoubtedly deepen the understanding of AdV-caused liver injury and be of benefit in the further safe development of AdVs.
Collapse
Affiliation(s)
- Zeng Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.
| | - Xiaozhan Zhang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| |
Collapse
|
5
|
Cervera-Carrascon V, Havunen R, Hemminki A. Oncolytic adenoviruses: a game changer approach in the battle between cancer and the immune system. Expert Opin Biol Ther 2019; 19:443-455. [PMID: 30905206 DOI: 10.1080/14712598.2019.1595582] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Oncolytic adenoviruses are among the most studied oncolytic viruses because of their tumor selectivity, safety, and transgene-delivery capability. With a growing number of different immunotherapies against cancer, the extraordinary immunogenicity of the adenovirus has emerged as a differentiating strength. Enabling T-cell related therapies with oncolytic adenoviruses appears a promising approach due to its inherent ability to elicit responses from the adaptive immune compartment. AREAS COVERED These viruses have successfully enhanced both adoptive T-cell therapies and immune-checkpoint therapies. Oncolytic viruses induce several effects at the tumor and on the systemic level that help to circumvent current limitations of T-cells and related therapies, such as T-cell trafficking, tumor immune suppressivity and antigen spreading EXPERT OPINION Taking into account the multitude of possibilities of treating cancer with immunotherapies, learning to optimize the combinations and administration strategies of these drugs, could lead to durable responses in patients with currently incurable cancers.
Collapse
Affiliation(s)
- Victor Cervera-Carrascon
- a Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine , University of Helsinki , Helsinki , Finland.,b TILT Biotherapeutics Ltd , Helsinki , Finland
| | - Riikka Havunen
- a Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine , University of Helsinki , Helsinki , Finland.,b TILT Biotherapeutics Ltd , Helsinki , Finland
| | - Akseli Hemminki
- a Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine , University of Helsinki , Helsinki , Finland.,b TILT Biotherapeutics Ltd , Helsinki , Finland.,c Hospital Comprehensive Cancer Center , Helsinki University , Helsinki , Finland
| |
Collapse
|
6
|
Hao S, Du X, Song Y, Ren M, Yang Q, Wang A, Wang Q, Zhao H, Du Z, Zhang G. Targeted gene therapy of the HSV-TK/hIL-12 fusion gene controlled by the hSLPI gene promoter of human non-small cell lung cancer in vitro. Oncol Lett 2018; 15:6503-6512. [PMID: 29731853 DOI: 10.3892/ol.2018.8148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 11/07/2017] [Indexed: 12/12/2022] Open
Abstract
The incidence of lung cancer and lung cancer-associated mortality have markedly increased worldwide, and gene-targeted therapy has emerged as a promising treatment strategy. The present study aimed to explore the targeted antitumor effect of the herpes simplex virus-thymidine kinase/human interleukin-12 (HSV-TK/hIL-12) fusion gene regulated by the human secretory leukocyte protease inhibitor (hSLPI) promoter of human non-small cell lung cancer (hNSCLC). There were four recombinant eukaryotic expression vectors: pcDNA3.1-CMV-TK, pcDNA3.1-CMV-TK/hIL-12, pcDNA3.1-phSLP-TK and pcDNA3.1-phSLP-TK/hIL-12. These were constructed and transfected into the A549, SPC-A1 and HepG2 cell lines in vitro. The expression of the HSV-TK/hIL-12 fusion gene was detected with reverse transcription-polymerase chain reaction (RT-PCR), and the content of hIL-12 was measured using an ELISA. The antitumor effect of the fusion gene on the A549, SPC-A1 and HepG2 cell lines was determined using an MTT assay. Analysis of the experimental data demonstrated that genes regulated by the cytomegalovirus promoter were expressed at the same level in three different tumor cell lines. Genes regulated by the hSLPI promoter were expressed in the A549 and SPC-A1 cell lines, but not in the HepG2 cell line. Coincidentally, the hIL-12 expression levels were similar to those observed in previous RT-PCR findings. In the Pcmv-TK/Pcmv-TK-hIL-12 group for all three cell lines, as well as in the PSLPI-TK/PSLPI-TK-hIL-12 group for the A549 and SPC-A1 cell lines, the cell survival rate declined significantly and the fusion gene transfection group indicated a lower cell survival rate, when compared with single gene transfection group. The present study indicated that the fusion gene regulated by the hSLPI promoter had a targeted antitumor effect on hNSCLC, and that the combined suicide gene and immune gene therapy had a stronger antitumor effect, compared with single gene therapy.
Collapse
Affiliation(s)
- Shuhong Hao
- Department of Hematology and Oncology, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaoyuan Du
- Department of Pathology, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Yang Song
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China
| | - Ming Ren
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China
| | - Qiwei Yang
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China.,Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ao Wang
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China
| | - Qingyu Wang
- The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China.,Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Haiyue Zhao
- Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhenwu Du
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China.,Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guizhen Zhang
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China.,Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
7
|
Hernandez-Alcoceba R, Poutou J, Ballesteros-Briones MC, Smerdou C. Gene therapy approaches against cancer using in vivo and ex vivo gene transfer of interleukin-12. Immunotherapy 2016; 8:179-98. [PMID: 26786809 DOI: 10.2217/imt.15.109] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IL-12 is an immunostimulatory cytokine with strong antitumor properties. Systemic administration of IL-12 in cancer patients led to severe toxic effects, prompting the development of gene therapy vectors able to express this cytokine locally in tumors. Both nonviral and viral vectors have demonstrated a high antitumor efficacy in preclinical tumor models. Some of these vectors, including DNA electroporation, adenovirus and ex vivo transduced dendritic cells, were tested in patients, showing low toxicity and moderate antitumor efficacy. IL-12 activity can be potentiated by molecules with immunostimulatory, antiangiogenic or cytotoxic activity. These combination therapies are of clinical interest because they could lower the threshold for IL-12 efficacy, increasing the therapeutic potential of gene therapy and preventing the toxicity mediated by this cytokine.
Collapse
Affiliation(s)
- Ruben Hernandez-Alcoceba
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| | - Joanna Poutou
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| | - María Cristina Ballesteros-Briones
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| | - Cristian Smerdou
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| |
Collapse
|
8
|
Higashi K, Hazama S, Araki A, Yoshimura K, Iizuka N, Yoshino S, Noma T, Oka M. A novel cancer vaccine strategy with combined IL-18 and HSV-TK gene therapy driven by the hTERT promoter in a murine colorectal cancer model. Int J Oncol 2014; 45:1412-20. [PMID: 25051201 PMCID: PMC4151806 DOI: 10.3892/ijo.2014.2557] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/20/2014] [Indexed: 01/17/2023] Open
Abstract
A therapeutic vaccine against minimal residual cancer cells is needed for the treatment of patients with colorectal cancer. Several gene therapy studies have revealed that the combination of a suicide gene and cytokine gene might induce effective antitumor immunity. In this study, we constructed an interleukin (IL)-18 and herpes simplex virus-thymidine kinase (HSV-TK) expression vector driven by the human telomerase reverse transcriptase (hTERT) promoter to study the efficacy of combination gene therapy with IL-18 and the HSV-TK suicide gene. Low immunogenic colon 26 cells were used for transfection and inoculation into syngeneic BALB/c mice. Large established tumors of colon 26 transfectants expressing IL-18 and HSV-TK driven by the hTERT promoter were completely eradicated after GCV administration in syngeneic BALB/c mice. Immunohistochemical analysis at the tumor rejection sites revealed enormous infiltrations of CD8+ T lymphocytes as well as CD4+ T lymphocytes and CD11b+ monocytes. Moreover, established distant tumors were completely eradicated by vaccination with the IL-18 and HSV-TK transfectants in combination with GCV. These data suggest that the IL-18 and suicide gene therapy can elicit antitumor specific immunity. In conclusion, gene therapy with IL-18 and HSV-TK plasmid vector driven by the hTERT promoter may be useful for cancer vaccination.
Collapse
Affiliation(s)
- Kosuke Higashi
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Shoichi Hazama
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Atsuhiro Araki
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Kiyoshi Yoshimura
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Norio Iizuka
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Shigefumi Yoshino
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Takafumi Noma
- Department of Molecular Biology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Masaaki Oka
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
9
|
Cerullo V, Koski A, Vähä-Koskela M, Hemminki A. Chapter eight--Oncolytic adenoviruses for cancer immunotherapy: data from mice, hamsters, and humans. Adv Cancer Res 2013; 115:265-318. [PMID: 23021247 DOI: 10.1016/b978-0-12-398342-8.00008-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adenovirus is one of the most commonly used vectors for gene therapy and two products have already been approved for treatment of cancer in China (Gendicine(R) and Oncorine(R)). An intriguing aspect of oncolytic adenoviruses is that by their very nature they potently stimulate multiple arms of the immune system. Thus, combined tumor killing via oncolysis and inherent immunostimulatory properties in fact make these viruses in situ tumor vaccines. When further engineered to express cytokines, chemokines, tumor-associated antigens, or other immunomodulatory elements, they have been shown in various preclinical models to induce antigen-specific effector and memory responses, resulting both in full therapeutic cures and even induction of life-long tumor immunity. Here, we review the state of the art of oncolytic adenovirus, in the context of their capability to stimulate innate and adaptive arms of the immune system and finally how we can modify these viruses to direct the immune response toward cancer.
Collapse
Affiliation(s)
- Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Division of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| | | | | | | |
Collapse
|
10
|
Chi J, Wang F, Li L, Feng D, Qin J, Xie F, Zhou F, Chen Y, Wang J, Yao K. The role of MAPK in CD4(+) T cells toll-like receptor 9-mediated signaling following HHV-6 infection. Virology 2011; 422:92-8. [PMID: 22055432 DOI: 10.1016/j.virol.2011.09.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 09/07/2011] [Accepted: 09/28/2011] [Indexed: 10/15/2022]
Abstract
Human herpesvirus-6 (HHV-6) is an important immunosuppressive and immunomodulatory virus that primarily infects immune cells (mainly CD4(+) T cells) and strongly suppresses the proliferation of infected cells. Toll-like receptors are pattern-recognition receptors essential for the development of an appropriate innate immune defense against infection. To understand the role of CD4(+) T cells in the innate response to HHV-6 infection and the involvement of TLRs, we used an in vitro infection model and observed that the infection of CD4(+) T cells resulted in the activation of JNK/SAPK via up-regulation of toll-like receptor 9 (TLR9). Associated with JNK activation, annexin V-PI staining indicated that HHV-6A was a strong inducer of apoptosis. Apoptotic response associated cytokines, IL-6 and TNF-α also induced by HHV-6A infection.
Collapse
Affiliation(s)
- Jing Chi
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Singh P, Yam M, Russell PJ, Khatri A. Molecular and traditional chemotherapy: a united front against prostate cancer. Cancer Lett 2010; 293:1-14. [PMID: 20117879 DOI: 10.1016/j.canlet.2009.11.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 11/23/2009] [Accepted: 11/27/2009] [Indexed: 01/28/2023]
Abstract
Castrate resistant prostate cancer (CRPC) is essentially incurable. Recently though, chemotherapy demonstrated a survival benefit ( approximately 2months) in the treatment of CRPC. While this was a landmark finding, suboptimal efficacy and systemic toxicities at the therapeutic doses warranted further development. Smart combination therapies, acting through multiple mechanisms to target the heterogeneous cell populations of PC and with potential for reduction in individual dosing, need to be developed. In that, targeted molecular chemotherapy has generated significant interest with the potential for localized treatment to generate systemic efficacy. This can be further enhanced through the use of oncolytic conditionally replicative adenoviruses (CRAds) to deliver molecular chemotherapy. The prospects of chemotherapy and molecular-chemotherapy as single and as components of combination therapies are discussed.
Collapse
Affiliation(s)
- P Singh
- Centre for Medicine and Oral Health, Griffith University - Gold Coast GH1, High Street, Southport, Gold Coast, QLD 4215, Australia
| | | | | | | |
Collapse
|
12
|
Liu WQ, Yang J, Dong J. Effects of HSV-TK+GFP/GCV suicide gene therapy system on mouse pancreatic cancer cells. Shijie Huaren Xiaohua Zazhi 2009; 17:1498-1503. [DOI: 10.11569/wcjd.v17.i15.1498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study in vitro therapeutic effect on mouse pancreatic cancer, as well as the bystander effect with HSV-TK suicide gene in combination with prodrug GCV.
METHODS: HSV-TK and GFP were inserted into pcDNA3.1 (+) to construct pcDNA3.1+/HSV-TK+GFP, and pcDNA3.1+/HSV-TK+GFP was transferred into mouse pancreatic cancer cell MPC by Lipofectin. We then added GCV to these gene-modified cells and studied the sensitivity of the cells to GCV as well as the bystander effect.
RESULTS: The gene modified pancreatic cancer cells MPC/HSV-TK+GFP were successfully developed. In vitro experiments showed that when the MPC/HSV-TK+GFP cells accounted for 10% of hybrid cells, the low concentration (20 mg/L) of GCV was about 50% of tumor cell killing. In vivo results showed that the low concentration of GCV killed the cells. And tumor growth of the mouse model was inhibited.
CONCLUSION: Our data demonstrate MPC/HSV-TK+GFP cells are sensitive to the treatment of GCV compared with unmodified tumor cells, and remarkable bystander effect is seen.
Collapse
|
13
|
Abstract
Type 3 (T3) reovirus strains induce apoptotic neuronal cell death and lethal encephalitis in infected mice. T3 strain Dearing (T3D)-induced apoptosis in primary neuronal cultures occurs by a Fas-mediated mechanism and requires the activation of caspase 8. We now show that Fas mRNA is upregulated in the brains of mice infected with encephalitic reovirus T3D and T3 strain Abney (T3A) but not following infection with nonencephalitic reovirus type 1 strain Lang. Fas is upregulated in regions of the brain that are injured during infection with T3 reovirus strains and colocalizes with virus antigen in individual neurons. In contrast, levels of FasL mRNA induced by encephalitic and nonencephalitic reovirus strains do not differ significantly. Caspase 8, the initiator caspase associated with Fas-mediated apoptosis, is activated in the cortex and hippocampal regions of both T3D- and T3A-infected mice. Furthermore, Bid cleavage and the activation of caspase 9 in the brains of T3D-infected mice suggest that the caspase 8-dependent activation of mitochondrial apoptotic signaling contributes to virus-induced apoptosis. We have previously shown that the inhibition of c-Jun N-terminal kinase (JNK) signaling blocks T3D-induced apoptosis and improves the outcome of virus-induced encephalitis. We now show that the reovirus-induced upregulation of Fas requires JNK signaling, thereby providing a link between reovirus-induced death receptor signaling and mitogen-activated protein kinase pathways and a potential mechanism for the therapeutic action of JNK inhibition.
Collapse
|
14
|
Induction of protective antitumor activity of tumor lysate-pulsed dendritic cells vaccine in RM-1 prostate cancer mode. ACTA ACUST UNITED AC 2009. [DOI: 10.1016/s1000-1948(09)60012-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
15
|
TNF-alpha mediates pseudorabies virus-induced apoptosis via the activation of p38 MAPK and JNK/SAPK signaling. Virology 2008; 381:55-66. [PMID: 18799179 DOI: 10.1016/j.virol.2008.08.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 08/07/2008] [Accepted: 08/14/2008] [Indexed: 11/24/2022]
Abstract
PRV infection causes apoptosis in vitro and in vivo. However, the significance of PRV-induced apoptosis and its signaling pathways is still unknown. This work investigates the role of MAPK pathways in mediating PRV-induced apoptosis. Flow cytometry, apoptosis ELISA and western blotting using antibodies against cleaved caspase-3, -6 and PARP demonstrated that PRV induces apoptosis in a time- and dose-dependent manner. p38 and JNK/SAPK inhibitors significantly protected cells from PRV-induced apoptosis. Inhibitor treatment did not affect Us3a gene transcription and progeny virus production. Western blotting revealed that PRV activates p38 and JNK/SAPK signaling. Inhibition of NF-kappaB had no effect on PRV-mediated apoptosis. Non-replicative PRV failed to activate p38 and JNK/SAPK or induce apoptosis. PRV infection increases TNF-alpha transcription, translation and secretion, as well as TNF-alpha receptor expression. Inhibition of p38 and JNK/SAPK reduced PRV-induced TNF-alpha up-regulation. Neutralization assay confirmed that TNF-alpha is a key mediator involved in PRV-induced apoptosis.
Collapse
|
16
|
Stoff-Khalili MA, Rivera AA, Nedeljkovic-Kurepa A, DeBenedetti A, Li XL, Odaka Y, Podduturi J, Sibley DA, Siegal GP, Stoff A, Young S, Zhu ZB, Curiel DT, Mathis JM. Cancer-specific targeting of a conditionally replicative adenovirus using mRNA translational control. Breast Cancer Res Treat 2008; 108:43-55. [PMID: 17508279 PMCID: PMC2268614 DOI: 10.1007/s10549-007-9587-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2007] [Accepted: 03/26/2007] [Indexed: 12/14/2022]
Abstract
BACKGROUND In view of the limited success of available treatment modalities for a wide array of cancer, alternative and complementary therapeutic strategies need to be developed. Virotherapy employing conditionally replicative adenoviruses (CRAds) represents a promising targeted intervention relevant to a wide array of neoplastic diseases. Critical to the realization of an acceptable therapeutic index using virotherapy in clinical trials is the achievement of oncolytic replication in tumor cells, while avoiding non-specific replication in normal tissues. In this report, we exploited cancer-specific control of mRNA translation initiation in order to achieve enhanced replicative specificity of CRAd virotherapy agents. Heretofore, the achievement of replicative specificity of CRAd agents has been accomplished either by viral genome deletions or incorporation of tumor selective promoters. In contrast, control of mRNA translation has not been exploited for the design of tumor specific replicating viruses to date. We show herein, the utility of a novel approach that combines both transcriptional and translational regulation strategies for the key goal of replicative specificity. METHODS We describe the construction of a CRAd with cancer specific gene transcriptional control using the CXCR4 gene promoter (TSP) and cancer specific mRNA translational control using a 5'-untranslated region (5'-UTR) element from the FGF-2 (Fibroblast Growth Factor-2) mRNA. RESULTS Both in vitro and in vivo studies demonstrated that our CRAd agent retains anti-tumor potency. Importantly, assessment of replicative specificity using stringent tumor and non-tumor tissue slice systems demonstrated significant improvement in tumor selectivity. CONCLUSIONS Our study addresses a conceptually new paradigm: dual targeting of transgene expression to cancer cells using both transcriptional and mRNA translational control. Our novel approach addresses the key issue of replicative specificity and can potentially be generalized to a wide array of tumor types, whereby tumor selective patterns of gene expression and mRNA translational control can be exploited.
Collapse
Affiliation(s)
- Mariam A. Stoff-Khalili
- Division of Human Gene Therapy, Departments of Medicine, Surgery, Pathology and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
- Department of Obstetrics and Gynecology, University of Duesseldorf, Medical Center, 40225 Duesseldorf, Germany
| | - Angel A. Rivera
- Division of Human Gene Therapy, Departments of Medicine, Surgery, Pathology and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
| | - Ana Nedeljkovic-Kurepa
- Gene Therapy Program, Departments of Cellular Biology and Anatomy and Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130 USA
| | - Arrigo DeBenedetti
- Gene Therapy Program, Departments of Cellular Biology and Anatomy and Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130 USA
| | - Xiao-Lin Li
- Gene Therapy Program, Departments of Cellular Biology and Anatomy and Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130 USA
| | - Yoshinobu Odaka
- Gene Therapy Program, Departments of Cellular Biology and Anatomy and Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130 USA
| | - Jagat Podduturi
- Gene Therapy Program, Departments of Cellular Biology and Anatomy and Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130 USA
| | - Don A. Sibley
- Gene Therapy Program, Departments of Cellular Biology and Anatomy and Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130 USA
| | - Gene P. Siegal
- Division of Human Gene Therapy, Departments of Medicine, Surgery, Pathology and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
| | - Alexander Stoff
- Division of Human Gene Therapy, Departments of Medicine, Surgery, Pathology and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
- Department of Plastic and Reconstructive Surgery, Dreifaltigkeits-Hospital, 50389 Wesseling, Germany
| | - Scott Young
- Division of Human Gene Therapy, Departments of Medicine, Surgery, Pathology and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
| | - Zheng B. Zhu
- Division of Human Gene Therapy, Departments of Medicine, Surgery, Pathology and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
| | - David T. Curiel
- Division of Human Gene Therapy, Departments of Medicine, Surgery, Pathology and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
| | - J. Michael Mathis
- Gene Therapy Program, Departments of Cellular Biology and Anatomy and Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130 USA
| |
Collapse
|
17
|
Tsuchiyama T, Nakamoto Y, Sakai Y, Marukawa Y, Kitahara M, Mukaida N, Kaneko S. Prolonged, NK cell-mediated antitumor effects of suicide gene therapy combined with monocyte chemoattractant protein-1 against hepatocellular carcinoma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:574-583. [PMID: 17182598 DOI: 10.4049/jimmunol.178.1.574] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tumor recurrence rates remain high after curative treatments for hepatocellular carcinoma (HCC). Immunomodulatory agents, including chemokines, are believed to enhance the antitumor effects of tumor cell apoptosis induced by suicide gene therapy. We therefore evaluated the immunomodulatory effects of a bicistronic recombinant adenovirus vector (rAd) expressing both HSV thymidine kinase and MCP-1 on HCC cells. Using an athymic nude mouse model (BALB/c-nu/nu), primary s.c. tumors (HuH7; human HCC cells) were completely eradicated by rAd followed by treatment with ganciclovir. The same animals were subsequently rechallenged with HCC cells, tumor development was monitored, and the recruitment or activation of NK cells was analyzed immunohistochemically or by measuring IFN-gamma mRNA expression. Tumor growth was markedly suppressed as compared with that in mice treated with a rAd expressing the HSV thymidine kinase gene alone (p < 0.001). Suppression of tumor growth was associated with the elevation of serum IL-12 and IL-18. During suppression, NK cells were recruited exclusively, and Th1 cytokine gene expression was enhanced in tumor tissues. The antitumor activity, however, was abolished either when the NK cells were inactivated with anti-asialo GM1 Ab or when anti-IL-12 and anti-IL-18 Abs were administered. These results indicate that suicide gene therapy, together with delivery of MCP-1, eradicates HCC cells and exerts prolonged NK cell-mediated antitumor effects in a model of HCC, suggesting a plausible strategy to prevent tumor recurrence.
Collapse
Affiliation(s)
- Tomoya Tsuchiyama
- Department of Gastroenterology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | |
Collapse
|
18
|
Russell PJ, Khatri A. Novel gene-directed enzyme prodrug therapies against prostate cancer. Expert Opin Investig Drugs 2006; 15:947-61. [PMID: 16859396 DOI: 10.1517/13543784.15.8.947] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
There is no effective cure for late-stage hormone (androgen) refractory prostate cancer. Although chemotherapy offers palliation to these late-stage patients, it also leads to systemic toxicities leading to poor quality of life. Clearly, the focus is on the development and evaluation of novel biologically relevant alternatives such as cytoreductive gene-directed enzyme prodrug therapy (GDEPT). With the current limitations of effective gene delivery in vivo, the in situ amplification of cytotoxicity due to bystander effects of GDEPT has special attraction for patients with prostate cancer, the prostate being dispensable. This review focuses on the development, application and potential of various GDEPTs for treating prostate cancer. The current status of research related to the issues of enhancement of in situ GDEPT delivery and prostate cancer-specific targeting of vectors (especially viral vectors) is assessed. Finally, the scope and progress of synergies between GDEPT and other treatment modalities, both traditional and alternate, are discussed.
Collapse
Affiliation(s)
- Pamela J Russell
- Oncology Research Centre, Prince of Wales Hospital Sydney, Level 2, Clinical Sciences Building, Barker Street, Randwick, NSW 2031, Australia.
| | | |
Collapse
|
19
|
Yan Y, Rubinchik S, Wood AL, Gillanders WE, Dong JY, Watson DK, Cole DJ. Bystander effect contributes to the antitumor efficacy of CaSm antisense gene therapy in a preclinical model of advanced pancreatic cancer. Mol Ther 2005; 13:357-65. [PMID: 16226492 DOI: 10.1016/j.ymthe.2005.06.485] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Revised: 06/21/2005] [Accepted: 06/21/2005] [Indexed: 01/29/2023] Open
Abstract
Pancreatic adenocarcinoma (PC) is an aggressive malignancy resistant to standard treatment modalities. Previously, we have reported that cancer-associated Sm-like protein (CaSm) contributes to the neoplastic transformation of PC. In this study, we utilized a recently established preclinical model of PC to determine if molecular targeting of CaSm can serve as the basis for a novel PC therapy. In a subcutaneous tumor model, intratumoral administration of an adenoviral vector encoding CaSm antisense RNA (Ad-alphaCaSm) significantly inhibited Panc02 tumor growth. Furthermore, in a metastatic tumor model, systemic administration of Ad-alphaCaSm resulted in a significant decrease in the number of hepatic metastases and increased survival time. We assessed the efficiency of in vivo delivery and observed significant levels of vector transduction in tissues containing PC, as well as a bystander effect that was amplifying the efficacy of CaSm gene therapy. This bystander effect was also active in vitro and was shown to be at least partially independent of host-related mechanisms. We conclude that CaSm antisense gene therapy is an effective novel therapy for PC and that the antitumor efficacy is dependent on both direct and bystander mechanisms.
Collapse
Affiliation(s)
- Yan Yan
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, 29425, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Nikitina EY, Desai SA, Zhao X, Song W, Luo AZ, Gangula RD, Slawin KM, Spencer DM. Versatile Prostate Cancer Treatment with Inducible Caspase and Interleukin-12. Cancer Res 2005; 65:4309-19. [PMID: 15899823 DOI: 10.1158/0008-5472.can-04-3119] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To establish optimized conditions for immunity against prostate cancer, we compared the efficacy of multiple approaches in autochthonous and s.c. transgenic adenocarcinoma of the mouse prostate (TRAMP)-based models. Mice immunized with interleukin (IL)-12-containing apoptotic, but not necrotic TRAMP-C2 cell-based, vaccines were resistant to TRAMP-C2 tumor challenge and re-challenge, independently of the route of vaccination (s.c. or i.p.). Administration of gamma-irradiated TRAMP-C2 cells preinfected with adenovirus containing both B7-1 and IL-12 genes, unlike adenovirus containing B7-1 alone, considerably protected C57BL/6 mice from TRAMP-C2 tumor growth and extended the life span of TRAMP mice. Vaccines that included dendritic cells, instead of IL-12, were equally efficient. Whereas injections of ligand-inducible caspase-1- and IL-12-containing adenoviruses cured small s.c. TRAMP-C2 tumors, nanopump-regulated delivery of viruses led to elimination of much larger tumors. The antitumor immune responses involved CD4+-, CD8+-, and natural killer cells and were strengthened by increasing the number of vaccinations. Intraprostatic administration of inducible caspase-1- and IL-12-containing adenoviruses resulted in local cell death and improved survival of adenocarcinoma-bearing TRAMP mice. Thus, tumor cell apoptosis induced by caspase in situ and accompanied by IL-12 is efficient against prostate cancer in a preclinical model.
Collapse
Affiliation(s)
- Ekaterina Yu Nikitina
- Department of Immunology and Scott Department of Urology, Baylor College of Medicine, Texas Medical Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Sakakima Y, Hayashi S, Yagi Y, Hayakawa A, Tachibana K, Nakao A. Gene therapy for hepatocellular carcinoma using sonoporation enhanced by contrast agents. Cancer Gene Ther 2005; 12:884-9. [PMID: 15891773 DOI: 10.1038/sj.cgt.7700850] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We examined whether sonoporation enhanced by a contrast agent (BR14) was effective in gene therapy for hepatocelluar carcinoma (HCC). Human hepatic cancer cells (SK-Hep1) and plasmid cDNAs expressing green fluorescent protein (GFP), interferonbeta (IFNbeta), and LacZ were used. In vitro, SK-Hep1 cell suspensions with DNA and BR14 were sonoporated. Expressions of every plasmid cDNA and the antitumor effect of IFNbeta were analyzed. In vivo, GFP and IFNbeta genes with BR14 were directly injected into subcutaneous tumors using SK-Hep1 in nude mice, and transcutaneous sonoporation of the tumors was performed. GFP gene transfections and tumor diameters after IFNbeta gene transfection were examined. In vitro, no SK-Hep1 cells were transfected without sonication, whereas transfections were successful after sonication with BR14. Antitumor effect of IFNbeta gene transfection by ultrasound (US) and with BR14 was revealed. In vivo, the SK-Hep1 cells expressed GFP, and the IFNbeta gene transfection by US with BR14 reduced tumor size significantly. In conclusion, gene therapy with sonoporation enhanced by a contrast agent may become a new treatment option for HCC.
Collapse
Affiliation(s)
- Yoshikazu Sakakima
- Department of Surgery II, Nagoya University School of Medicine, Nagoya 466-8550, Japan.
| | | | | | | | | | | |
Collapse
|
22
|
Goto T, Nishi T, Kobayashi O, Tamura T, Dev SB, Takeshima H, Kochi M, Kuratsu JI, Sakata T, Ushio Y. Combination electro-gene therapy using herpes virus thymidine kinase and interleukin-12 expression plasmids is highly efficient against murine carcinomas in vivo. Mol Ther 2004; 10:929-37. [PMID: 15509510 DOI: 10.1016/j.ymthe.2004.07.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2004] [Accepted: 07/27/2004] [Indexed: 10/26/2022] Open
Abstract
We report the use of plasmid DNA-mediated combination gene therapy for tumor-bearing mice using in vivo electroporation, also called electro-gene therapy (EGT), that resulted in uncomplicated and complete cures in more than 90% of the mice. Subcutaneously inoculated CT26 tumors in syngeneic BALB/c mice were subjected to repeated EGT treatments consisting of intratumoral co-injection of naked plasmids encoding the cytokine interleukin-12 (IL-12) (p35 and p40 subunits) and the suicide gene herpes simplex virus thymidine kinase (HSV-tk), followed by in vivo electroporation. The early anti-tumor effect was always stronger, and the rate of cure, as seen in the long-term follow-up, was always greater in the groups treated with combination EGT than in those treated with IL-12 or HSV-tk EGT alone. Systemic levels of IL-12 and IFN-gamma increased in both combination and IL-12-alone EGT-treated groups. Moreover, combination EGT for established subcutaneous tumors strongly reduced hematogenous lung metastases and increased survival time when live CT26 tumor cells were injected through the tail vein. Limited experiments on C57/B16 mice with murine melanoma also showed very similar trends. These results suggest that this simple and safe method of plasmid-mediated combination EGT may provide a potentially effective gene therapy for cancer.
Collapse
Affiliation(s)
- Tomoaki Goto
- Department of Neurosurgery, Saiseikai Kumamoto Hospital, 5-3-1 Chikami, Kumamoto 861-4193, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ziller C, Lincet H, Muller CD, Staedel C, Behr JP, Poulain L. The cyclin-dependent kinase inhibitor p21cip1/waf1 enhances the cytotoxicity of ganciclovir in HSV-tk transfected ovarian carcinoma cells. Cancer Lett 2004; 212:43-52. [PMID: 15246560 DOI: 10.1016/j.canlet.2004.03.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2004] [Accepted: 03/24/2004] [Indexed: 11/22/2022]
Abstract
Suicide gene therapy could be an attractive addition to the treatment of ovarian carcinomas, for which acquired chemoresistance frequently results in treatment failure. Here we show that transfection of the HSV-tk gene, followed by incubation with up to 1 mM ganciclovir fails to induce cell death in SKOV3 chemoresistant human ovarian carcinoma cells. However, co-transfection of HSV-tk with Cip1/Waf1 encoding the p21(cip1/waf1) inhibitor of cdks, allows 100 microM ganciclovir to eradicate the population of tumor cells. Potentiation of a drug by co-transfer of HSV-tk with Cip1/Waf1could thus represent another therapeutic approach for tumours that are resistant to conventional therapy.
Collapse
Affiliation(s)
- Christelle Ziller
- Laboratoire de Chimie Génétique, CNRS UMR 7514, Université Louis Pasteur Strasbourg I, Faculté de Pharmacie, 74 route du Rhin, BP 24, 67401 Illkirch, France
| | | | | | | | | | | |
Collapse
|
24
|
Zhang JH, Wan MX, Yuan JY, Pan BR. Construction and identification of recombinant vectors carrying herpes simplex virus thymidine kinase and cytokine genes expressed in gastric carcinoma cell line SGC7901. World J Gastroenterol 2004; 10:26-30. [PMID: 14695763 PMCID: PMC4717072 DOI: 10.3748/wjg.v10.i1.26] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To construct and identify the recombinant vectors carrying herpes simplex virus thymidine kinase (HSV-TK) and tumor necrosis factor alpha (TNF-α) or interleukin-2 (IL-2) genes expressed in gastric carcinoma cell line SGC7901.
METHODS: The fragments of HSV-TK, internal ribosome entry sites (IRES) and TNF-α or IL-2 genes were inserted in a TK-IRES-TNF-α or TK-IRES-IL-2 order into pEGFP-N3 and pLXSN to generate the therapeutic vectors pEGFP-TT, pEGFP-TI, pL(TT)SN and pL(TI)SN respectively, which were structurally confirmed by the digestion analysis of restriction endonuclease. The former two plasmids were used for the transient expression of recombinant proteins in the target cells while pL(TT)SN and pL(TI)SN were transfected into SGC7901 cells by lipofectamine for the stable expression of objective genes through G418 selection. The protein products expressed transiently and stably in SGC7901 cells by the constructed vectors were confirmed by fluorescent microscopy and Western blot respectively.
RESULTS: The inserted fragments in all constructed plasmids were structurally confirmed to be consistent with that of the published data. In the transient expression, both pEGFP-TT and pEGFP-TI were shown expressed in nearly 50% of the transfected SGC7901 cells. Similarly, the G418 selected vectors PL(TT)SN and PL(TI)SN were confirmed to be successful in the stable expression of the objective proteins in the target cells.
CONCLUSION: The constructed recombinant vectors in the present study that can express the suicide gene TK in combination with cytokines genes may serve as the potential tools to perform more effective investigations in future for the gene therapy of gastric carcinoma.
Collapse
Affiliation(s)
- Jian-Hua Zhang
- Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, Shaanxi Province, China
| | | | | | | |
Collapse
|
25
|
Zhang JH, Wan MX, Yuan JY, Pan BR. Do there exist synergistic antitumor effects by coexpression of herpes simplex virus thymidine kinase with cytokine genes on human gastric cancer cell line SGC7901? World J Gastroenterol 2004; 10:147-51. [PMID: 14695787 PMCID: PMC4717068 DOI: 10.3748/wjg.v10.i1.147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To evaluate the synergistic antitumor effects of herpes simplex virus thymidine kinase (HSV-TK) together with tumor necrosis factor alpha (TNF-α) or interleukin-2 (IL-2) gene expression on gastric cancer cell line SGC7901.
METHODS: Recombinant vectors pL(TT)SN and pL(TI)SN, which express TK-IRES-TNF-α and TK-IRES-IL-2 genes separately, as well as the control plasmids pL(TK)SN and pLXSN were employed to transfect PA317 cells respectively to generate the viruses that can stably express the objective genes through G418 selection. The gastric cancer cells were then transfected by the retroviral serum from the package cells and maintained in culture to determine the cell growth and apoptosis. The cytotoxic effects of HSV-TK together with TNF-α or IL-2 gene expression on the transfected cancer cells were evaluated by the cell viability and bystander effects in the presence of GCV supplemented in the cultural medium.
RESULTS: Expression of recombinant proteins including TNF-α and IL-2 by stable transfectants was confirmed by Western blotting. The percentage of cell apoptosis in the SGC/0, SGC/TK-TNF-α, SGC/TK-IL-2 and SGC/TK clone was 2.3%, 12.3%, 11.1% and 10.9% respectively at 24 h post-transfection. Cell growth status among all the experimental groups as judged by cell absorbance (A) at 570nm did not exhibit any significant difference (P > 0.05); although it was noted to be slightly lower in the SGC/TT group. Cell survival rate in SGC/TI, SGC/TT and SGC/TK group was significantly decreased in a dose-dependent manner of GCV compared with that of the SGC/0 group (P < 0.05-0.01). Among all studied cells, the SGC/TT was shown most sensitive to GCV with a half lethal dose of 0.5 mg·L-1. In contrast, the survival rate of SGC/0 cells was not affected by the presence of GCV with the doses less than 10 mg·L-1. The half lethal dose of GCV for SGC/0 cells was more than 100 mg·L-1. Marked bystander effect induced by SGC/TI, SGC/TT and SGC/TK cells was confirmed by the fact that 20% of these stable transfectants could kill 50% of the co-cultured cells, in which the most prominent bystander effect was found in the circumstance of SGC/TT presence. However, no significant difference of these variables was found among SGC/TI, SGC/TT and SGC/TK cells (P > 0.05).
CONCLUSION: The synergistic antitumor effects produced by the co-expression of HSV-TK with TNF-α or IL-2 genes were not present in the transfected SGC7901 cells. The mechanism underlying these phenomena was not known.
Collapse
Affiliation(s)
- Jian-Hua Zhang
- Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, Shaanxi Province, China.
| | | | | | | |
Collapse
|
26
|
Yamazaki M, Straus FH, Messina M, Robinson BG, Takeda T, Hashizume K, DeGroot LJ. Adenovirus-mediated tumor-specific combined gene therapy using Herpes simplex virus thymidine/ganciclovir system and murine interleukin-12 induces effective antitumor activity against medullary thyroid carcinoma. Cancer Gene Ther 2003; 11:8-15. [PMID: 14681722 DOI: 10.1038/sj.cgt.7700636] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The present treatment of advanced and metastatic medullary thyroid carcinoma (MTC) is unsatisfactory. Tissue-specific cancer gene therapy is a novel alternative approach. We developed a recombinant adenovirus expressing Herpes simplex type 1 thymidine kinase (HSVtk) driven by a modified CALC-I promoter TCP (AdTCPtk). Infection with this virus showed efficient cytotoxic effect on MTC cell lines (rMTC and TT cells) after treatment with ganciclovir (GCV) in vitro. In a syngenic WAG/Rij rat model, the combination of AdTCPtk/GCV treatment with administration of murine interleukin-12 (mIL-12) expressing adenovirus under control of TCP (AdTCPmIL-12) resulted in effective growth suppression of tumor at the treated site and also at a distant untreated site, in comparison to treatment with AdTCPtk/GCV or AdTCPmIL-12 alone. Moreover, intravenous injection of AdTCPtk, or AdTCPtk+AdTCPmIL-12, followed by administration of GCV, did not cause evident toxicity after administration of GCV. These results indicate that this combined system can provide an effective therapy for metastatic MTC with minimal toxicity.
Collapse
Affiliation(s)
- Masanori Yamazaki
- Thyroid Study Unit, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
OBJECTIVES To give oncology nurses an overview on the vectors and selected approaches used in the current clinical trials involving gene transfer to cancer patients. DATA SOURCES Peer-reviewed scientific papers, review articles, and book chapters. CONCLUSION Significant progress has been made in the field of cancer gene therapy. Different phases of clinical protocols derived from new generations of vectors and novel approaches are being tested for use in the treatment of patients with cancer. IMPLICATIONS FOR NURSING PRACTICE Oncology nurses need to be familiar with current advances in the field of cancer gene therapy to expand their role as health care professional, patient educator, and advocate for the treatment of patients with cancer.
Collapse
Affiliation(s)
- Ke Liu
- Surgery Branch, Center for Cancer Research, National Institutes of Health, 9000 Rockville Pike, Bldg 10, Room 2B08, Bethesda, MD 20892-1502, USA
| |
Collapse
|
28
|
Abstract
Tumor necrosis factor (TNF)-related cytokines are critical effector molecules in the immune response to viral pathogens. Engagement of the TNF receptors by their cognate ligands activates apoptotic and non-apoptotic signaling pathways, both of which can mediate antiviral activity. In response, viruses have evolved mechanisms to inhibit signaling by some cytokines of the TNF superfamily. These strategies are largely unique to each class of virus, but are similar in that they all target key regulatory checkpoints of the TNF pathway. In recent years, studies directed towards dissecting the mechanisms of TNF signaling and the viral retort have led to several significant discoveries, and form the basis for this review.
Collapse
Affiliation(s)
- Chris A Benedict
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, 10355 Science Center Drive, San Diego, CA 92121, USA.
| |
Collapse
|
29
|
Hyer ML, Sudarshan S, Schwartz DA, Hannun Y, Dong JY, Norris JS. Quantification and characterization of the bystander effect in prostate cancer cells following adenovirus-mediated FasL expression. Cancer Gene Ther 2003; 10:330-9. [PMID: 12679806 DOI: 10.1038/sj.cgt.7700576] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Inducing Fas-mediated apoptosis in prostate cancer (PCa) is a promising new therapeutic approach with the potential to overcome delivery issues currently problematic in cancer gene therapy. We have previously demonstrated that a Fas Ligand (FasL) expressing adenovirus (AdGFPFasL(TET)) was able to induce Fas-mediated apoptosis in a panel of PCa cell lines regardless of their Fas-sensitivity as determined by the agonistic Fas antibody CH-11. We now report that AdGFPFasL(TET)-infected cells produce apoptotic bodies and cellular debris that continues to elicit FasL-mediated bystander killing in uninfected neighboring cells. Using light microscopy, we demonstrate that AdGFPFasL(TET)-infected cells release apoptotic bodies and cellular debris into the local environment and that this material will induce bystander killing in Jurkat, PPC-1, and PC-3 target cells, but not in DU145 and K-562 cells. The bystander killing mechanism is mediated through Fas/FasL interaction because it is significantly inhibited if target cells are pretreated with the pan spectrum caspase inhibitor Z-VAD-FMK or the Fas neutralizing antibody ZB-4. Coincubation of PPC-1 target cells with apoptotic bodies and cellular debris (effector material) induce nearly complete target cell killing at a ratio of 1:1 target to effector. Collectively, these data indicate that AdGFPFasL(TET)-infected PCa cells release apoptotic and cellular debris capable of inducing bystander killing in PCa and supports the development of FasL as a gene therapy agent.
Collapse
Affiliation(s)
- Marc L Hyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | |
Collapse
|
30
|
Benedict CA, Banks TA, Ware CF. Death and survival: viral regulation of TNF signaling pathways. Curr Opin Immunol 2003; 15:59-65. [PMID: 12495734 DOI: 10.1016/s0952-7915(02)00018-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Chris A Benedict
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, 10355 Science Center Drive, San Diego, CA 92121, USA
| | | | | |
Collapse
|
31
|
Abstract
Despite advances in surgery, radiotherapy, and chemotherapy, the survival of patients with oral squamous cell carcinoma has not significantly improved over the past several decades. Treatment options for recurrent or refractory oral cancers are limited. Gene therapy for oral cancer is currently under investigation in clinical trials. The goal of cancer gene therapy is to introduce new genetic material into target cells without toxicity to non-target tissues. This review discusses the techniques used in cancer gene therapy for oral squamous cell carcinoma and summarizes the ongoing strategies that are being evaluated in clinical trials.
Collapse
Affiliation(s)
- S Xi
- Department of Otolaryngology, University of Pittsburgh School of Medicine, PA, USA
| | | |
Collapse
|
32
|
Tarner IH, Fathman CG. The potential for gene therapy in the treatment of autoimmune disease. Clin Immunol 2002; 104:204-16. [PMID: 12217329 DOI: 10.1006/clim.2002.5235] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Ingo H Tarner
- Department of Rheumatology, Stanford University School of Medicine, California 94305-5166, USA
| | | |
Collapse
|