1
|
Yang K, Feng S, Luo Z. Oncolytic Adenovirus, a New Treatment Strategy for Prostate Cancer. Biomedicines 2022; 10:biomedicines10123262. [PMID: 36552019 PMCID: PMC9775875 DOI: 10.3390/biomedicines10123262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Prostate cancer is the most common cancer and one of the leading causes of cancer mortality in males. Androgen-deprivation therapy (ADT) is an effective strategy to inhibit tumour growth at early stages. However, 10~50% of cases are estimated to progress to metastatic castration-resistant prostate cancer (mCRPC) which currently lacks effective treatments. Clinically, salvage treatment measures, such as endocrine therapy and chemotherapy, are mostly used for advanced prostate cancer, but their clinical outcomes are not ideal. When the existing clinical therapeutic methods can no longer inhibit the development of advanced prostate cancer, human adenovirus (HAdV)-based gene therapy and viral therapy present promising effects. Pre-clinical studies have shown its powerful oncolytic effect, and clinical studies are ongoing to further verify its effect and safety in prostate cancer treatment. Targeting the prostate by HAdV alone or in combination with radiotherapy and chemotherapy sheds light on patients with castration-resistant and advanced prostate cancer. This review summarizes the advantages of oncolytic virus-mediated cancer therapy, strategies of HAdV modification, and existing preclinical and clinical investigations of HAdV-mediated gene therapy to further evaluate the potential of oncolytic adenovirus in prostate cancer treatment.
Collapse
Affiliation(s)
- Kaiyi Yang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
- Correspondence: (K.Y.); (Z.L.)
| | - Shenghui Feng
- Provincial Key Laboratory of Tumour Pathogens and Molecular Pathology, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhijun Luo
- Provincial Key Laboratory of Tumour Pathogens and Molecular Pathology, Queen Mary School, Nanchang University, Nanchang 330031, China
- Correspondence: (K.Y.); (Z.L.)
| |
Collapse
|
2
|
Hu H, Xia Q, Hu J, Wang S. Oncolytic Viruses for the Treatment of Bladder Cancer: Advances, Challenges, and Prospects. J Clin Med 2022; 11:jcm11236997. [PMID: 36498574 PMCID: PMC9738443 DOI: 10.3390/jcm11236997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Bladder cancer is one of the most prevalent cancers. Despite recent advancements in bladder cancer therapy, new strategies are still required for improving patient outcomes, particularly for those who experienced Bacille Calmette-Guerin failure and those with locally advanced or metastatic bladder cancer. Oncolytic viruses are either naturally occurring or purposefully engineered viruses that have the ability to selectively infect and lyse tumor cells while avoiding harming healthy cells. In light of this, oncolytic viruses serve as a novel and promising immunotherapeutic strategy for bladder cancer. A wide diversity of viruses, including adenoviruses, herpes simplex virus, coxsackievirus, Newcastle disease virus, vesicular stomatitis virus, alphavirus, and vaccinia virus, have been studied in many preclinical and clinical studies for their potential as oncolytic agents for bladder cancer. This review aims to provide an overview of the advances in oncolytic viruses for the treatment of bladder cancer and highlights the challenges and research directions for the future.
Collapse
Affiliation(s)
| | | | - Jia Hu
- Correspondence: (J.H.); (S.W.)
| | | |
Collapse
|
3
|
Efficacy and Safety of Oncolytic Viruses in Randomized Controlled Trials: A Systematic Review and Meta-Analysis. Cancers (Basel) 2020; 12:cancers12061416. [PMID: 32486227 PMCID: PMC7352817 DOI: 10.3390/cancers12061416] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
Oncolytic virotherapy is a promising antitumor therapeutic strategy. It is based on the ability of viruses to selectively kill cancer cells and induce host antitumor immune responses. However, the clinical outcomes of oncolytic viruses (OVs) vary widely. Therefore, we performed a meta-analysis to illustrate the efficacy and safety of oncolytic viruses. The Cochrane Library, PubMed, and EMBASE databases were searched for randomized controlled trials (RCTs) published up to 31 January 2020. The data for objective response rate (ORR), overall survival (OS), progression-free survival (PFS), and adverse events (AEs) were independently extracted by two investigators from 11 studies that met the inclusion criteria. In subgroup analyses, the objective response rate benefit was observed in patients treated with oncolytic DNA viruses (odds ratio (OR) = 4.05; 95% confidence interval (CI): 1.96–8.33; p = 0.0002), but not in those treated with oncolytic RNA viruses (OR = 1.00, 95% CI: 0.66–1.52, p = 0.99). Moreover, the intratumoral injection arm yielded a statistically significant improvement (OR = 4.05, 95% CI: 1.96–8.33, p = 0.0002), but no such improvement was observed for the intravenous injection arm (OR = 1.00, 95% CI: 0.66–1.52, p = 0.99). Among the five OVs investigated in RCTs, only talimogene laherparepvec (T-VEC) effectively prolonged the OS of patients (hazard ratio (HR), 0.79; 95% CI: 0.63–0.99; p = 0.04). None of the oncolytic virotherapies improved the PFS (HR = 1.00, 95% CI: 0.85–1.19, p = 0.96). Notably, the pooled rate of severe AEs (grade ≥3) was higher for the oncolytic virotherapy group (39%) compared with the control group (27%) (risk difference (RD), 12%; risk ratio (RR), 1.44; 95% CI: 1.17–1.78; p = 0.0006). This review offers a reference for fundamental research and clinical treatment of oncolytic viruses. Further randomized controlled trials are needed to verify these results.
Collapse
|
4
|
Fan J, Liu Y, Liu L, Huang Y, Li X, Huang W. A Multifunction Lipid-Based CRISPR-Cas13a Genetic Circuit Delivery System for Bladder Cancer Gene Therapy. ACS Synth Biol 2020; 9:343-355. [PMID: 31891494 DOI: 10.1021/acssynbio.9b00349] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The treatment of bladder cancer has recently shown minimal progress. Gene therapy mediated by CRISPR provides a new option for bladder cancer treatment. In this study, we developed a versatile liposome system to deliver the CRISPR-Cas13a gene circuits into bladder cancer cells. After in vitro studies and intravesical perfusion studies in mice, this system showed five advantages: (1) CRISPR-Cas13a, a transcriptional targeting and cleavage tool for gene expression editing, did not affect the stability of the cell genome; (2) the prepared liposome systems were targeted to hVEGFR2, which is always highly expressed in bladder cancer cells; (3) the CRISPR-Cas13a sequence was driven by an artificial tumor specific promoter to achieve further targeting; (4) a near-infrared photosensitizer released using near-infrared light was introduced to control the delivery system; and (5) the plasmids were constructed with three crRNA tandem sequences to achieve multiple targeting and wider therapeutic results. This tumor cell targeting lipid delivery system with near-infrared laser-controlled ability provided a versatile strategy for CRISPR-Cas13a based gene therapy of bladder cancer.
Collapse
Affiliation(s)
- Jing Fan
- Department of Urology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518039, China
- Reproductive Medicine Center, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
- The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yuchen Liu
- Department of Urology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518039, China
| | - Lisa Liu
- Department of Urology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518039, China
| | - Yikun Huang
- Department of Urology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518039, China
| | - Xuemei Li
- Reproductive Medicine Center, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
| | - Weiren Huang
- Department of Urology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518039, China
| |
Collapse
|
5
|
Tessmann JW, Buss J, Begnini KR, Berneira LM, Paula FR, de Pereira CMP, Collares T, Seixas FK. Antitumor potential of 1-thiocarbamoyl-3,5-diaryl-4,5-dihydro-1H-pyrazoles in human bladder cancer cells. Biomed Pharmacother 2017; 94:37-46. [PMID: 28750358 DOI: 10.1016/j.biopha.2017.07.060] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/01/2017] [Accepted: 07/09/2017] [Indexed: 11/19/2022] Open
Abstract
Bladder cancer is a genitourinary malignant disease common worldwide. Current chemotherapy is often limited mainly due to toxicity and drug resistance. Thus, there is a continued need to discover new therapies. Recently evidences shows that pyrazoline derivatives are promising antitumor agents in many types of cancers, but there are no studies with bladder cancer. In order to find potent and novel chemotherapy drugs for bladder cancer, a series of pyrazoline derivatives 2a-2d were tested for their antitumor activity in two human bladder cancer cell lines 5647 and T24. The MTT assay showed that the compounds 1-thiocarbamoyl-3,5-diphenyl-4,5-dihydro-1H-pyrazole (2a) and 1-thiocarbamoyl-5-(4-chlorophenyl)-3-phenyl-4,5-dihydro-1H-pyrazole (2c) decrease the cell viability of 5637 cells. Molecular modeling indicated that these compounds had a good oral bioavailability and low toxicities. Clonogenic assay and flow cytometric analysis were used to assess colony formation, apoptosis induction and cell cycle distribution. Overall, our results suggest that pyrazoline 2a and 2c, with the substituents hydrogen and chlorine respectively, may decrease cell viability and colony formation of bladder cancer 5637 cell line by inhibition of cell cycle progression, and for pyrazoline 2a, by induction of apoptosis. As indicated by the physicochemical properties of these compounds, the steric factor influences the activity. Therefore, these pyrazoline derivatives can be considered promising anticancer agents for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Josiane Weber Tessmann
- Programa de Pós-Graduação em Biotecnologia (PPGB), Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil; Grupo de Pesquisa em Oncologia Celular e Molecular (GPO), Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil.
| | - Julieti Buss
- Grupo de Pesquisa em Oncologia Celular e Molecular (GPO), Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil.
| | - Karine Rech Begnini
- Programa de Pós-Graduação em Biotecnologia (PPGB), Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil; Grupo de Pesquisa em Oncologia Celular e Molecular (GPO), Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil.
| | - Lucas Moraes Berneira
- Laboratório de Lipidômica e Bio-orgânica, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil.
| | - Favero Reisdorfer Paula
- Laboratório de Desenvolvimento e Controle de Qualidade em Medicamentos, Universidade Federal do Pampa, BR 472 - Km 592, Uruguaiana, RS, Cep: 97508-000, Brazil.
| | - Claudio Martin Pereira de Pereira
- Laboratório de Lipidômica e Bio-orgânica, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil; Programa de Pós-Graduação em Bioquímica e Bioprospecção, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil.
| | - Tiago Collares
- Programa de Pós-Graduação em Biotecnologia (PPGB), Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil; Grupo de Pesquisa em Oncologia Celular e Molecular (GPO), Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil; Programa de Pós-Graduação em Bioquímica e Bioprospecção, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil.
| | - Fabiana Kömmling Seixas
- Programa de Pós-Graduação em Biotecnologia (PPGB), Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil; Grupo de Pesquisa em Oncologia Celular e Molecular (GPO), Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário s/n, Capão do Leão, RS, Cep: 96010-900, Brazil.
| |
Collapse
|
6
|
Svyatchenko VA, Tarasova MV, Netesov SV, Chumakov PM. Oncolytic adenoviruses in anticancer therapy: Current status and prospects. Mol Biol 2012. [DOI: 10.1134/s0026893312040103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
7
|
Virotherapy induces massive infiltration of neutrophils in a subset of tumors defined by a strong endogenous interferon response activity. Cancer Gene Ther 2011; 18:785-94. [PMID: 21869820 PMCID: PMC3196785 DOI: 10.1038/cgt.2011.46] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Oncolytic virotherapy has shown substantial promises as an alternative therapeutic modality for solid tumors in both preclinical studies and clinical trials. The main therapeutic activity of virotherapy derives from the direct lytic effect associated with virus replication and the induction of host immune responses to the infected tumor cells. Here we show that some human and murine tumor cell lines are highly resistant to the lytic effect of a type II herpes simplex virus-derived oncolytic virus, FusOn-H2, which was constructed by deleting the N-terminal region of the ICP10 gene. However, these tumor cells still respond exceptionally well to FusOn-H2 virotherapy in vivo. Histological examination of the treated tumors revealed that, in contrast to tumors supporting FusOn-H2 replication, implants of these highly resistant lines showed massive infiltration of neutrophils after virotherapy. Further analysis showed that there is a correlation between an intrinsically strong interferon response activity and the recruitment of neutrophils in these tumors. These results suggest that an innate immune response mainly represented by neutrophils may be part of the virotherapy-mediated antitumor mechanism in these tumors.
Collapse
|
8
|
Toth K, Dhar D, Wold WSM. Oncolytic (replication-competent) adenoviruses as anticancer agents. Expert Opin Biol Ther 2010; 10:353-68. [PMID: 20132057 DOI: 10.1517/14712590903559822] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE OF THE FIELD Whilst therapies for neoplasies have advanced tremendously in the last few decades, there is still a need for new anti-cancer treatments. One option is genetically-engineered oncolytic adenovirus (Ad) 'vectors'. These kill cancer cells via the viral replication cycle, and amplify the anti-tumor effect by producing progeny virions able to infect neighboring tumor cells. AREAS COVERED IN THIS REVIEW We provide a description of basic Ad biology and summarize the literature for oncolytic Ads from 1996 to the present. WHAT THE READER WILL GAIN An overall view of oncolytic Ads, the merits and drawbacks of the various features of these vectors, and obstacles to further development and future directions for research. TAKE HOME MESSAGE Ads are attractive for gene therapy because they are relatively innocuous, easy to produce in large quantities, genetically stable, and easy to manipulate. A variety of have been constructed and tested, in pre-clinical and clinical experiments. Oncolytic Ads proved to be remarkably safe; no dose-limiting toxicity was observed in any clinical trial, and the maximum tolerated dose was not reached. At present, the major challenge for researchers is to increase the efficacy of the vectors, and to incorporate oncolytic virotherapy into existing treatment protocols.
Collapse
Affiliation(s)
- Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA.
| | | | | |
Collapse
|
9
|
Retargeted adenoviral cancer gene therapy for tumour cells overexpressing epidermal growth factor receptor or urokinase-type plasminogen activator receptor. Gene Ther 2010; 17:1000-10. [PMID: 20410926 DOI: 10.1038/gt.2010.45] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We have assessed the ability of bispecific fusion proteins to improve adenovirus-mediated transfer of therapeutic and marker transgenes. We constructed an expression vector that can be easily modified to synthesize a variety of fusion proteins for retargeting adenoviral gene therapy vectors to cell surface markers, which are differentially expressed between normal and cancer cells. Adenoviral transduction can be improved in a number of tumour cell lines which overexpress EGFR (epidermal growth factor receptor) or uPAR (urokinase-type plasminogen activator receptor), but which have only low levels of endogenous hCAR (human coxsackie B and adenovirus receptor) expression. Up to 40-fold improvement in beta-galactosidase transgene expression was seen using an EGFR retargeting protein, and up to 16-fold using a second fusion protein targeting uPAR. In vitro, our uPAR retargeting fusion protein improved the sensitivity to adenoviral herpes simplex virus thymidine kinase/ganciclovir by an order of magnitude, whereas in vivo, our EGFR retargeting protein is able to significantly delay tumour growth in rodent animal models in a dose-dependent manner. The 'cassette' design of our fusion protein constructs offers a flexible method for the straightforward synthesis of multiple adenoviral retargeting proteins, directed against a variety of tumour-associated antigens, for use in clinical trials.
Collapse
|
10
|
Lu Y, Madu CO. Viral-based gene delivery and regulated gene expression for targeted cancer therapy. Expert Opin Drug Deliv 2010; 7:19-35. [PMID: 19947888 DOI: 10.1517/17425240903419608] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
IMPORTANCE OF THE FIELD Cancer is both a major health concern and a care-cost issue in the US and the rest of the world. It is estimated that there will be a total of 1,479,350 new cancer cases and 562,340 cancer deaths in 2009 within the US alone. One of the major obstacles in cancer therapy is the ability to target specifically cancer cells. Most existing chemotherapies and other routine therapies (such as radiation therapy and hormonal manipulation) use indiscriminate approaches in which both cancer cells and non-cancerous surrounding cells are treated equally by the toxic treatment. As a result, either the cancer cell escapes the toxic dosage necessary for cell death and consequently resumes replication, or an adequate lethal dose that kills the cancer cell also causes the cancer patient to perish. Owing to this dilemma, cancer- or organ/tissue-specific targeting is greatly desired for effective cancer treatment and the reduction of side effect cytotoxicity within the patient. AREAS COVERED IN THIS REVIEW In this review, the strategies of targeted cancer therapy are discussed, with an emphasis on viral-based gene delivery and regulated gene expression. WHAT THE READER WILL GAIN Numerous approaches and updates in this field are presented for several common cancer types. TAKE HOME MESSAGE A summary of existing challenges and future directions is also included.
Collapse
Affiliation(s)
- Yi Lu
- University of Tennessee Health Science Center, Department of Pathology and Laboratory Medicine, Cancer Research Building, Room 218, 19 South Manassas Street, Memphis, TN 38163, USA.
| | | |
Collapse
|
11
|
Kasman LM, Barua S, Lu P, Rege K, Voelkel-Johnson C. Polymer-enhanced adenoviral transduction of CAR-negative bladder cancer cells. Mol Pharm 2009; 6:1612-9. [PMID: 19655763 DOI: 10.1021/mp9000958] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The application of adenoviral gene therapy for cancer is limited by immune clearance of the virus as well as poor transduction efficiency, since the protein used for viral entry (CAR) serves physiological functions in adhesion and is frequently decreased among cancer cells. Cationic polymers have been used to enhance adenoviral gene delivery, but novel polymers with low toxicity are needed to realize this approach. We recently identified polymers that were characterized by high transfection efficiency of plasmid DNA and a low toxicity profile. In this study we evaluated the novel cationic polymer EGDE-3,3' for its potential to increase adenoviral transduction of the CAR-negative bladder cancer cell line TCCSUP. The amount of adenovirus required to transduce 50-60% of the cells was reduced 100-fold when Ad.GFP was preincubated with the EGDE-3,3' polymer. Polyethyleneimine (pEI), a positively charged polymer currently used as a standard for enhancing adenoviral transduction, also increased infectivity, but transgene expression was consistently higher with EGDE-3,3'. In addition, EGDE-3,3'-supplemented transduction of an adenovirus expressing an apoptosis inducing transgene, Ad.GFP-TRAIL, significantly enhanced the amount of cell death. Thus, our results indicate that novel biocompatible polymers may be useful in improving the delivery of adenoviral gene therapy.
Collapse
Affiliation(s)
- Laura M Kasman
- Department of Microbiology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | |
Collapse
|
12
|
Advances in Gene Therapy for Bladder Cancer. Bladder Cancer 2009. [DOI: 10.1007/978-1-59745-417-9_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Plattner VE, Wagner M, Ratzinger G, Gabor F, Wirth M. Targeted drug delivery: Binding and uptake of plant lectins using human 5637 bladder cancer cells. Eur J Pharm Biopharm 2008; 70:572-6. [DOI: 10.1016/j.ejpb.2008.06.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 06/10/2008] [Accepted: 06/10/2008] [Indexed: 11/29/2022]
|
14
|
|
15
|
Yang ZR, Wang HF, Zhao J, Peng YY, Wang J, Guinn BA, Huang LQ. Recent developments in the use of adenoviruses and immunotoxins in cancer gene therapy. Cancer Gene Ther 2007; 14:599-615. [PMID: 17479105 DOI: 10.1038/sj.cgt.7701054] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite setbacks in the past and apparent hurdles ahead, gene therapy is advancing toward reality. The past several years have witnessed this new field of biomedicine developing rapidly both in breadth and depth, especially for the treatment of cancer, thanks largely to the better understanding of molecular and genetic basis of oncogenesis and the development of new and improved vectors and technologies for gene delivery and targeting. This article is intended to provide a brief review of recent advances in cancer gene therapy using adenoviruses, both as vectors and as oncolytic agents, and some of the recent progress in the development of immunotoxins for use in cancer gene therapy.
Collapse
Affiliation(s)
- Z R Yang
- Center for Biotech & BioMedicine and Division of Life Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | | | | | | | | | | | | |
Collapse
|
16
|
Witlox M, Lamfers M, Wuisman P, Curiel D, Siegal G. Evolving gene therapy approaches for osteosarcoma using viral vectors: review. Bone 2007; 40:797-812. [PMID: 17189720 PMCID: PMC2731716 DOI: 10.1016/j.bone.2006.10.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Revised: 10/25/2006] [Accepted: 10/26/2006] [Indexed: 12/11/2022]
Abstract
This review begins with an introduction to the malignant bone tumor, osteosarcoma [OS] and then moves to a discussion of the commonly used vectors for gene transfer. We first briefly highlight non-viral vectors including polymeric and liposomal delivery systems but concentrate predominantly on the 5 leading viral vectors used in cancer gene therapy, specifically retroviruses, adeno-associated viruses, herpes viruses and lentiviruses with the most detailed analysis reserved for adenoviruses. The 3 main strategies for gene therapy in osteosarcoma are next summarized. As part of this review, the several prodrug-converting enzymes utilized in OS suicide gene therapy are examined. The text then turns to a discussion of adenovirus-mediated gene transfer and the need for tumor targeting via transductional or transcriptional approaches. Because of practical problems with use of replication-incompetent viruses in achieving complete tumor kill in vivo, virotherapy utilizing replication competent viruses has come to the fore. This topic is, thus, next reviewed which allows for a natural transition to a discussion of armed therapeutic viruses many of which are conditionally replicating adenoviruses carrying transgenes with established anti-tumor efficacy. We recognize that several other issues have arisen which hamper progress in the field of cancer gene therapy. We, therefore, review viral-induced toxicity in the host and vector delivery issues which have been found to potentially influence safety. We end with a brief perspective including commenting on animal models used in examining delivery strategies for osteosarcoma gene therapy. The challenges remaining are touched upon most especially the need to deal with pulmonary metastatic disease from OS.
Collapse
Affiliation(s)
- M.A. Witlox
- Department of Orthopedic Surgery, VU University Medical Center, Amsterdam, NL, ,
- Divison of Gene Therapy, Department of Medical Oncology, VU University Medical Center, Amsterdam, NL
| | - M.L. Lamfers
- Department of Neurosurgery, VU University Medical Center, Amsterdam, NL,
| | - P.I.J.M. Wuisman
- Department of Orthopedic Surgery, VU University Medical Center, Amsterdam, NL, ,
| | - D.T. Curiel
- Division of Human Gene Therapy, Depts. Of Medicine, Surgery, Pathology & Ob/Gyn and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL, USA,
| | - G.P. Siegal
- Departments of Pathology, Cell Biology, and Surgery, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL, USA,
| |
Collapse
|
17
|
Paraskevakou G, Allen C, Nakamura T, Zollman P, James CD, Peng KW, Schroeder M, Russell SJ, Galanis E. Epidermal growth factor receptor (EGFR)-retargeted measles virus strains effectively target EGFR- or EGFRvIII expressing gliomas. Mol Ther 2007; 15:677-86. [PMID: 17299404 DOI: 10.1038/sj.mt.6300105] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
A retargeted measles virus strain MV-GFP-H(AA)-scEGFR was generated by engineering the MV-NSe Edmonston vaccine strain to incorporate both CD46 (Y481A) and signaling lymphocyte activation molecule (SLAM) (R533A) ablating mutations in the hemagglutinin protein in combination with the display of a single-chain antibody against epidermal growth factor receptor (EGFR) at the C terminus of hemagglutinin. The unmodified MV-GFP virus was used as a positive control. Specificity of the EGFR retargeted virus was demonstrated in non-permissive Chinese hamster ovary (CHO) cells stably transfected to express either the natural receptors CD46 or SLAM or the target receptors EGFR and EGFRvIII. In vitro, the retargeted virus had potent antitumor activity against EGFR- or EGFRvIII-overexpressing primary glioblastoma multi-forme (GBM) cell lines that was comparable to the activity of the unmodified MV-GFP virus. Intratumoral administration of MV-GFP-H(AA)-scEGFRvIII in orthotopic GBM12 xenografts resulted in tumor regression, as demonstrated by bioluminescence imaging and significant prolongation of survival, that was comparable to the effect of the unmodified strain. In contrast to MV-GFP, central nervous system administration of the targeted MV-GFP-H(AA)-scEGFR virus in measles replication-permissive Ifnar(ko) CD46 transgenic mice resulted in no neurotoxicity. In conclusion, EGFR-retargeted measles virus strains have comparable therapeutic efficacy to the unmodified virus in glioma cells overexpressing EGFR or EGFRvIII in vivo and in vitro, and improved therapeutic index, a finding with potential translational implications in glioma virotherapy.
Collapse
|
18
|
Fu X, Tao L, Li M, Fisher WE, Zhang X. Effective treatment of pancreatic cancer xenografts with a conditionally replicating virus derived from type 2 herpes simplex virus. Clin Cancer Res 2006; 12:3152-7. [PMID: 16707615 DOI: 10.1158/1078-0432.ccr-06-0045] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Pancreatic cancer is a devastating disease that is almost universally fatal because of the lack of effective treatments. We recently constructed a novel oncolytic virus (FusOn-H2) from the type 2 herpes simplex virus. Because the replication potential of FusOn-H2 depends on the activation of the Ras signaling pathway, we evaluated its antitumor effect against pancreatic cancer, which often harbors K-ras gene mutations. EXPERIMENTAL DESIGN Human pancreatic cancer xenografts were established in nude mice either s.c. or orthotopically (n = 8/group). FusOn-H2 was injected either directly (s.c. tumors) or by the i.v. or i.p. route (orthotopic tumors). Tumor volume, weight, and survival time were recorded for each animal. Statistical analyses were done by Student's t test. RESULTS A single intratumor injection of FusOn-H2 completely eradicated s.c. pancreatic cancers in all animals. Systemic injection of the oncolytic virus produced clear antitumor effects but did not abolish tumors in any animal. The most striking antitumor effect was seen when the virus was given i.p. Delivery of FusOn-H2 by this route completely eradicated established orthotopic tumors in 75% of the animals and completely prevented local metastases. CONCLUSIONS FusOn-H2 has potent activity against human pancreatic cancer xenografts and may be a promising candidate for investigative virotherapy of this malignancy.
Collapse
Affiliation(s)
- Xinping Fu
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
19
|
Abstract
The application of replicating viruses for the treatment of cancers represents a novel therapy that is distinct from traditional treatment modalities. It is apparent that the genetic changes that a virus produces within an infected cell in order to create an environment conducive to viral replication are often similar to the processes involved in cellular transformation. These include uncontrolled cellular proliferation, prevention of apoptosis, and resistance to host organism immune effector mechanisms. Deletions of viral genes involved in these processes have been exploited to produce viral mutants whose replication is selective for transformed cells. The use of tissue-specific transcriptional response or RNA stability elements to control the expression of critical viral genes has also resulted in targeted viruses. Work also is being undertaken to restrict or alter the tropism of viruses by altering their ability to infect certain cell types. Finally, the addition of exogenous genes can be used to increase the virus's lytic potential and/or bystander killing; to further induce the host's immune response against cancer cells; and/or to permit the controlled downregulation of viral replication if necessary. The combination of different tumor-targeting mutations in parallel with the expression of foreign genes has resulted in the evolution of second- and third-generation viruses that continue to become further distinct from their native parental strains. The movement of these viruses into the clinic has begun to demonstrate the potential of this approach in the treatment of cancers.
Collapse
Affiliation(s)
- Stephen H Thorne
- Department of Pediatrics and Bio-X Program, Stanford University School of Medicine, Stanford, CA 94305-5427, USA.
| | | | | |
Collapse
|
20
|
Mathis JM, Stoff-Khalili MA, Curiel DT. Oncolytic adenoviruses - selective retargeting to tumor cells. Oncogene 2005; 24:7775-91. [PMID: 16299537 DOI: 10.1038/sj.onc.1209044] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Virotherapy is an approach for the treatment of cancer, in which the replicating virus itself is the anticancer agent. Virotherapy exploits the lytic property of virus replication to kill tumor cells. As this approach relies on viral replication, the virus can self-amplify and spread in the tumor from an initial infection of only a few cells. The success of this approach is fundamentally based on the ability to deliver the replication-competent viral genome to target cells with a requisite level of efficiency. With virotherapy, while a number of transcriptional retargeting strategies have been utilized to restrict viral replication to tumor cells, this review will focus primarily on transductional retargeting strategies, whereby oncolytic viruses can be designed to selectively infect tumor cells. Using the adenoviral vector paradigm, there are three broad strategies useful for viral retargeting. One strategy uses heterologous retargeting ligands that are bispecific in that they bind both to the viral vector as well as to a cell surface target. A second strategy uses genetically modified viral vectors in which a cellular retargeting ligand is incorporated. A third strategy involves the construction of chimeric recombinant vectors, in which a capsid protein from one virus is exchanged for that of another. These transductional retargeting strategies have the potential for reducing deleterious side effects, and increasing the therapeutic index of virotherapeutic agents.
Collapse
Affiliation(s)
- J Michael Mathis
- Gene Therapy Program, Department of Cellular Biology and Anatomy, LSU Health Sciences Center, Shreveport, LA 71130, USA
| | | | | |
Collapse
|
21
|
Zhang J, Liu YF, Yang SJ, Qiao Q, Cheng H, Zhang CS, Ma FC, Guo HZ. Primary targeting of recombinant Fv-immunotoxin hscFv 25-mTNFα against hepatocellular carcinoma. World J Gastroenterol 2004; 10:1872-5. [PMID: 15222026 PMCID: PMC4572220 DOI: 10.3748/wjg.v10.i13.1872] [Citation(s) in RCA: 4] [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 obtain human recombinant Fv-immunotoxin hscFv25-mTNFα (mutant human TNFα fused to human scFv25) against hepatocellular carcinoma (HCC).
METHODS: Two relevant sites of enzymatic digestion were added to mTNFα by PCR. mTNFα was linked to the 3’ end of hscFv25 in pGEX4T-1 vector. This anti-HCC recombinant Fv-immunotoxin hscFv25-mTNFα was expressed in Escherichia coli and purified from inclusions. After purified by glutathione-S-transferase affinity chromatography and thrombin digestion, it was identified by electrophoresis and Western blot. And then, the purified recombinant Fv-immunotoxin was injected into nude mice with HCC xenografts through their tail veins. mTNFα protein and PBS were used as control at the same time. After treated for two weeks, nude mice were executed. The bulk and weight of tumors were observed. The tumor tissues were stained by immunohistochemical method with TNFα antibody.
RESULTS: The expression ratio of recombinant Fv-immunotoxin hscFv25-mTNFα was 12% of bacterial protein. The result of tumor restraining trials of hscFv25-mTNFα showed 2/5 complete remission and 3/5 partial remission. mTNFα restraining trials showed 5/5 partial remission. The therapeutic result of hscFv25-mTNFα was better than that of mTNFα (F = 8.70, P < 0.05). The hscFv25-mTNFα remedial tumor tissues were positive for TNFα by immunohistochemical staining. The positive granules mainly existed in the cytoplasm of tumor cell.
CONCLUSION: Recombinant Fv-immunotoxin hscFv25-mTNFα has better therapeutic effect than mTNFα. It can inhibit the cellular growth of HCC and has some potential of clinical application.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Belousova N, Korokhov N, Krendelshchikova V, Simonenko V, Mikheeva G, Triozzi PL, Aldrich WA, Banerjee PT, Gillies SD, Curiel DT, Krasnykh V. Genetically targeted adenovirus vector directed to CD40-expressing cells. J Virol 2003; 77:11367-77. [PMID: 14557622 PMCID: PMC229360 DOI: 10.1128/jvi.77.21.11367-11377.2003] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2003] [Accepted: 07/30/2003] [Indexed: 11/20/2022] Open
Abstract
The success of gene therapy depends on the specificity of transgene delivery by therapeutic vectors. The present study describes the use of an adenovirus (Ad) fiber replacement strategy for genetic targeting of the virus to human CD40, which is expressed by a variety of diseased tissues. The tropism of the virus was modified by the incorporation into its capsid of a protein chimera comprising structural domains of three different proteins: the Ad serotype 5 fiber, phage T4 fibritin, and the human CD40 ligand (CD40L). The tumor necrosis factor-like domain of CD40L retains its functional tertiary structure upon incorporation into this chimera and allows the virus to use CD40 as a surrogate receptor for cell entry. The ability of the modified Ad vector to infect CD40-positive dendritic cells and tumor cells with a high efficiency makes this virus a prototype of choice for the derivation of therapeutic vectors for the genetic immunization and targeted destruction of tumors.
Collapse
Affiliation(s)
- Natalya Belousova
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Cote RJ, Datar RH. Therapeutic approaches to bladder cancer: identifying targets and mechanisms. Crit Rev Oncol Hematol 2003; 46 Suppl:S67-83. [PMID: 12850529 DOI: 10.1016/s1040-8428(03)00066-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Transitional cell carcinoma is the second most common genitourinary malignancy in US and third most common cause of death among genitourinary tumors. Treatment options for bladder cancer include surgery, often combined with chemotherapy, radiation, and/or immunotherapy. The MVAC adjuvant chemotherapy regimen has been most widely used in locally invasive as well as metastatic disease. Only a proportion of patients at risk will respond to therapy. There is thus need to identify good responder patients for adjuvant therapy and to identify new targets to treat a greater range of patients. Based upon patient-specific aberrations in pathways or known markers, both existing and new therapies can be tailored to benefit patients based on the risk of progression and molecular alterations specific to a patient's tumor. Targeted therapy, therefore, is defined as therapy that targets mechanism and risk. Utilizing the available knowledge of the molecular biology of cell-cycle regulation, signal transduction, apoptosis, and angiogenesis in bladder cancer, we review the potential therapeutic targets for rational drug development. Finally, using bladder cancer as a model for translational research, requirements for a desired clinical trial are presented.
Collapse
Affiliation(s)
- Richard J Cote
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
| | | |
Collapse
|
24
|
Hsieh JL, Wu CL, Lai MD, Lee CH, Tsai CS, Shiau AL. Gene therapy for bladder cancer using E1B-55 kD-deleted adenovirus in combination with adenoviral vector encoding plasminogen kringles 1-5. Br J Cancer 2003; 88:1492-9. [PMID: 12778082 PMCID: PMC2741042 DOI: 10.1038/sj.bjc.6600908] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mutations or loss of heterozygosity of p53 are detected in approximately 50% of bladder cancers. E1B-55 kD-deleted adenovirus has been shown to kill tumour cells with defective p53 function while sparing normal cells. Here, we examined the cytolytic effect and replication of E1B-55 kD-deleted adenovirus, designated Ad5WS1, on human bladder cancer cell lines with various p53 status. Ad5WS1 caused more severe cytolytic effect and replicated more efficiently in J82 and TCC-SUP bladder cancer cells carrying mutant p53 compared with TSGH-8301 and BFTC-905 bladder cancer cells retaining wild-type p53. Introduction of dominant negative p53 into BFTC-905 cells rendered them more susceptible to Ad5WS1-induced cytolysis. Furthermore, cells susceptible to lysis caused by Ad5WS1 were not attributable to their greater infectability by adenovirus. Finally, Ad5WS1 suppressed the growth of TCC-SUP bladder tumour xenografts, which could be augmented when combined with replication-defective adenoviral vector encoding kringles 1-5 of plasminogen (K1-5), an angiogenic inhibitor. Taken together, our results show that E1B-55 kD-deleted adenovirus replicates and hence lyses bladder cancer cells with mutant p53 much more efficient than those with wild-type p53. Thus, E1B-deleted adenovirus may have therapeutic potential, especially in combination with adenoviral vector expressing K1-5, for the treatment of bladder cancer.
Collapse
Affiliation(s)
- J-L Hsieh
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
| | - C-L Wu
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
- Department of Biochemistry, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
| | - M-D Lai
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
- Department of Biochemistry, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
| | - C-H Lee
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
| | - C-S Tsai
- Department of Microbiology and Immunology, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
| | - A-L Shiau
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
- Department of Microbiology and Immunology, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
- Department of Microbiology and Immunology, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan. E-mail:
| |
Collapse
|
25
|
Zhang J, Liu YF, Yang SJ, Qiao Q, Zhang SZ, Cheng H. Targeting therapeutic of humanized scFv25 and the fusion to mutant TNFα against hepatocellular carcinoma: a preliminary study. Shijie Huaren Xiaohua Zazhi 2003; 11:285-288. [DOI: 10.11569/wcjd.v11.i3.285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the cytotoxic effects of humanized scFv25 and the fusion to mutant TNFα on HCC xenografts in nude mice.
METHODS: The mice with HCC xenografts were injected the purified recombinant immunotoxin through tail vain and executed after two weeks. The bulk and weight of tumor were observed. Expression of TNFα was detected by immunohistochemical staining in the tumor tissues.
RESULTS: The tumor regression trials of hscFv25-mTNFα showed 5/5 effective, with 2/5 completely remission and 3/5 partly remission. The therapeutic result of hscFv25-mTNFα was better than that of mutant TNFα (Xh2 = 6.62, P < 0.05). The HCC tissue treated by hscFv25-mTNFα expressed TNFα, and the positive granules were mainly existed in cytoplasm.
CONCLUSION: Recombinant immunotoxin the hscFV25-mTNFα can regress the growth of HCC with a great potential.
Collapse
|
26
|
Abstract
Although the cytotoxic effects of viruses are usually viewed in terms of pathogenicity, it is possible to harness this activity for therapeutic purposes. Viral genomes are highly versatile, and can be modified to direct their cytotoxicity towards cancer cells. These viruses are known as oncolytic viruses. How are viruses engineered to become tumour specific, and can they be used to safely treat cancer in humans?
Collapse
Affiliation(s)
- E Antonio Chiocca
- Molecular Neuro-Oncology Laboratories, Neurosurgery Service, Massachusetts General Hospital, East Building, 13th Street, Bldg 149, Charlestown, Massachusetts 02129, USA. chioccahelix.mgh.harvard.edu
| |
Collapse
|