1
|
Smith JA, Goldspiel BR. Cancer gene therapy update. J Oncol Pharm Pract 2016. [DOI: 10.1177/107815529900500101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective. To provide an update about gene marking and gene therapy trials in cancer patients. Data Sources. A MEDLINE search using the term “gene therapy” was conducted for the period 1985 to 1998. The reference lists from retrieved articles were reviewed. Meeting abstracts from the American Society of Clinical Oncology annual meeting (published in their proceedings) and the Annual Cancer Gene Therapy Symposium (published in Cancer Gene Therapy) that concerned gene therapy in cancer patients were also included. Data Extraction. Both authors reviewed the retrieved material and included preclinical data, case reports, and clinical trials related to gene transfer or gene therapy in cancer patients. Data Synthesis. There are several possible approaches to using gene therapy for the diagnosis and treatment of cancer and for the monitoring of cancer therapy. Exogenous genes may be used to mark cells to help better understand cancer biology or may be used directly for cancer treatment. Gene-marking trials have already provided new information about cancer biology and have demonstrated that reinfused progenitor cells may be a source of relapse in patients with acute or chronic myelogenous leukemia and neuroblastoma. Approaches using gene therapy for cancer treatment include: using lymphocytes as gene carriers, using foreign genes to increase tumor immunogenicity, introducing tumor regression antigen genes into viruses, introducing “sensitivity” genes to produce new cytotoxic agent(s) within tumors, producing new protein product(s) to protect normal cells, replacing missing or mutant tumor suppressor genes, and inactivating oncogenes. Clinical trials using these strategies have demonstrated that gene transfer is feasible (albeit with low transduction efficiency) and that gene expression occurs; in addition, clinical responses have been noted.
Collapse
Affiliation(s)
- Judith A Smith
- National Institutes of Health Clinical Center Pharmacy Department, Bethesda, Maryland
| | - Barry R Goldspiel
- National Institutes of Health Clinical Center Pharmacy Department, Bethesda, Maryland
| |
Collapse
|
2
|
Saito K, Shirasawa H, Isegawa N, Shiiba M, Uzawa K, Tanzawa H. Oncolytic virotherapy for oral squamous cell carcinoma using replication-competent viruses. Oral Oncol 2009; 45:1021-7. [DOI: 10.1016/j.oraloncology.2009.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 08/29/2009] [Accepted: 09/02/2009] [Indexed: 01/02/2023]
|
3
|
Kirn DH. Section Review: Oncologic, Endocrine & Metabolic: New treatment approaches for metastatic breast cancer. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.4.12.1233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
4
|
Russell SJ, Peng KW. Viruses as anticancer drugs. Trends Pharmacol Sci 2007; 28:326-33. [PMID: 17573126 PMCID: PMC3125087 DOI: 10.1016/j.tips.2007.05.005] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Revised: 04/11/2007] [Accepted: 05/25/2007] [Indexed: 01/14/2023]
Abstract
Oncolytic viruses are being developed as anticancer drugs. They propagate selectively in tumor tissue and destroy it without causing excessive damage to normal non-cancerous tissues. When used as drugs, they must meet stringent criteria for safety and efficacy and be amenable to pharmacological study in human subjects. Specificity for neoplastic tissue is the key to safety, and this goal can be achieved through a variety of ingenious virus-engineering strategies. Antiviral immunity remains a significant barrier to the clinical efficacy of oncolytic viruses but this is being addressed by using novel immune-evasive delivery strategies and immunosuppressive drugs. Noninvasive pharmacokinetic monitoring is facilitated by engineering marker genes into the viral genome. Clinical data on the pharmacokinetics of oncolytic viruses will be the key to accelerating their development and approval as effective anticancer drugs. This review introduces concepts relevant to the use of viruses as anticancer drugs, emphasizing targeting mechanisms as well as safety and efficacy issues that are currently limiting their clinical success.
Collapse
Affiliation(s)
- Stephen J Russell
- Molecular Medicine Program, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| | | |
Collapse
|
5
|
Abstract
Since the turn of the nineteenth century, when their existence was first recognized, viruses have attracted considerable interest as possible agents of tumor destruction. Early case reports emphasized regression of cancers during naturally acquired virus infections, providing the basis for clinical trials where body fluids containing human or animal viruses were used to transmit infections to cancer patients. Most often the viruses were arrested by the host immune system and failed to impact tumor growth, but sometimes, in immunosuppressed patients, infection persisted and tumors regressed, although morbidity as a result of the infection of normal tissues was unacceptable. With the advent of rodent models and new methods for virus propagation, there were numerous attempts through the 1950s and 1960s to force the evolution of viruses with greater tumor specificity, but success was limited and many researchers abandoned the field. Technology employing reverse genetics later brought about a renewal of interest in virotherapy that allowed the generation of more potent, tumor-specific oncolytics. Here, examination of early oncolytic virotherapy before genetic engineering serves to highlight tremendous advances, yet also hints at ways to penetrate host immune defenses, a significant remaining challenge in modern virotherapy research.
Collapse
Affiliation(s)
- Elizabeth Kelly
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
6
|
Kim JH, Oh JY, Park BH, Lee DE, Kim JS, Park HE, Roh MS, Je JE, Yoon JH, Thorne SH, Kirn D, Hwang TH. Systemic armed oncolytic and immunologic therapy for cancer with JX-594, a targeted poxvirus expressing GM-CSF. Mol Ther 2006; 14:361-70. [PMID: 16905462 DOI: 10.1016/j.ymthe.2006.05.008] [Citation(s) in RCA: 240] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Revised: 05/16/2006] [Accepted: 05/16/2006] [Indexed: 12/17/2022] Open
Abstract
Targeted oncolytic viruses and immunostimulatory therapeutics are being developed as novel cancer treatment platforms. These approaches can be combined through the expression of immunostimulatory cytokines from targeted viruses, including adenoviruses and herpesviruses. Although intratumoral injection of such viruses has been associated with tumor growth inhibition, eradication of distant metastases was not reported. The major limitations for this approach to date have been (1) inefficient intravenous virus delivery to tumors and (2) the lack of predictive, immunocompetent preclinical models. To overcome these hurdles, we developed JX-594, a targeted, thymidine kinase(-) vaccinia virus expressing human GM-CSF (hGM-CSF), for intravenous (i.v.) delivery. We evaluated two immunocompetent liver tumor models: a rabbit model with reproducible, time-dependent metastases to the lungs and a carcinogen-induced rat liver cancer model. Intravenous JX-594 was well tolerated and had highly significant efficacy, including complete responses, against intrahepatic primary tumors in both models. In addition, whereas lung metastases developed in all control rabbits, none of the i.v. JX-594-treated rabbits developed detectable metastases. Tumor-specific virus replication and gene expression, systemically detectable levels of hGM-CSF, and tumor-infiltrating CTLs were also demonstrated. JX-594 holds promise as an i.v.-delivered, targeted virotherapeutic. These two tumor models hold promise for the optimization of this approach.
Collapse
Affiliation(s)
- J H Kim
- Department of Pharmacology and MRCCMT, Dong-A University Medical College, Busan 604-714, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Trajcevski S, Solly SK, Frisén C, Trenado A, Cosset FL, Klatzmann D. Characterization of a semi-replicative gene delivery system allowing propagation of complementary defective retroviral vectors. J Gene Med 2005; 7:276-87. [PMID: 15515136 DOI: 10.1002/jgm.663] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Recently, several cancer gene therapy studies have shown that replication-competent retroviral vectors represent a major improvement over replication-defective ones in terms of transgene propagation efficiency. However, this positive effect is somewhat spoiled by the increased risk of dissemination and oncogenesis that replication-competent retroviral vectors entail. To enhance both their integral safety and their transgene capacity, we developed a semi-replication-competent retroviral vector system. METHODS The semi-replication-competent retroviral vector system is based on two transcomplementing replication-defective retroviral vectors termed gag-pol vector (GPv) and env vector (Ev). Vector propagation was monitored in vitro and in solid tumors in vivo, using different reporter transgenes for GPv and Ev. Systemic vector dissemination and leukemogenesis was assessed by direct intravenous vector injection and subsequent bone marrow transplantation, in MLV-sensitive mice. RESULTS In vitro and in vivo the semi-replication-competent retroviral vectors propagate transgenes almost as efficiently as replication-competent ones. The semi-replication-competent retroviral vector system does not lead to detectable dissemination or leukemogenesis as does the replication-competent vector or the parental virus. Additionally, the vector duo allows co-propagation of different transgenes as well as mobilization of a third replication-defective vector. CONCLUSIONS This study is an initial proof of principle for the use of complementary retroviral vectors to deliver and propagate transgenes in vitro and in solid tumors in vivo, but with reduced pathogenicity compared to its parental virus. In-between replication-defective and replication-competent retroviral vectors, this semi-replicative system offers good grounds for its application in in vitro studies and allows envisioning its further development for cancer gene therapy.
Collapse
Affiliation(s)
- Stéphane Trajcevski
- Laboratoire de biologie et thérapeutiques des pathologies immunitaires, CNRS UMR7087, Université Pierre et Marie Curie, Groupe hospitalier Pitié-Salpêtrière, 83 boulevard de l'hôpital, 75651 Paris cedex 13, France
| | | | | | | | | | | |
Collapse
|
8
|
Abstract
Molecular research has vastly advanced our understanding of the mechanism of cancer growth and spread. Targeted approaches utilizing molecular science have yielded provocative results in the treatment of cancer. Oncolytic viruses genetically programmed to replicate within cancer cells and directly induce toxic effect via cell lysis or apoptosis are currently being explored in the clinic. Safety has been confirmed and despite variable efficacy results several dramatic responses have been observed with some oncolytic viruses. This review summarizes results of clinical trials with oncolytic viruses in cancer.
Collapse
Affiliation(s)
- Eugene Lin
- Mary Crowley Medical Research Center, Dallas, Texas, USA
| | | |
Collapse
|
9
|
Finger C, Sun Y, Sanz L, Alvarez-Vallina L, Buchholz CJ, Cichutek K. Replicating retroviral vectors mediating continuous production and secretion of therapeutic gene products from cancer cells. Cancer Gene Ther 2005; 12:464-74. [PMID: 15692609 DOI: 10.1038/sj.cgt.7700805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The successful application of cancer gene therapy has been hampered by the low efficiency of in vivo gene delivery by currently used replication-defective vectors. Accordingly, considerable efforts are now being directed toward development and use of vectors capable of replicating in cancer cells. However, for replicating retroviruses, insertion of additional reading frames into the viral genome often resulted in the generation of unstable viruses. Here, we report a novel concept for the generation of replication-competent murine leukemia virus (MLV) vectors capable of mediating the secretion of soluble therapeutic proteins from infected cells. As a proof of principle, we inserted transgene regions encoding either a single-chain variable region fragment (scFv), here, the laminin-specific L36-scFv, or the T-cell-specific 7A5-scFv, or the cytokine GM-CSF into the MLV envelope (env) gene after +1 codon of the envelope (Env) protein, followed by a sequence specifying a furin protease cleavage site. The resulting viruses, termed L36-furin-A, 7A5-furin-A and GMCSF-furin-Mo, respectively, infected a variety of human cell lines, including HMEC-1 (endothelial), A301 (lymphoid), MDA-MB231 and MDA-MB468 (breast cancer) and HT1080 (fibrosarcoma) cells. Western blot analysis of conditioned culture medium from HT1080 cells infected by replicating L36-furin A, as an example, revealed that more than 90% of the Env fusion protein molecules were indeed intracellularly cleaved. After 5 days of infection, up to 3-4 mug/ml of soluble L36-scFv accumulated in the supernatant of HT1080 cells. The eukaryotically produced L36-scFv and 7A5-scFv were able to recognize their native antigens with high avidity, as assessed by ELISA and flow cytometry. Furthermore, the replicating viruses were genetically stable for more than 12 cell passages. In conclusion, a new generation of replication-competent retroviral vectors capable of mediating long-term and efficient secretion of therapeutic proteins suitable for cancer therapy was generated.
Collapse
Affiliation(s)
- Carsten Finger
- Division of Medical Biotechnology, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | | | | | | | | | | |
Collapse
|
10
|
Abstract
Viruses capable of inducing lysis of malignant cells through their replication process are known as "oncolytic" viruses. Clinical trials in oncology have been performed with oncolytic viruses for nearly fifty years. Both systemic and intratumoral routes of administration have been explored. Toxicity has generally been limited to injection site pain, transient fever, and tumor necrosis. Responses with early crude materials were usually short in duration; however, recent trials with gene-attenuated viruses suggest a more prolonged duration of responses.
Collapse
|
11
|
Gómez-Treviño A, Castel S, López-Iglesias C, Cortadellas N, Comas-Riu J, Mercadé E. Effects of adenovirus-mediated SV5 fusogenic glycoprotein expression on tumor cells. J Gene Med 2003; 5:483-92. [PMID: 12797113 DOI: 10.1002/jgm.371] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The fusogenic (F) membrane glycoprotein of the paramyxovirus SV5 allows virus to enter host cells and mediates fusion between neighboring cells, which leads to cell death. F glycoprotein is synthesized as an inactive precursor (F(0)) that is cleaved by cellular protease furine to form the active heterodimer F(1) + F(2). The active protein can induce syncytium formation in the absence of another integral glycoprotein (HN), a property that appears to be unique among paramyxoviruses. METHODOLOGY We constructed a non-replicative adenovirus to express SV5 F protein in tumor cells, and its fusion capacity was analyzed by fluorescent and confocal microscopy. Cell viability and bystander effect were compared with the thymidine kinase/ganciclovir suicide gene therapy. The structure of F-expressing cells was studied using electron microscopy. RESULTS F glycoprotein expression induced syncytium formation to a maximum at 72 h, after which syncytia progressively lost viability and detached. The cell membrane was disrupted while nuclear structure was preserved. Over-expression of SV5 F protein in tumor cells led to high cytotoxicity comparable with that associated with the thymidine kinase/ganciclovir. A potent bystander killing effect was detected until the ratio of F-transduced to non-transduced cells was 1 : 100. CONCLUSIONS These results indicate that the fusogenic glycoprotein of the paramyxovirus SV5 could be used to eliminate tumor cells and may encourage studies aimed at modifying its selectivity and combining its expression with other cytotoxic strategies to improve their efficacy.
Collapse
Affiliation(s)
- A Gómez-Treviño
- Department of Microbiology, University of Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
12
|
Solly SK, Trajcevski S, Frisén C, Holzer GW, Nelson E, Clerc B, Abordo-Adesida E, Castro M, Lowenstein P, Klatzmann D. Replicative retroviral vectors for cancer gene therapy. Cancer Gene Ther 2003; 10:30-9. [PMID: 12489026 DOI: 10.1038/sj.cgt.7700521] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2002] [Indexed: 11/08/2022]
Abstract
Poor efficiency of gene transfer into cancer cells constitutes the major bottleneck of current cancer gene therapy. We reasoned that because tumors are masses of rapidly dividing cells, they would be most efficiently transduced with vector systems allowing transgene propagation. We thus designed two replicative retrovirus-derived vector systems: one inherently replicative vector, and one defective vector propagated by a helper retrovirus. In vitro, both systems achieved very efficient transgene propagation. In immunocompetent mice, replicative vectors transduced >85% tumor cells, whereas defective vectors transduced <1% under similar conditions. It is noteworthy that viral propagation could be efficiently blocked by azido-thymidine, in vitro and in vivo. In a model of established brain tumors treated with suicide genes, replicative retroviral vectors (RRVs) were approximately 1000 times more efficient than defective adenoviral vectors. These results demonstrate the advantage and potential of RRVs and strongly support their development for cancer gene therapy.
Collapse
Affiliation(s)
- Sounkary K Solly
- Biologie et Thérapeutique des Pathologies Immunitaires, CNRS/UPMC UMR 7087, Hôpital Pitié-Salpêtrière, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Logg CR, Logg A, Matusik RJ, Bochner BH, Kasahara N. Tissue-specific transcriptional targeting of a replication-competent retroviral vector. J Virol 2002; 76:12783-91. [PMID: 12438603 PMCID: PMC136666 DOI: 10.1128/jvi.76.24.12783-12791.2002] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2002] [Accepted: 09/13/2002] [Indexed: 01/06/2023] Open
Abstract
The inability of replication-defective viral vectors to efficiently transduce tumor cells in vivo has prevented the successful application of such vectors in gene therapy of cancer. To address the need for more efficient gene delivery systems, we have developed replication-competent retroviral (RCR) vectors based on murine leukemia virus (MLV). We have previously shown that such vectors are capable of transducing solid tumors in vivo with very high efficiency. While the natural requirement of MLV infection for cell division imparts a certain degree of specificity for tumor cells, additional means for confining RCR vector replication to tumor cells are desirable. Here, we investigated the parameters critical for successful tissue-specific transcriptional control of RCR vector replication by replacing various lengths of the MLV enhancer/promoter with sequences derived either from the highly prostate-specific probasin (PB) promoter or from a more potent synthetic variant of the PB promoter. We assessed the transcriptional specificity of the resulting hybrid long terminal repeats (LTRs) and the cell type specificity and efficiency of replication of vectors containing these LTRs. Incorporation of PB promoter sequences effectively restricted transcription from the LTR to prostate-derived cells and imparted prostate-specific RCR vector replication but required the stronger synthetic promoter and retention of native MLV sequences in the vicinity of the TATA box for optimal replicative efficiency and specificity. Our results have thus identified promoter strength and positioning within the LTR as important determinants for achieving both high transduction efficiency and strict cell type specificity in transcriptionally targeted RCR vectors.
Collapse
Affiliation(s)
- Christopher R Logg
- Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, USA
| | | | | | | | | |
Collapse
|
14
|
Abstract
RNA viruses are rapidly emerging as extraordinarily promising agents for oncolytic virotherapy. Integral to the lifecycles of all RNA viruses is the formation of double-stranded RNA, which activates a spectrum of cellular defense mechanisms including the activation of PKR and the release of interferon. Tumors are frequently defective in their PKR signaling and interferon response pathways, and therefore provide a relatively permissive substrate for the propagation of RNA viruses. For most of the oncolytic RNA viruses currently under study, tumor specificity is either a natural characteristic of the virus, or a serendipitous consequence of adapting the virus to propagate in human tumor cell lines. Further refinement and optimization of these oncolytic agents can be achieved through virus engineering. This article provides a summary of the current status of oncolytic virotherapy efforts for seven different RNA viruses, namely, mumps, Newcastle disease virus, measles virus, vesicular stomatitis virus, influenza, reovirus, and poliovirus.
Collapse
Affiliation(s)
- Stephen J Russell
- Molecular Medicine Program, Mayo Clinic, Rochester, Minnesota 55905, USA.
| |
Collapse
|
15
|
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
|
16
|
Advani SJ, Weichselbaum RR, Whitley RJ, Roizman B. Friendly fire: redirecting herpes simplex virus-1 for therapeutic applications. Clin Microbiol Infect 2002; 8:551-63. [PMID: 12427216 DOI: 10.1046/j.1469-0691.2002.00432.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Herpes simplex virus-1 (HSV-1) is a relatively large double-stranded DNA virus encoding at least 89 proteins with well characterized disease pathology. An understanding of the functions of viral proteins together with the ability to genetically engineer specific viral mutants has led to the development of attenuated HSV-1 for gene therapy. This review highlights the progress in creating attenuated genetically engineered HSV-1 mutants that are either replication competent (viral non-essential gene deleted) or replication defective (viral essential gene deleted). The choice between a replication-competent or -defective virus is based on the end-goal of the therapeutic intervention. Replication-competent HSV-1 mutants have primarily been employed as antitumor oncolytic viruses, with the lytic nature of the virus harnessed to destroy tumor cells selectively. In replacement gene therapy, replication-defective viruses have been utilized as delivery vectors. The advantages of HSV-1 vectors are that they infect quiescent and dividing cells efficiently and can encode for relatively large transgenes.
Collapse
Affiliation(s)
- S J Advani
- The Marjorie B. Kovler Viral Oncology Laboratories, The University of Chicago, Chicago, Illinois 60637, USA.
| | | | | | | |
Collapse
|
17
|
Harrington K, Alvarez-Vallina L, Crittenden M, Gough M, Chong H, Diaz RM, Vassaux G, Lemoine N, Vile R. Cells as vehicles for cancer gene therapy: the missing link between targeted vectors and systemic delivery? Hum Gene Ther 2002; 13:1263-80. [PMID: 12162810 DOI: 10.1089/104303402760128504] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Systemic administration of currently manufactured viral stocks has not so far achieved sufficient circulating titers to allow therapeutic targeting of metastatic disease. This is due to low initial viral titers, immune inactivation, nonspecific adhesion, and loss of particles. One way to exploit the elegant molecular manipulations that have been made to increase vector targeting is to protect these vectors until they reach the local sites of tumor growth. Various cell types home preferentially to tumors and can be loaded with the constructs required to produce targeted vectors. Here we discuss the potential of using such cell carriers to chaperone precious vectors directly to the tumors. The vectors can incorporate mechanisms to achieve tumor site-inducible expression, along with tumor cell-specific expression of the therapeutic gene and/or replicating viral genomes that would be released at the tumor. In this way, the great advances that have so far been made with the engineering of vector tropisms might be genuinely exploited and converted into clinical benefit.
Collapse
Affiliation(s)
- Kevin Harrington
- Cancer Research Campaign, Centre for Cell and Molecular Biology, Chester Beatty Laboratories, Institute of Cancer Research, London SW3 6JB, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Blank SV, Rubin SC, Coukos G, Amin KM, Albelda SM, Molnar-Kimber KL. Replication-selective herpes simplex virus type 1 mutant therapy of cervical cancer is enhanced by low-dose radiation. Hum Gene Ther 2002; 13:627-39. [PMID: 11916486 DOI: 10.1089/10430340252837224] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1)-based oncolytic treatment is a promising therapeutic approach for malignancy. Recombinant strains of HSV-1 containing mutations in the ICP 34.5 protein have been shown to replicate preferentially in rapidly proliferating malignant cells, resulting in a direct cytolytic effect. We assessed the efficacy of multimutated HSV-1 strains on human cervical cancer, and then used these viruses in combination with radiation therapy, the standard treatment for cervical cancer. The HSV-1 mutants 4009, 7020, 3616, and G207 induced significant lysis of three established human cervical cancer cell lines in vitro in a dose-dependent manner. G207 intratumoral treatment of established subcutaneous C33a tumors in severe combined immunodeficient (SCID) mice significantly reduced tumor burden by 50%. Weekly and triweekly treatments improved efficacy and inhibited flank tumor growth in an administration frequency-dependent manner without toxicity. Combination therapy of a low dose of radiation (1.5 or 3 Gy) and replication-selective HSV mutants infection exhibited increased antitumor effects against cervical cancer cells in vitro. The in vivo effect of G207 combined with low-dose radiation was studied in Me180 xenografts in athymic mice. Treatment of established Me180 tumor nodules with 3 Gy followed by intratumoral G207 administration greatly improved efficacy, resulting in 42% complete eradication of tumor. In conclusion, single and multiple intratumoral injections of G207 significantly reduced tumor burden in xenogeneic models of cervical cancer, and the addition of low-dose radiation further potentiated the effect. These results suggest that replication-selective HSV-1 mutants may be potent oncolytic agents for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Stephanie V Blank
- Department of Obstetrics and Gynecology, University of Pennsylvania Medical Center, Philadelphia, PA 19104, USA.
| | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Galanis E, Vile R, Russell SJ. Delivery systems intended for in vivo gene therapy of cancer: targeting and replication competent viral vectors. Crit Rev Oncol Hematol 2001; 38:177-92. [PMID: 11369253 DOI: 10.1016/s1040-8428(01)00103-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cancer gene therapy represents one of the most rapidly evolving areas in pre-clinical and clinical cancer research. Application of gene transfer techniques in clinical trials has made increasingly obvious that several issues will need to be addressed prior to meaningful incorporation of gene therapy in the care of cancer patients. Two of the most important problems to overcome are lack of selectivity of the existing vectors and low efficiency of gene transfer. This review focuses on use of targeting and replication competent vectors in order to overcome these obstacles. Targeted gene therapy of malignancies can be achieved through vector targeting or transcriptional targeting and can improve the therapeutic index of gene transfer by preventing damage of normal tissues, an important requirement if systemic gene delivery is contemplated. Replication competent viral vectors can improve the efficiency of gene transfer. Provisionally replicating viruses can also improve the therapeutic index by targeting toxicity to tumor cells. A variety of provisionally replicating viruses, such as the attenuated adenovirus ONYX-015, the adenovirus CN706 that selectively replicates in prostate cancer cells, the double mutant herpes simplex virus G207, the human reovirus, and the Newcastle disease virus are currently in clinical trials. Early clinical results and limitations in the application of these vectors are discussed.
Collapse
Affiliation(s)
- E Galanis
- Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
21
|
Lambright ES, Amin K, Wiewrodt R, Force SD, Lanuti M, Propert KJ, Litzky L, Kaiser LR, Albelda SM. Inclusion of the herpes simplex thymidine kinase gene in a replicating adenovirus does not augment antitumor efficacy. Gene Ther 2001; 8:946-53. [PMID: 11426335 DOI: 10.1038/sj.gt.3301489] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2000] [Accepted: 04/10/2001] [Indexed: 11/09/2022]
Abstract
Replication-incompetent adenoviruses (Ad) carrying the herpes simplex thymidine kinase (HSVtk) gene have been used in a number of human cancer gene therapy trials, however transduction has generally been limited to a small minority of tumor cells. To solve this problem, replication-competent adenoviral vectors carrying transgenes such as HSVtk have been developed. However, contradictory evidence exists regarding the efficacy of these new vectors. Accordingly, we constructed and tested a replication-competent E3-deleted adenoviral vector containing the HSVtk suicide gene driven by the endogenous E3 promoter (Ad.wt.tk). This virus showed high level production of the HSVtk transgene and was more efficacious than a non-replicating virus in vitro, after injection into flank tumors, and against established intraperitoneal tumors. However, addition of ganciclovir (GCV) therapy to cells or tumor-bearing animals treated with the replicating vector containing the HSVtk suicide gene did not result in increased cell killing. Our results indicate that addition of HSVtk to a replicating Ad virus will not likely be useful in augmenting antitumor effects.
Collapse
Affiliation(s)
- E S Lambright
- Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Logg CR, Tai CK, Logg A, Anderson WF, Kasahara N. A uniquely stable replication-competent retrovirus vector achieves efficient gene delivery in vitro and in solid tumors. Hum Gene Ther 2001; 12:921-32. [PMID: 11387057 PMCID: PMC8184367 DOI: 10.1089/104303401750195881] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A major obstacle in cancer gene therapy is the limited efficiency of in vivo gene transfer by replication-defective retrovirus vectors in current use. One strategy for circumventing this difficulty would be to use vectors capable of replication within tumor tissues. We have developed a replication-competent retrovirus (RCR) vector derived from murine leukemia virus (MuLV). This vector utilizes a unique design strategy in which an internal ribosome entry site-transgene cassette is positioned between the env gene and the 3' long terminal repeat (LTR). The ability of this vector to replicate and transmit a transgene was examined in culture and in a solid tumor model in vivo. The RCR vector exhibited replication kinetics similar to those of wildtype MuLV and mediated efficient delivery of the transgene throughout an entire population of cells in culture after an initial inoculation with 1 plaque-forming unit (PFU) of vector per 2000 cells. After injection of 6 x 10(3) PFU of vector into established subcutaneous tumors, highly efficient spread of the transgene was observed over a period of 7 weeks, in some cases resulting in spread of the transgene throughout the entire tumor. MuLV-based RCR vectors show significant advantages over standard replication-defective vectors in efficiency of gene delivery both in culture and in vivo. This represents the first example of the use of an RCR vector in an adult mammalian host, and their first application to transduction of solid tumors.
Collapse
Affiliation(s)
- C R Logg
- Department of Pathology and Institute for Genetic Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | | | | | | | | |
Collapse
|
23
|
Abstract
High grade gliomas in adults are devastating diseases, with very poor survival despite their lack of distant metastases. Local treatments, such as surgical resection and stereotactic radiosurgery, have been most successful, whereas systemic therapy (for example, chemotherapy and immunotherapy) have been rather disappointing. Several gene therapy systems have been successful in controlling or eradicating these tumours in animal models and are now being tested as a logical addition to current clinical management. This review describes the gene therapy clinical protocols that have been completed or that are ongoing for human gliomas. These include the prodrug activating system, herpes simplex thymidine kinase (HSVtk)/ganciclovir (GCV), utilising either retrovirus vector producer cells or adenovirus vectors; adenovirus mediated p53 gene transfer; adenovirus mediated IFN-beta gene transfer and oncolytic herpes virus and adenovirus vectors. To date, all of the clinical studies have used direct injection of the vector into the glioma. The Phase I clinical studies have demonstrated low to moderate toxicity and variable levels of gene transfer and in some cases anti-tumour effect. Future directions will rely upon improvements in gene delivery as well as gene therapies and combinations of gene therapy with other treatment modalities.
Collapse
Affiliation(s)
- J B Alavi
- Hematology-Oncology Division, Department of Medicine, University of Pennsylvania, 3400 Spruce St., Philadelphia 19104, USA.
| | | |
Collapse
|
24
|
Abstract
Viruses capable of inducing lysis of malignant cells through their replication process are known as "oncolytic" viruses. Clinical trials in oncology have been performed with oncolytic viruses for nearly fifty years. Both systemic and intratumoral routes of administration have been explored. Toxicity has generally been limited to injection site pain, transient fever and tumor necrosis. Responses with early crude materials were usually short in duration; however, recent trials with gene attenuated viruses suggest more prolonged duration to responses observed.
Collapse
|
25
|
Abstract
Gene therapy, the treatment of any disorder or pathophysiologic state on the basis of the transfer of genetic information, was a high-priority goal in the 1990s. The lung is a major target of gene therapy for genetic disorders, such as cystic fibrosis and alpha1-antitrypsin deficiency, and for other diseases, including lung cancer, malignant mesothelioma, pulmonary inflammation, surfactant deficiency, and pulmonary hypertension. This paper examines general concepts in gene therapy, summarizes the results of published clinical trials, and highlights areas of research aimed at overcoming challenges in the field. Although progress has been slower than anticipated, gene transfer has been safely achieved in patients with lung diseases. Recent advancements in understanding of the molecular basis of lung disease and the development of improved vector systems make it likely that gene therapy will be an important tool for the 21st-century clinician.
Collapse
Affiliation(s)
- S M Albelda
- University of Pennsylvania Medical Center, Philadelphia, USA.
| | | | | |
Collapse
|
26
|
Affiliation(s)
- J Gómez-Navarro
- Gene Therapy Center, University of Alabama, Birmingham, Alabama 35294-3300, USA
| | | | | |
Collapse
|
27
|
Lambright ES, Caparrelli DJ, Abbas AE, Toyoizumi T, Coukos G, Molnar-Kimber KL, Kaiser LR. Oncolytic therapy using a mutant type-1 herpes simplex virus and the role of the immune system. Ann Thorac Surg 1999; 68:1756-60; discussion 1761-2. [PMID: 10585055 DOI: 10.1016/s0003-4975(99)00852-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Herpes simplex virus (HSV)-1716, a replication-restricted herpes simplex virus type 1, has shown efficacy as an oncolytic treatment for central nervous system tumors, breast cancer, ovarian cancer, and malignant mesothelioma. We evaluated the efficacy of HSV-1716 in a murine lung cancer model, Lewis lung carcinoma. METHODS Lewis lung carcinoma cells were infected with HSV-1716 and implanted in the flanks of mice at varying ratios of infected to uninfected cells. Tumor burden was assessed by measurement of the weight of the tumor nodule. The role of the immune system was examined by performing experiments in both immunocompetent and SCID mice. Tumors were implanted in the opposite flank to evaluate the vaccine effect. RESULTS In immunocompetent and SCID animals, ratio of 1:10 (infected-to-uninfected) cells completely prevented tumor formation and ratio of 1:100 suppressed tumor growth. Established tumors at a distant site in the groups receiving HSV-1716 infected cells showed no difference in size versus control, suggesting absence of a vaccine effect. CONCLUSIONS We conclude that HSV-1716 may provide a oncolytic therapy for lung cancer even in the absence of immune system induction and a "carrier" cell could potentially deliver this vector.
Collapse
Affiliation(s)
- E S Lambright
- Harrison Department of Surgical Research, University of Pennsylvania Medical Center, Philadelphia, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Malignant pleural mesothelioma is a neoplasm that is commonly fatal and for which there are no widely accepted curative approaches. Mesothelioma is unresponsive to most chemotherapy and radiotherapy regimens, and it typically recurs even after the most aggressive attempts at surgical resection. Multimodality approaches have been of some benefit in prolonging survival of very highly selected subgroups of patients, but they have had a relatively small impact on the majority of the patients diagnosed with this disease. As the incidence of pleural mesothelioma peaks in the United States and Europe over the next 10 to 20 years, new therapeutic measures will be necessary. This review will discuss the roles of chemotherapy, radiotherapy, surgery, and combined modality approaches in the treatment of pleural mesothelioma, as well as scientific advances made in the past decade that have led to the development of experimental techniques, such as photodynamic therapy, immunotherapy, and gene therapy, that are currently undergoing human clinical trials. These promising new avenues may modify the therapeutic nihilism that is rampant among clinicians dealing with mesothelioma.
Collapse
Affiliation(s)
- D H Sterman
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania Medical Center, Philadelphia 19104, USA.
| | | | | |
Collapse
|
29
|
Hunter WD, Martuza RL, Feigenbaum F, Todo T, Mineta T, Yazaki T, Toda M, Newsome JT, Platenberg RC, Manz HJ, Rabkin SD. Attenuated, replication-competent herpes simplex virus type 1 mutant G207: safety evaluation of intracerebral injection in nonhuman primates. J Virol 1999; 73:6319-26. [PMID: 10400723 PMCID: PMC112710 DOI: 10.1128/jvi.73.8.6319-6326.1999] [Citation(s) in RCA: 142] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/1999] [Accepted: 04/16/1999] [Indexed: 11/20/2022] Open
Abstract
This study examined the safety of intracerebral inoculation of G207, an attenuated, replication-competent herpes simplex virus type 1 (HSV-1) recombinant, in nonhuman primates. Sixteen New World owl monkeys (Aotus nancymae [karyotype 1, formerly believed to be A. trivirgatus]), known for their exquisite susceptibility to HSV-1 infection, were evaluated. Thirteen underwent intracerebral inoculation with G207 at doses of 10(7) or 10(9) PFU, two were vehicle inoculated, and one served as an infected wild-type control and received 10(3) PFU of HSV-1 strain F. HSV-1 strain F caused rapid mortality and symptoms consistent with HSV encephalitis, including fever, hemiparesis, meningitis, and hemorrhage in the basal ganglia. One year after G207 inoculation, seven of the animals were alive and exhibited no evidence of clinical complications. Three deaths resulted from nonneurologic causes unrelated to HSV infection, and three animals were sacrificed for histopathologic examination. Two animals were reinoculated with G207 (10(7) PFU) at the same stereotactic coordinates 1 year after the initial G207 inoculation. These animals were alive and healthy 2 years after the second inoculation. Cerebral magnetic resonance imaging studies performed both before and after G207 inoculation failed to reveal radiographic evidence of HSV-related sequelae. Despite the lack of outwardly observable HSV pathology, measurable increases in serum anti-HSV titers were detected. Histopathological examination of multiple organ tissues found no evidence of HSV-induced histopathology or dissemination. We conclude that intracerebral inoculation of up to 10(9) PFU of G207, well above the efficacious dose in mouse tumor studies, is safe and therefore appropriate for human clinical trials.
Collapse
Affiliation(s)
- W D Hunter
- Departments of Neurosurgery, Georgetown University Medical Center, Washington, D.C. 20007, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Affiliation(s)
- J Gómez-Navarro
- Gene Therapy Center, University of Alabama at Birmingham 35294-3300, USA
| | | | | |
Collapse
|
31
|
Efficient Generation of Recombinant Adenoviral Vectors by Cre-lox Recombination In Vitro. Mol Med 1999. [DOI: 10.1007/bf03402119] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
32
|
|
33
|
Abstract
Gene therapy has received considerable attention and some speculation as to its value. Although few patients have been treated, the preliminary results of the phase I lung cancer gene therapy clinical trials are very promising. Clinically relevant basic research in the molecular pathogenesis and immunology of lung cancer is progressing. As improved vector technologies are developed, new opportunities will be available to initiate lung cancer gene therapy trials that are based on a more detailed understanding of lung cancer biology. In conclusion, although important biologic and technical questions remain unanswered, recent research suggests that gene therapy will have a profound impact on lung cancer treatment.
Collapse
Affiliation(s)
- S M Dubinett
- University of California at Los Angeles/Wadsworth Pulmonary Immunology Laboratory, Division of Pulmonary and Critical Care Medicine, USA
| | | | | | | |
Collapse
|
34
|
Abstract
Gene therapy for breast cancer is still in the very early stages of development. Many of the molecular strategies that have been proposed are also being developed for other cancers. Their application to breast cancer, however, needs to address several issues specific to this disease such as the widespread nature of metastases, the indolent growth of the tumor cells, and the production by the tumor of immunosuppressive agents. Nonetheless, these approaches appear promising, particularly those that employ a combination of strategies. Gene therapies that affect the biology of breast cancer cells or regulate host immune mechanisms have been most successful and may be paired with existing therapies for breast cancer.
Collapse
Affiliation(s)
- H K Boxhorn
- Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, USA
| | | |
Collapse
|
35
|
Abstract
Malignant mesothelioma (MM) is a fatal malignancy refractory to all forms of standard anticancer therapy. This article reports the results of a phase I clinical trial assessing the safety of intrapleural delivery and efficacy of intratumoral gene transfer of recombinant adenovirus (rAd) containing herpes simplex virus thymidine kinase (HSVtk) gene into the pleural space of patients with MM, followed by systematic treatment with the antiviral drug ganciclovir (GCV) for 14 days. AD.RSVtk/GCV gene therapy proved to be well tolerated, with evidence of significant gene transfer particularly at high vector doses and with elimination of preliminary biopsy. Ongoing gene therapy trials for mesothelioma at two other centers, focusing on immunostimulation and using suicide gene therapy as a tumor vaccine, are also reviewed in this article.
Collapse
Affiliation(s)
- D H Sterman
- Department of Medicine, University of Pennsylvania Health System, Philadelphia, USA
| | | | | |
Collapse
|
36
|
Vile RG, Sunassee K, Diaz RM. Strategies for achieving multiple layers of selectivity in gene therapy. MOLECULAR MEDICINE TODAY 1998; 4:84-92. [PMID: 9547795 DOI: 10.1016/s1357-4310(97)01157-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Here we review the progress towards the development of targeted vectors for direct in vivo delivery in gene therapy. Currently, there are many separate approaches. These include: simple physical/anatomical localization of administration of the vector at the site where gene transfer is required; exploitation of natural tropisms of plasmid, viral and cellular vectors; and the use of molecular engineering to change the specificity of proteins and nucleic acids so that they specifically recognize target ligands expressed on/in the target cells. Unfortunately, each of these approaches is usually imperfect by itself. However, combinations of these strategies might produce vectors in which several layers of imperfect targeting give an overall level of specificity that can justify systemic delivery of vectors to treat human disease.
Collapse
Affiliation(s)
- R G Vile
- Laboratory of Molecular Therapy, Hammersmith Hospital, London, UK.
| | | | | |
Collapse
|
37
|
Affiliation(s)
- R G Vile
- Imperial Cancer Research Fund, Laboratory of Molecular Therapy, ICRF Oncology Unit, Imperial College School of Medicine, Hammersmith Hospital, DuCane Road, London, W12 0NN, UK.
| |
Collapse
|
38
|
Niculescu-Duvaz I, Spooner R, Marais R, Springer CJ. Gene-directed enzyme prodrug therapy. Bioconjug Chem 1998; 9:4-22. [PMID: 9460542 DOI: 10.1021/bc970116t] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- I Niculescu-Duvaz
- CRC Centre for Cancer Therapeutics, Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | | | | | | |
Collapse
|
39
|
Abstract
Adenoviruses began to be developed into highly effective gene expression vectors in the early 1980s. Recently, the increased interest in utilizing this transfer system in vivo has posed new problems for heterologous gene-transfer, spurring a renewed effort in the field of vector development toward solving the structural, immunological and targeting problems posed by gene therapy applications.
Collapse
Affiliation(s)
- I Kovesdi
- GenVec Inc., Rockville, MD 20852, USA.
| | | | | | | |
Collapse
|
40
|
Abstract
Most of these therapies, although still in the infant stages of their development, offer the potential for major advances in colorectal cancer therapy. Gene therapy is an entirely new medicinal paradigm for the treatment of cancer. Currently, the clinical application of these methods is limited by the need for a more through understanding of cancer immunology and the availability of better vector systems for efficient and selective tumor gene transfer. As increasing numbers of scientists and clinicians address these issues, better therapies will likely emerge.
Collapse
Affiliation(s)
- S L Eck
- Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, USA
| |
Collapse
|
41
|
Affiliation(s)
- S M Albelda
- Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, USA.
| |
Collapse
|
42
|
Affiliation(s)
- May S.M. Smith
- Bureau of Biologies and Radiopharmaceuticals, Drugs Directorate, Health Protection Branch, Health Canada
| | - Anthony A.G. Ridgway
- Bureau of Biologies and Radiopharmaceuticals, Drugs Directorate, Health Protection Branch, Health Canada
| |
Collapse
|
43
|
Irie A, Kijima H, Ohkawa T, Bouffard DY, Suzuki T, Curcio LD, Holm PS, Sassani A, Scanlon KJ. Anti-oncogene ribozymes for cancer gene therapy. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1997; 40:207-57. [PMID: 9217927 DOI: 10.1016/s1054-3589(08)60141-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- A Irie
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California 91010, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Many viral and non-viral vector systems have now been developed for gene therapy applications. In this article, the pros and cons of these vector systems are discussed in relation to the different cancer gene therapy strategies. The protocols used in cancer gene therapy can be broadly divided into six categories including gene transfer to explanted cells for use as cell-based cancer vaccines; gene transfer to a small number of tumour cells in situ to achieve a vaccine effect; gene transfer to vascular endothelial cells (VECs) lining the blood vessels of the tumour to interfere with tumour angiogenesis; gene transfer to T lymphocytes to enhance their antitumour effector capability; gene transfer to haemopoietic stem cells (HSCs) to enhance their resistance to cytotoxic drugs and gene transfer to a large number of tumour cells in situ to achieve nonimmune tumour reduction with or without bystander effect. Each of the six strategies makes unique demands on the vector system and these are discussed with reference to currently available vectors. Aspects of vector biology that are in need of further development are discussed in some detail. The final section points to the potential use of replicating viruses as delivery vehicles for efficient in vivo gene transfer to disseminated cancers.
Collapse
Affiliation(s)
- J Zhang
- Cambridge Centre for Protein Engineering, MRC Centre, UK
| | | |
Collapse
|
45
|
Chong H, Vile RG. New therapeutic approaches based on gene transfer techniques. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1996; 18:149-70. [PMID: 8908697 DOI: 10.1007/bf00820663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- H Chong
- Imperial Cancer Research Fund Laboratory of Cancer Gene Therapy, Rayne Institute, St. Thomas' Hospital, London, UK
| | | |
Collapse
|