1
|
Nikrad JA, Galvin RT, Sheehy MM, Novacek EL, Jacobsen KL, Corbière SMAS, Beckmann PJ, Jubenville TA, Yamamoto M, Largaespada DA. Conditionally replicative adenovirus as a therapy for malignant peripheral nerve sheath tumors. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200783. [PMID: 38595983 PMCID: PMC10959710 DOI: 10.1016/j.omton.2024.200783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024]
Abstract
Oncolytic adenoviruses (Ads) stand out as a promising strategy for the targeted infection and lysis of tumor cells, with well-established clinical utility across various malignancies. This study delves into the therapeutic potential of oncolytic Ads in the context of neurofibromatosis type 1 (NF1)-associated malignant peripheral nerve sheath tumors (MPNSTs). Specifically, we evaluate conditionally replicative adenoviruses (CRAds) driven by the cyclooxygenase 2 (COX2) promoter, as selective agents against MPNSTs, demonstrating their preferential targeting of MPNST cells compared with non-malignant Schwann cell control. COX2-driven CRAds, particularly those with modified fiber-knobs exhibit superior binding affinity toward MPNST cells and demonstrate efficient and preferential replication and lysis of MPNST cells, with minimal impact on non-malignant control cells. In vivo experiments involving intratumoral CRAd injections in immunocompromised mice with human MPNST xenografts significantly extend survival and reduce tumor growth rate compared with controls. Moreover, in immunocompetent mouse models with MPNST-like allografts, CRAd injections induce a robust infiltration of CD8+ T cells into the tumor microenvironment (TME), indicating the potential to promote a pro-inflammatory response. These findings underscore oncolytic Ads as promising, selective, and minimally toxic agents for MPNST therapy, warranting further exploration.
Collapse
Affiliation(s)
- Julia A Nikrad
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Robert T Galvin
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Mackenzie M Sheehy
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Ethan L Novacek
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Kari L Jacobsen
- Department of Surgery, University of Minnesota, 516 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Stanislas M A S Corbière
- Institute for Research in Immunology and Cancer, Université de Montréal, 2950 Chemin de Polytechnique Marcelle-Coutu Pavilion, Montréal, QC H3T1J4, Canada
| | - Pauline J Beckmann
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Tyler A Jubenville
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, 516 Delaware Street SE, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - David A Largaespada
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
2
|
Zhang H, Wang H, An Y, Chen Z. Construction and application of adenoviral vectors. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102027. [PMID: 37808925 PMCID: PMC10556817 DOI: 10.1016/j.omtn.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Adenoviral vectors have been widely used as vaccine candidates or potential vaccine candidates against infectious diseases due to the convenience of genome manipulation, their ability to accommodate large exogenous gene fragments, easy access of obtaining high-titer of virus, and high efficiency of transduction. At the same time, adenoviral vectors have also been used extensively in clinical research for cancer gene therapy and treatment of diseases caused by a single gene defect. However, application of adenovirus also faces a series of challenges such as poor targeting, strong immune response against the vector itself, and they cannot be used repeatedly. It is believed that these problems will be solved gradually with further research and technological development in related fields. Here, we review the construction methods of adenoviral vectors, including "gutless" adenovirus and discuss application of adenoviral vectors as prophylactic vaccines for infectious pathogens and their application prospects as therapeutic vaccines for cancer and other kinds of chronic infectious disease such as human papillomavirus, hepatitis B virus, and hepatitis C virus.
Collapse
Affiliation(s)
- Hongbo Zhang
- Department of Basic Research, Ab&B Bio-Tech CO., LTD. JS, Taizhou, Jiangsu, China
| | - Hongdan Wang
- Department of Basic Research, Ab&B Bio-Tech CO., LTD. JS, Taizhou, Jiangsu, China
| | - Youcai An
- Department of Basic Research, Ab&B Bio-Tech CO., LTD. JS, Taizhou, Jiangsu, China
| | - Ze Chen
- Department of Basic Research, Ab&B Bio-Tech CO., LTD. JS, Taizhou, Jiangsu, China
| |
Collapse
|
3
|
Oncolytic viruses: A novel treatment strategy for breast cancer. Genes Dis 2021; 10:430-446. [DOI: 10.1016/j.gendis.2021.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/27/2021] [Accepted: 11/19/2021] [Indexed: 11/21/2022] Open
|
4
|
Kimura J, Ono HA, Kosaka T, Nagashima Y, Hirai S, Ohno S, Aoki K, Julia D, Yamamoto M, Kunisaki C, Endo I. Conditionally replicative adenoviral vectors for imaging the effect of chemotherapy on pancreatic cancer cells. Cancer Sci 2013; 104:1083-90. [PMID: 23679574 DOI: 10.1111/cas.12196] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 05/01/2013] [Accepted: 05/03/2013] [Indexed: 12/26/2022] Open
Abstract
Pancreatic cancer has a poor prognosis after complete macroscopic resection combined with chemotherapy. Even after neoadjuvant chemotherapy, R0 resection is often not possible. Moreover, current imaging techniques cannot reliably distinguish viable cancer cells from scar tissue at the resectional margin. We investigated the use of a conditionally replicative adenovirus (CRAd), Ad5/3Cox2CRAd-ΔE3ADP-Luc, for imaging the effects of chemotherapy. The CRAd infectivity of pancreatic cancer cells was enhanced by a chimeric Ad5/3 fiber, E1A expression was under the control of the Cox2 promoter, and the luciferase gene was inserted adjacent to the adenovirus death protein (ADP) gene. Subcutaneous xenografts of the pancreatic cancer cell line MiaPaCa-2 were established in 24 BALB/c nu/nu mice. When xenografts reached a diameter of 4-6 mm (day 1), the mice were injected i.p. with either PBS (group A; n = 12) or 1000 mg/kg gemcitabine (group B; n = 12), weekly. On days 19, 26, 33, and 40, CRAd were injected intratumorally into three mice in groups A and B. Bioluminescence was imaged 72 h after CRAd injection, and gross tumor volumes were measured then tumors were removed for ex vivo histopathology using H&E and Ki-67 staining. Correlations between gross tumor volume, pathological evaluation of the percentage of viable tumor area, and CRAd bioluminescence were analyzed. Bioluminescence correlated closely with the percentage of viable tumor area (R = 0.96), but not with gross tumor volume (R = 0.31). Therefore, CRAds might be reliable imaging tools for monitoring chemotherapy in pancreatic cancer, and could improve our ability to distinguish viable tumor cells from scar tissue.
Collapse
Affiliation(s)
- Jun Kimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama-city University, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Wu JH, Wang HF, Wang ZC, Xu K, Qi YL, Li JH, Gong SL, Liu Y, Liu Y. Conditionally replicating adenovirus combined with gene-targeted radiotherapy induces apoptosis via TRAIL death receptors in MDA-MB-231 cells. Mol Med Rep 2013; 8:299-305. [PMID: 23708314 DOI: 10.3892/mmr.2013.1488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 05/20/2013] [Indexed: 11/06/2022] Open
Abstract
Malignant tumors are usually treated using monotherapies, which are not always effective. Therefore, combination therapies have gained increasing attention. The aims of this study were to investigate the effects of conditionally replicating adenovirus (CRAd) in combination with X-ray irradiation on the proliferation and apoptosis of MDA-MB-231 cells, as well as to determine the molecular mechanisms involved. MDA-MB-231 cells were treated simultaneously with CRAd and X-ray irradiation. Then, cell viability was measured using Cell Counting Kit-8. Cell apoptosis was assessed by flow cytometry with Annexin V and propidium iodide (PI) double-staining. The expression of tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL), death receptor 5 (DR5), caspase-3 and caspase-8 mRNA was detected by quantitative polymerase chain reaction. The expression of TRAIL, DR5, caspase-3 and caspase-8 protein was measured by enzyme-linked immunosorbent assay (ELISA) and western blotting, respectively. The results showed that CRAd, in combination with irradiation, inhibited cell proliferation, promoted cell apoptosis and significantly increased the expression of TRAIL, DR5, caspase-3 and caspase-8 mRNA and proteins in MDA-MB-231 cells. Therefore, three aspects, including the targeted killing effect of CRAd, the apoptosis-promoting role of TRAIL and the direct killing effect of ionizing radiation to MDA-MB-231 cells, contribute to the mechanisms of CRAd in combination with irradiation to inhibit the proliferation of MDA-MB-231 cells. The pro-apoptotic effect may involve the interaction between TRAIL, DR5, caspase-3 and caspase-8.
Collapse
Affiliation(s)
- Jia Hui Wu
- Key Laboratory of Radiobiology, Ministry of Health, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Armstrong L, Arrington A, Han J, Gavrikova T, Brown E, Yamamoto M, Vickers SM, Davydova J. Generation of a novel, cyclooxygenase-2-targeted, interferon-expressing, conditionally replicative adenovirus for pancreatic cancer therapy. Am J Surg 2012; 204:741-50. [PMID: 22748294 DOI: 10.1016/j.amjsurg.2012.02.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 02/16/2012] [Accepted: 02/16/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND Oncolytic adenoviruses provide a promising alternative for cancer treatment. Recently, adjuvant interferon (IFN)-alfa has shown significant survival benefits for pancreatic cancer, yet was impeded by systemic toxicity. To circumvent these problems adenovirus with high-level targeted IFN-alfa expression can be generated. METHODS Conditionally replicative adenoviruses (CRAds) with improved virulence and selectivity for pancreatic cancer were generated. The vectors were tested in vitro, in vivo, and in human pancreatic cancer and normal tissue specimens. RESULTS Adenoviral death protein and fiber modifications significantly improved oncolysis. CRAds selectively replicated in vitro, in vivo and showed persistent spread in cancer xenografts. They showed high-level replication in human pancreatic cancer specimens, but not in normal tissues. Improved IFN-CRAd oncolytic efficiency was shown. CONCLUSIONS Optimized cyclooxygenase-2 CRAds show highly favorable effects in vitro and in vivo. We report a pancreatic cancer-specific, highly virulent, IFN-expressing CRAd, and we believe that adenovirus-based IFN therapy offers a new treatment opportunity for pancreatic cancer patients.
Collapse
Affiliation(s)
- Leonard Armstrong
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Paul A, Binsalamah ZM, Khan AA, Abbasia S, Elias CB, Shum-Tim D, Prakash S. A nanobiohybrid complex of recombinant baculovirus and Tat/DNA nanoparticles for delivery of Ang-1 transgene in myocardial infarction therapy. Biomaterials 2011; 32:8304-18. [DOI: 10.1016/j.biomaterials.2011.07.042] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 07/13/2011] [Indexed: 01/03/2023]
|
8
|
In Vivo Anti-Tumor Effect of Expressing p14ARF-TAT Using a FGF2-Targeted Cationic Lipid Vector. Pharm Res 2011; 28:720-30. [DOI: 10.1007/s11095-010-0353-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 12/14/2010] [Indexed: 11/29/2022]
|
9
|
Bourke MG, Salwa S, Harrington KJ, Kucharczyk MJ, Forde PF, de Kruijf M, Soden D, Tangney M, Collins JK, O'Sullivan GC. The emerging role of viruses in the treatment of solid tumours. Cancer Treat Rev 2011; 37:618-32. [PMID: 21232872 DOI: 10.1016/j.ctrv.2010.12.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 12/04/2010] [Accepted: 12/07/2010] [Indexed: 12/13/2022]
Abstract
There is increasing optimism for the use of non-pathogenic viruses in the treatment of many cancers. Initial interest in oncolytic virotherapy was based on the observation of an occasional clinical resolution of a lymphoma after a systemic viral infection. In many cancers, by comparison with normal tissues, the competency of the cellular anti-viral mechanism is impaired, thus creating an exploitable difference between the tumour and normal cells, as an unimpeded viral proliferation in cancer cells is eventually cytocidal. In addition to their oncolytic capability, these particular viruses may be engineered to facilitate gene delivery to tumour cells to produce therapeutic effects such as cytokine secretion and anti -tumour immune responses prior to the eventual cytolysis. There is now promising clinical experience with these viral strategies, particularly as part of multimodal studies, and already several clinical trials are in progress. The limitations of standard cancer chemotherapies, including their lack of specificity with consequent collateral toxicity and the development of cross-resistance, do not appear to apply to viral-based therapies. Furthermore, virotherapy frequently restores chemoradiosensitivity to resistant tumours and has also demonstrated efficacy against cancers that historically have a dismal prognosis. While there is cause for optimism, through continued improvements in the efficiency and safety of systemic delivery, through the emergence of alternative viral agents and through favourable clinical experiences, clinical trials as part of multimodal protocols will be necessary to define clinical utility. Significant progress has been made and this is now a major research area with an increasing annual bibliography.
Collapse
Affiliation(s)
- M G Bourke
- Cork Cancer Research Centre, Leslie C. Quick Jnr. Laboratory, Biosciences Institute, University College Cork, Ireland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Sagawa T, Yamada Y, Takahashi M, Sato Y, Kobune M, Takimoto R, Fukaura J, Iyama S, Sato T, Miyanishi K, Matsunaga T, Takayama T, Kato J, Sasaki K, Hamada H, Niitsu Y. Treatment of hepatocellular carcinoma by AdAFPep/rep, AdAFPep/p53, and 5-fluorouracil in mice. Hepatology 2008; 48:828-40. [PMID: 18756484 DOI: 10.1002/hep.22420] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
UNLABELLED Although conditionally replicable adenovirus (CRAd) has been used in the clinical treatment of hepatocellular carcinoma (HCC), it suffers from the inherent drawback of having relatively low antitumor activity. Here, we have sought to overcome this drawback. First, we combined CRAd (AdAFPep/Rep) driven by alpha-fetoprotein enhancer/promoter (AFPep) with a replication-incompetent adenovirus carrying a p53 transgene that is also driven by AFPep. The synergism of this combination produced a significantly improved tumoricidal effect on the human HCC cell line Hep3B, which has a relatively short doubling time in comparison with other human HCC cell lines, through the transactivation of p53 by early region 1A transcribed by AdAFPep/Rep. This synergistic interaction was augmented by the addition of a subtumoricidal dose (0.5 microg/mL) of 5-fluorouracil (5-FU), which enhanced p53 expression and facilitated the release of virions from tumor cells. When relatively large (10-mm-diameter) Hep3B tumors grown in nude mice were injected with the two viruses in combination, they showed significantly impaired growth in comparison with those treated with each virus separately. The growth suppression effect of the virus combination was enhanced by a low dose (600 microg) of 5-FU. Survival of the tumor-bearing mice treated with these three agents was significantly longer than that of control mice. Moreover, the tumor completely disappeared with the repeated injection of these agents. CONCLUSION This combination strategy holds promise for the treatment of relatively large and rapidly growing HCCs that may be encountered clinically.
Collapse
Affiliation(s)
- Tamotsu Sagawa
- Fourth Department of Internal Medicine, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Ko YT, Hartner WC, Kale A, Torchilin VP. Gene delivery into ischemic myocardium by double-targeted lipoplexes with anti-myosin antibody and TAT peptide. Gene Ther 2008; 16:52-9. [PMID: 18701915 DOI: 10.1038/gt.2008.135] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The treatment of myocardial ischemia using gene therapy is a rather novel but promising approach. Gene delivery to target cells may be enhanced by using double-targeted delivery systems simultaneously capable of extracellular accumulation and intracellular penetration. With this in mind, we have used low cationic liposomes-plasmid DNA complexes (lipoplexes) modified with cell-penetrating transactivating transcriptional activator (TAT) peptide (TATp) and/or with monoclonal anti-myosin monoclonal antibody 2G4 (mAb 2G4) specific toward cardiac myosin, for targeted gene delivery to ischemic myocardium. In vitro transfection of both normoxic and hypoxic cardiomyocytes was enhanced by the presence of TATp as determined by fluorescence microscopy and ELISA. The in vitro transfection was further enhanced by the additional modification with mAb 2G4 antibody in the case of hypoxic, but not normoxic cardiomyocytes. However, we did not observe a synergism between TATp and mAb 2G4 ligands under our experimental condition. In in vivo experiments, we have clearly demonstrated an increased accumulation of mAb 2G4-modified TATp lipoplexes in the ischemic rat myocardium and significantly enhanced transfection of cardiomyocytes in the ischemic zone. Thus, the genetic transformation of normoxic and hypoxic cardiomyocytes can be enhanced by using lipoplexes modified with TATp and/or mAb 2G4. Such complexes also demonstrate an increased accumulation in the ischemic myocardium and effective transfection of hypoxic cardiomyocytes in vivo.
Collapse
Affiliation(s)
- Y T Ko
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
12
|
Singh R, Tian B, Kostarelos K. Artificial envelopment of nonenveloped viruses: enhancing adenovirus tumor targeting
in vivo. FASEB J 2008; 22:3389-402. [DOI: 10.1096/fj.08-103275] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Ravi Singh
- Nanomedicine Laboratory, Centre for Drug Delivery ResearchThe School of PharmacyUniversity of London London UK
| | - Bowen Tian
- Nanomedicine Laboratory, Centre for Drug Delivery ResearchThe School of PharmacyUniversity of London London UK
| | - Kostas Kostarelos
- Nanomedicine Laboratory, Centre for Drug Delivery ResearchThe School of PharmacyUniversity of London London UK
| |
Collapse
|
13
|
Abstract
Leiomyomas (fibroids) are common estrogen-dependent uterine tumours that cause significant morbidity for women and a substantial economic impact on health delivery systems. Currently, there is no effective medical treatment option for this condition-hysterectomy is the mainstay of management. This is not an attractive choice for many women, especially patients desiring to preserve their fertility potential. Gene therapy is becoming a clinical reality, with more than 600 clinical trials worldwide. Researchers have recently attempted to develop a gene-therapy-based approach for the ablation of uterine fibroids. The localized nature of this condition and its accessibility using different imaging or endoscopic techniques make it an attractive target for direct delivery of gene-based vectors. Recent work from our laboratory suggests the potential use of a dominant-negative form of estrogen receptor (ER) to inactivate estrogen signalling in leiomyoma cells and induce apoptosis. Our in vivo data in a mouse model demonstrate the ability of an adenovirus-expressing dominant-negative ER to arrest leiomyoma growth. We and others also have described the utility of the herpes simplex virus-thymidine kinase (HSV-TK) plus ganciclovir (GCV) suicide gene-therapy system to effectively eradicate leiomyoma cells by utilizing the bystandard effect phenomena and the high expression of gap-junction protein in these tumours. Further work on rat models will pave the way for future leiomyoma gene-therapy clinical trials and allow the realization of gene therapy as a viable non-surgical option for this common problem in women's health.
Collapse
Affiliation(s)
- Ayman Al-Hendy
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.
| | | |
Collapse
|
14
|
Kirby TO, Rivera A, Rein D, Wang M, Ulasov I, Breidenbach M, Kataram M, Contreras JL, Krumdieck C, Yamamoto M, Rots MG, Haisma HJ, Alvarez RD, Mahasreshti PJ, Curiel DT. A novel ex vivo model system for evaluation of conditionally replicative adenoviruses therapeutic efficacy and toxicity. Clin Cancer Res 2005; 10:8697-703. [PMID: 15623655 DOI: 10.1158/1078-0432.ccr-04-1166] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Current animal tumor models are inadequate for the evaluation of toxicity and efficacy of conditionally replicative adenoviruses. A novel model system is needed that will provide insight into the anticipated therapeutic index of conditionally replicative adenoviruses preclinically. We endeavored to show a novel model system, which involves ex vivo evaluation of conditionally replicative adenovirus toxicity and therapeutic efficacy in thin, precision-cut slices of human primary tumor and liver. EXPERIMENTAL DESIGN The Krumdieck thin-slice tissue culture system was used to obtain and culture slices of tumor xenografts of ovarian cancer cell lines, human primary ovarian tumors, and human liver. We determined the viability of slices in culture over a period of 36 to 48 hours by ([3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxphenyl-2-(4-sulfophenyl)-2H-tetrazolium, inner salt)]) (MTS) assay. In vitro Hey cells, slices of Hey xenografts, and human ovarian tumor or human liver slices were infected with 500vp/cell of either replication competent wild-type adenovirus (Ad5/3wt), conditionally replicative adenovirus (Ad5/3cox-2), or the replication deficient adenovirus (Ad5/3luc1). At 12-, 24-, and 36-hour intervals, the replication of adenoviruses in these slices was determined by quantitative reverse transcription-PCR of adenoviral E4 copy number. RESULTS Primary tumor slices were able to maintain viability for up to 48 hours after infection with nonreplicative virus (Ad5luc1). Infection of Hey xenografts with Ad5/3cox-2 showed replication consistent with that seen in Hey cells infected in an in vitro setting. Primary tumor slices showed replication of both Ad5/3wt and Ad5/3cox over a 36-hour time period. Human liver slices showed replication of Ad5/3wt but a relative reduction in replication of Ad5/3cox-2 indicative of conditional replication "liver off" phenotype, thus predicting lower toxicity. CONCLUSIONS The thin-slice model system represents a stringent method of ex vivo evaluation of novel replicative adenoviral vectors and allows assessment of human liver replication relative to human tumor replication. This is the first study to incorporate this system for evaluation of therapeutic efficacy and replicative specificity of conditionally replicative adenoviruses. Also, the study is the first to provide a valid means for preclinical assay of potential conditionally replicative adenovirus-based hepatotoxicities, thus providing a powerful tool to determine therapeutic index for clinical translation of conditionally replicative adenoviruses.
Collapse
Affiliation(s)
- Tyler O Kirby
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-2172, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Treatment of cancer is limited by toxicity to normal tissue with standard approaches (chemotherapy, surgery and radiotherapy). The use of selective replicating viral vectors may enable the targeting of gene-modified viruses to malignant tissue without toxic effect. Studies of these vectors have demonstrated tumour-selective replication and minimal evidence of replication in normal tissue. The most advanced clinical results reported involve gene-modified adenoviral vectors. Several completed, histologically confirmed responses to local/regional injection have been induced, particularly in recurrent squamous cell carcinoma involving the head and neck region. Dose limiting toxicity above 10(13) viral particles per injection has been observed. Anti-tumour effect is demonstrable in animal models without evidence of significant toxicity when these vectors are used alone or in combination with chemotherapy, radiation therapy or as gene delivery vehicles. Preliminary clinical trials, particularly with E1B-deleted adenoviruses, report evidence of clinical activity in comparison with expected historical responses. Enhancement in replication selectivity to malignant tissue is also demonstrated preclinically and clinically with an E1B-deleted adenovirus utilising a prostate-specific antigen promoter. Other selective replicating viral vectors such as herpes simplex virus and vaccinia virus have also been explored clinically and suggest evidence of activity in patients with cancer. Modifications may one day enable more aggressive use of these new and exciting therapeutics as systemic gene delivery vehicles.
Collapse
|
16
|
Zhang ZL, Zou WG, Luo CX, Li BH, Wang JH, Sun LY, Qian QJ, Liu XY. An armed oncolytic adenovirus system, ZD55-gene, demonstrating potent antitumoral efficacy. Cell Res 2004; 13:481-9. [PMID: 14728805 DOI: 10.1038/sj.cr.7290191] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
ONYX-015 is an attractive therapeutic adenovirus for cancer because it can selectively replicate in tumor cells and kill them. To date, clinical trials of this adenovirus have demonstrated marked safety but not potent enough when it was used alone. In this paper, we put forward a novel concept of Gene-ViroTherapy strategy and in this way, we constructed an armed therapeutic oncolytic adenovirus system, ZD55-gene, which is not only deleted of E1B 55-kD gene similar to ONYX-015, but also armed with foreign antitumor gene. ZD55-gene exhibited similar cytopathic effects and replication kinetics to that of ONYX-015 in vitro. Importantly, the carried gene is expressed and the expression level can increase with the replication of virus. Consequently, a significant antitumoral efficacy was observed when ZD55-CD/5-FU was used as an example in nude mice with subcutaneous human SW620 colon cancer. Our data demonstrated that ZD55-gene, which utilizing the Gene-ViroTherapy strategy, is more efficacious than each individual component in vivo.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Antineoplastic Agents/therapeutic use
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Cell Line, Tumor
- Cells, Cultured
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Colorectal Neoplasms/therapy
- Cytopathogenic Effect, Viral/drug effects
- Female
- Fluorouracil/therapeutic use
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Genetic Therapy
- Genetic Vectors
- HeLa Cells
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Transplantation
- Oncogenes/drug effects
- Transplantation, Heterologous
- Virus Replication
Collapse
Affiliation(s)
- Zi Lai Zhang
- Laboratory of Biotechnology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The field of cancer gene therapy is in continuous expansion, and technology is quickly moving ahead as far as gene targeting and regulation of gene expression are concerned. This review focuses on the endocrine aspects of gene therapy, including the possibility to exploit hormone and hormone receptor functions for regulating therapeutic gene expression, the use of endocrine-specific genes as new therapeutic tools, the effects of viral vector delivery and transgene expression on the endocrine system, and the endocrine response to viral vector delivery. Present ethical concerns of gene therapy and the risk of germ cell transduction are also discussed, along with potential lines of innovation to improve cell and gene targeting.
Collapse
Affiliation(s)
- Luisa Barzon
- Department of Histology, Microbiology, and Medical Biotechnologies, University of Padova, I-35121 Padua, Italy
| | | | | |
Collapse
|
18
|
Abstract
Gastric cancer is common in China, and its early diagnosis and treatment are difficult. In recent years great progress has been achieved in gene therapy, and a wide array of gene therapy systems for gastric cancer has been investigated. The present article deals with the general principles of gene therapy and then focuses on how these principles may be applied to gastric cancer.
Collapse
Affiliation(s)
- Chao Zhang
- Department of General Surgery, Southwest Hospital, Third Military Medical University, Gaotan Yan, Chongqing 400038, China.
| | | |
Collapse
|
19
|
Abstract
Over the past ten years significant advances have been made in the fields of gene therapy and tumour immunology, such that there now exists a considerable body of evidence validating the proof in the principle of gene therapy based cancer vaccines. While clinical benefit has so far been marginal, data from preclinical and early clinical trials of gene therapy combined with standard therapies are strongly suggestive of additional benefit. Many reasons have been proposed to explain the paucity of clinical responses to single agent vaccination strategies including the poor antigenicity of tumour cells and the development of tolerance through down-regulation of MHC, costimulatory, signal transduction, and other molecules essential for the generation of strong immune responses. In addition, there is now evidence from animal models that the growing tumour may actively inhibit the host immune response. Removal of the primary tumour prior to T cell transfer from the spleen of cancer bearing animals, led to effective tumour cell line specific immunity in the recipient mouse suggesting that there is an ongoing tumour-host interaction. This model also illustrates the potential difficulties of clinical vaccine trials in patients with advanced stage disease.
Collapse
Affiliation(s)
- Susy M Scholl
- Department of Medical Oncology, Institut Curie, 26 rue D'ulm 75248, Paris Cedex 05, France
| | - Silke Michaelis
- Department of Medical Oncology, Institut Curie, 26 rue D'ulm 75248, Paris Cedex 05, France
| | - Ray McDermott
- Department of Medical Oncology, Institut Curie, 26 rue D'ulm 75248, Paris Cedex 05, France
| |
Collapse
|
20
|
Salloum RM, Mauceri HJ, Hanna NN, Gorski DH, Posner MC, Weichselbaum RR. Dual Induction of the Epo-Egr-TNF-α Plasmid in Hypoxic Human Colon Adenocarcinoma Produces Tumor Growth Delay. Am Surg 2003. [DOI: 10.1177/000313480306900105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gene therapy is a modality for the treatment of solid tumors that involves the introduction of a suicide gene into the tumor cells. Genetic radiotherapy involves the placement of a radiation-sensitive promoter upstream from a suicide gene. Because of their irregular vasculature some solid tumors are chronically hypoxic and hence are resistant to conventional treatment with chemotherapy and ionizing radiation (IR). The purpose of this study was to demonstrate that regional tumor hypoxia could be exploited to improve local tumor control. The cDNA coding the erythropoietin hypoxia-responsive element (EPO) was placed upstream from the Egr-TNF-α construct. WIDR human colon adenocarcinoma cells were injected into the right hind limb of nude mice and treated with Epo-Egr-TNF-α plasmid with or without IR. Tumor volumes were measured by calipers and tumor necrosis factor (TNF)-α content of the tumor was determined by enzyme-linked immunosorbent assay. Treatment with the combined regimen of Epo-Egr-TNF-α plasmid + IR resulted in significant tumor growth delay. Tumor TNF-α content was increased by 30 per cent in the combined treatment group compared with each treatment alone. Regional tumor hypoxia can be exploited successfully to induce tumor growth delay, enhance local control, and enhance the therapeutic ratio.
Collapse
Affiliation(s)
- Rabih M. Salloum
- Department of Surgery, University of Rochester School of Medicine, Rochester, New York
| | | | - Nader N. Hanna
- Department of Surgery, University of Kentucky School of Medicine, Lexington, Kentucky
| | - David H. Gorski
- Department of Surgery, University of Medicine and Dentistry of New Jersey, New Brunswick, New Jersey
| | - Mitchell C. Posner
- Department of Surgery, University of Chicago School of Medicine, Chicago, Illinois
| | | |
Collapse
|
21
|
Nemunaitis J, Edelman J. Selectively replicating viral vectors. Cancer Gene Ther 2002; 9:987-1000. [PMID: 12522438 DOI: 10.1038/sj.cgt.7700547] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2002] [Indexed: 01/26/2023]
Affiliation(s)
- John Nemunaitis
- US Oncology, Inc., Collins Building, 5th Floor, Dallas, Texas 75246, USA.
| | | |
Collapse
|
22
|
Abstract
The feasibility of using adenoviruses for gene therapy has been under close scrutiny recently, as it has become clear that significant toxicity can result from the strong immune response created by intravenous administration of large doses of first generation adenovirus vectors. This suggests that other vectors could be more useful for treatment of metabolic and hereditary disease, where widespread transduction is often necessary for effective gene replacement, and the viability of target cells is important. However, promising recent results in human cancer trials have confirmed that adenoviruses can be very useful in oncology. For cancer treatment, the unparalleled transduction efficacy of adenovirus in dividing and dormant cells is a major benefit. As the goal in cancer gene therapy is to kill infected tumour cells, long-term transgene expression is not necessary. In addition, the immune response generated against infected cells could be useful for eradicating uninfected tumour. Importantly, more than 670 cancer patients have been treated with adenovirus intratumorally, intra-arterially, intraperitoneally and intravenously with very manageable adverse effects and no unexpected severe or lethal toxicity. Currently, the most promising approaches are based on replication-competent agents that allow efficient tumour penetration because of their capacity for tissue-specific replication. In addition to transcriptional control, it is becoming clear that targeting is necessary for efficient tumour transduction and less infection of normal tissues. Exciting results are anticipated when the first selectively replicating targeted adenoviruses go to clinical trials. In conclusion, intense gene therapy and virological research have suggested that while other vectors could be more useful for treatment of hereditary disease, adenoviruses are highly promising and safe agents for oncology, as suggested in a number of early phase clinical trials.
Collapse
Affiliation(s)
- Akseli Hemminki
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | |
Collapse
|
23
|
van der Poel HG, Molenaar B, van Beusechem VW, Haisma HJ, Rodriguez R, Curiel DT, Gerritsen WR. Epidermal growth factor receptor targeting of replication competent adenovirus enhances cytotoxicity in bladder cancer. J Urol 2002. [PMID: 12050554 DOI: 10.1016/s0022-5347(05)64905-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE We evaluated the delivery and oncolytic potential of targeted replication competent adenoviruses in bladder cancer lines. MATERIALS AND METHODS Seven established human bladder cancer tumor lines (5637, SW800, TCCsup, J82, Scaber, T24 and 253J) were studied for the expression of integrins alpha(v)beta3, alpha(v)beta5, Coxsackievirus and adenovirus receptor, epidermal growth factor receptor (EGF-R) and epithelial cell adhesion molecule antigens using flow cytometry analysis. Bispecific single chain Fv fragments were used to target replication deficient luciferase reporter adenovirus to EGF-R (425-s11) or to epithelial cell adhesion molecule (C28-s11) antigens. Moreover, a fiber modified adenovirus targeting alpha(v)-integrins was studied. Replication competent serotype-5 adenoviruses attenuated to replicate specifically in retinoblastoma pRb (Ad5-d24) or p53 deficient (Ad5-d55K) cells were tested in vitro for oncolytic properties. RESULTS Low to absent Coxsackievirus and adenovirus receptor expression was found in 5 of the 7 tumor lines (SW800, J82, T24, 5637 and Scaber). EGF-R expression was found in all cell lines, whereas elevated epithelial cell adhesion molecule expression was seen in 3 (5637, Scaber and TCCsup), alpha(v)beta3-integrin was found in 1 (Scaber) and alpha(v)beta5-integrin was found in 3 (TCCsup, 253J and T24). EGF-R targeting using 425-s11 improved transgene expression in all cell lines from 2.1 to 12.5 times over nontargeted viruses. Epithelial cell adhesion molecule and integrin targeting was inferior to EGF-R targeting with a maximal increase in transgene expression of 2 times for epithelial cell adhesion molecule in 5637cells and 1.6 times for integrin targeting in T24 cells. Comparison of the wild-type replication competent virus with conditionally replicating adenoviruses (Ad5-d55K and Ad5-d24) showed superior oncolytic activity for the latter 2 in all lines. Furthermore, improved cytotoxicity (29% to 33%) was obtained in 4 of the 7 lines after pre-incubation of Ad5-d24 with 425-s11. CONCLUSIONS EGF-R directed bispecific single chain antibodies enhance adenovirus mediated transgene expression and oncolysis in bladder cancer lines.
Collapse
Affiliation(s)
- H G van der Poel
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
24
|
van der Poel HG, Molenaar B, van Beusechem VW, Haisma HJ, Rodriguez R, Curiel DT, Gerritsen WR. Epidermal growth factor receptor targeting of replication competent adenovirus enhances cytotoxicity in bladder cancer. J Urol 2002; 168:266-72. [PMID: 12050554 DOI: 10.1097/00005392-200207000-00089] [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] [Indexed: 11/25/2022]
Abstract
PURPOSE We evaluated the delivery and oncolytic potential of targeted replication competent adenoviruses in bladder cancer lines. MATERIALS AND METHODS Seven established human bladder cancer tumor lines (5637, SW800, TCCsup, J82, Scaber, T24 and 253J) were studied for the expression of integrins alpha(v)beta3, alpha(v)beta5, Coxsackievirus and adenovirus receptor, epidermal growth factor receptor (EGF-R) and epithelial cell adhesion molecule antigens using flow cytometry analysis. Bispecific single chain Fv fragments were used to target replication deficient luciferase reporter adenovirus to EGF-R (425-s11) or to epithelial cell adhesion molecule (C28-s11) antigens. Moreover, a fiber modified adenovirus targeting alpha(v)-integrins was studied. Replication competent serotype-5 adenoviruses attenuated to replicate specifically in retinoblastoma pRb (Ad5-d24) or p53 deficient (Ad5-d55K) cells were tested in vitro for oncolytic properties. RESULTS Low to absent Coxsackievirus and adenovirus receptor expression was found in 5 of the 7 tumor lines (SW800, J82, T24, 5637 and Scaber). EGF-R expression was found in all cell lines, whereas elevated epithelial cell adhesion molecule expression was seen in 3 (5637, Scaber and TCCsup), alpha(v)beta3-integrin was found in 1 (Scaber) and alpha(v)beta5-integrin was found in 3 (TCCsup, 253J and T24). EGF-R targeting using 425-s11 improved transgene expression in all cell lines from 2.1 to 12.5 times over nontargeted viruses. Epithelial cell adhesion molecule and integrin targeting was inferior to EGF-R targeting with a maximal increase in transgene expression of 2 times for epithelial cell adhesion molecule in 5637cells and 1.6 times for integrin targeting in T24 cells. Comparison of the wild-type replication competent virus with conditionally replicating adenoviruses (Ad5-d55K and Ad5-d24) showed superior oncolytic activity for the latter 2 in all lines. Furthermore, improved cytotoxicity (29% to 33%) was obtained in 4 of the 7 lines after pre-incubation of Ad5-d24 with 425-s11. CONCLUSIONS EGF-R directed bispecific single chain antibodies enhance adenovirus mediated transgene expression and oncolysis in bladder cancer lines.
Collapse
Affiliation(s)
- H G van der Poel
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
25
|
Kruyt FAE, Curiel DT. Toward a new generation of conditionally replicating adenoviruses: pairing tumor selectivity with maximal oncolysis. Hum Gene Ther 2002; 13:485-95. [PMID: 11874627 DOI: 10.1089/10430340252809784] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Conditionally replicating adenoviruses (CRADs) represent a promising new platform for the treatment of cancer. CRADs have been demonstrated to kill tumor cells when other therapies fail, indicating that their antitumor properties are complementary to, and distinct from, those of standard treatments such as chemotherapy and radiation. In clinic trials CRADs have shown encouraging results, demonstrating mild side effects when administered at high doses and via different routes, including intratumorally, intraperitoneally, and intravenously. Tumor-selective replication has been detected, although as a single agent the efficacy appears to be limited. Interestingly, combined treatment with radiation or chemotherapy has been found to enhance CRAD efficacy considerably. To date, the molecular mechanisms underlying adenovirus-mediated oncolysis, and the way in which chemotherapy enhances oncolysis, are not well understood. A fuller knowledge of these processes will open up new strategies to improve the cell-killing potential of CRADs. Here, we discuss several possibilities that may lead to CRADs with enhanced oncolytic activity. These approaches include strategies to functionally couple tumor targeting and optimal oncolytic activity, and ways to further increase tumor cell disruption at later stages of infection to facilitate the spreading of virus throughout the tumor mass. In addition, improved methods to evaluate the efficacy of these agents in animal models, and in the clinic, will be required to systematically test and optimize CRAD efficacy, also taking into account the influence of tumor characteristics and the administration route.
Collapse
Affiliation(s)
- Frank A E Kruyt
- Division of Gene Therapy, Department of Medical Oncology, VU University Medical Center, NL-1007 MB Amsterdam, The Netherlands
| | | |
Collapse
|
26
|
Abstract
The delineation of the molecular basis of neoplasia provides the possibility of specific intervention by gene therapy through the introduction of genetic material for therapeutic purposes. In this regard, several gene therapy approaches have been developed for the treatment of cancer: mutation compensation, genetic immunopotentiation, molecular chemotherapy, inhibition of angiogenesis, replicative vector oncolysis, and chemosensitization or radiosensitization. Clinical trials have been initiated to evaluate safety, toxicity, and efficacy of each of these approaches, based on promising preclinical results. Various limitations that have been identified include lack of in vivo selective tumor delivery of vectors, minimal expression of therapeutic genes, immune response against vectors, and normal tissue toxicity. Combined modality therapy with gene therapy and chemotherapy or radiation therapy has shown promising results. It is expected that as new therapeutic targets and approaches are identified, combined with advances in vector design, that gene therapy will play an increasing role in clinical cancer treatment.
Collapse
Affiliation(s)
- D J Buchsbaum
- Department of Radiation Oncology, University of Alabama at Birmingham, 1530 3rd Avenue South, WTI 674, Birmingham, AL 35294, USA.
| | | |
Collapse
|