全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Retargeting of Viruses to Generate Oncolytic Agents

DOI: 10.1155/2012/798526

Full-Text   Cite this paper   Add to My Lib

Abstract:

Oncolytic virus therapy is based on the ability of viruses to effectively infect and kill tumor cells without destroying the normal tissues. While some viruses seem to have a natural preference for tumor cells, most viruses require the modification of their tropism to specifically enter and replicate in such cells. This review aims to describe the transductional targeting strategies currently employed to specifically redirect viruses towards surface receptors on tumor cells. Three major strategies can be distinguished; they involve (i) the incorporation of new targeting specificity into a viral surface protein, (ii) the incorporation of a scaffold into a viral surface protein to allow the attachment of targeting moieties, and (iii) the use of bispecific adapters to mediate targeting of a virus to a specified moiety on a tumor cell. Of each strategy key features, advantages and limitations are discussed and examples are given. Because of their potential to cause sustained, multiround infection—a desirable characteristic for eradicating tumors—particular attention is given to viruses engineered to become self-targeted by the genomic expression of a bispecific adapter protein. 1. Introduction Cancer is one of the major health problems of our times. Though the prognosis for people diagnosed with, at least some forms of, cancer has increased considerably, it is more typical a disease of which treatment is initially effective, to be followed later by an irreversible and eventually fatal relapse. Already for decades, cancer treatment is based on three types of approaches: surgery, radio-, and chemotherapy. While the scientific and technological advancements have improved the efficacy of each of these classical approaches tremendously, and while also some new therapies have evolved including immunotherapy, the treatments apparently fail to eradicate all residual tumor cells or metastases completely. Therefore, additional means are urgently required to support or replace the conventional therapies. Hence, a variety of new approaches is currently being explored, one of which is based on the use of viruses. Oncolytic viruses are defined by their ability to specifically kill tumor cells, but to leave the normal tissues unharmed. Their most characteristic features, thus, are their target specificity and their cytolytic capacity. Ideally, they exhibit additional features including, but not limited to, a high reproductive capacity in vivo, the ability to recruit uninfected neighboring cells (syncytia formation), the ability to infect both dividing and nondividing

References

[1]  P. J. Lech and S. J. Russell, “Use of attenuated paramyxoviruses for cancer therapy,” Expert Review of Vaccines, vol. 9, no. 11, pp. 1275–1302, 2010.
[2]  G. N. Barber, “Vesicular stomatitis virus as an oncolytic vector,” Viral Immunology, vol. 17, no. 4, pp. 516–527, 2004.
[3]  C. Thirukkumaran and D. G. Morris, “Oncolytic viral therapy using reovirus,” Methods in Molecular Biology, vol. 542, pp. 607–634, 2009.
[4]  A. Hesse, D. Kosmides, R. E. Kontermann, and D. M. Nettelbeck, “Tropism modification of adenovirus vectors by peptide ligand insertion into various positions of the adenovirus serotype 41 short-fiber knob domain,” Journal of Virology, vol. 81, no. 6, pp. 2688–2699, 2007.
[5]  D. T. Rein, M. Breidenbach, H. Wu et al., “Gene transfer to cervical cancer with fiber-modified adenoviruses,” International Journal of Cancer, vol. 111, no. 5, pp. 698–704, 2004.
[6]  N. Belousova, G. Mikheeva, J. Gelovani, and V. Krasnykh, “Modification of adenovirus capsid with a designed protein ligand yields a gene vector targeted to a major molecular marker of cancer,” Journal of Virology, vol. 82, no. 2, pp. 630–637, 2008.
[7]  K. L. Poulin, R. M. Lanthier, A. C. Smith et al., “Retargeting of adenovirus vectors through genetic fusion of a single-chain or single-domain antibody to capsid protein IX,” Journal of Virology, vol. 84, no. 19, pp. 10074–10086, 2010.
[8]  R. Waehler, S. J. Russell, and D. T. Curiel, “Engineering targeted viral vectors for gene therapy,” Nature Reviews Genetics, vol. 8, no. 8, pp. 573–587, 2007.
[9]  J. M. Mathis, M. A. Stoff-Khalili, and D. T. Curiel, “Oncolytic adenoviruses—selective retargeting to tumor cells,” Oncogene, vol. 24, no. 52, pp. 7775–7791, 2005.
[10]  J. N. Glasgow, M. Everts, and D. T. Curiel, “Transductional targeting of adenovirus vectors for gene therapy,” Cancer Gene Therapy, vol. 13, no. 9, pp. 830–844, 2006.
[11]  K. Hall, M. E. Blair Zajdel, and G. E. Blair, “Unity and diversity in the human adenoviruses: exploiting alternative entry pathways for gene therapy,” Biochemical Journal, vol. 431, no. 3, pp. 321–336, 2010.
[12]  T. Nakamura and S. J. Russell, “Oncolytic measles viruses for cancer therapy,” Expert Opinion on Biological Therapy, vol. 4, no. 10, pp. 1685–1692, 2004.
[13]  A. L. Hammond, R. K. Plemper, J. Zhang, U. Schneider, S. J. Russell, and R. Cattaneo, “Single-chain antibody displayed on a recombinant measles virus confers entry through the tumor-associated carcinoembryonic antigen,” Journal of Virology, vol. 75, no. 5, pp. 2087–2096, 2001.
[14]  E. Galanis, “Therapeutic potential of oncolytic measles virus: promises and challenges,” Clinical Pharmacology and Therapeutics, vol. 88, no. 5, pp. 620–625, 2010.
[15]  B. Blechacz and S. J. Russell, “Measles virus as an oncolytic vector platform,” Current Gene Therapy, vol. 8, no. 3, pp. 162–175, 2008.
[16]  S. J. Russell and K. W. Peng, “Measles virus for cancer therapy,” Current Topics in Microbiology and Immunology, vol. 330, pp. 213–241, 2009.
[17]  K. Shah and X. O. Breakefield, “HSV amplicon vectors for cancer therapy,” Current Gene Therapy, vol. 6, no. 3, pp. 361–370, 2006.
[18]  G. Campadelli-Fiume, C. De Giovanni, V. Gatta, P. Nanni, P. L. Lollini, and L. Menotti, “Rethinking herpes simplex virus: the way to oncolytic agents,” Reviews in Medical Virology, vol. 21, no. 4, pp. 213–226, 2011.
[19]  R. Manservigi, R. Argnani, and P. Marconi, “HSV recombinant vectors for gene therapy,” The Open Virology Journal, vol. 4, pp. 123–156, 2010.
[20]  S. J. Turrell and A. Whitehouse, “Mutation of herpesvirus saimiri ORF51 glycoprotein specifically targets infectivity to hepatocellular carcinoma cell lines,” Journal of Biomedicine and Biotechnology, vol. 2011, Article ID 785158, 14 pages, 2011.
[21]  K. Park, W. J. Kim, Y. H. Cho et al., “Cancer gene therapy using adeno-associated virus vectors,” Frontiers in Bioscience, vol. 13, no. 7, pp. 2653–2659, 2008.
[22]  A. Girod, M. Ried, C. Wobus et al., “Genetic capsid modifications allow efficient re-targeting of adeno-associated virus type 2,” Nature Medicine, vol. 5, no. 9, pp. 1052–1056, 1999.
[23]  M. Grifman, M. Trepel, P. Speece et al., “Incorporation of tumor-targeting peptides into recombinant adeno-associated virus capsids,” Molecular Therapy, vol. 3, no. 6, pp. 964–975, 2001.
[24]  W. Shi, G. S. Arnold, and J. S. Bartlett, “Insertional mutagenesis of the adeno-associated virus type 2 (AAV2) capsid gene and generation of AAV2 vectors targeted to alternative cell-surface receptors,” Human Gene Therapy, vol. 12, no. 14, pp. 1697–1711, 2001.
[25]  W. Shi and J. S. Bartlett, “RGD inclusion in VP3 provides adeno-associated virus type 2 (AAV2)-based vectors with a heparan sulfate-independent cell entry mechanism,” Molecular Therapy, vol. 7, no. 4, pp. 515–525, 2003.
[26]  C. Dalba, D. Klatzmann, C. R. Logg, and N. Kasahara, “Beyond oncolytic virotherapy: replication-competent retrovirus vectors for selective and stable transduction of tumors,” Current Gene Therapy, vol. 5, no. 6, pp. 655–667, 2005.
[27]  C. K. Tai and N. Kasahara, “Replication-competent retrovirus vectors for cancer gene therapy,” Frontiers in Bioscience, vol. 13, no. 8, pp. 3083–3095, 2008.
[28]  S. J. Russell and F. L. Cosset, “Modifying the host range properties of retroviral vectors,” Journal of Gene Medicine, vol. 1, no. 5, pp. 300–311, 1999.
[29]  O. Erlwein, W. Wels, and B. S. Schnierle, “Chimeric ecotropic MLV envelope proteins that carry EGF receptor-specific ligands and the Pseudomonas exotoxin a translocation domain to target gene transfer to human cancer cells,” Virology, vol. 302, no. 2, pp. 333–341, 2002.
[30]  M. P. Chadwick, F. J. Morling, F. L. Cosset, and S. J. Russell, “Modification of retroviral tropism by display of IGF-I,” Journal of Molecular Biology, vol. 285, no. 2, pp. 485–494, 1999.
[31]  M. Pizzato, E. D. Blair, M. Fling et al., “Evidence for nonspecific adsorption of targeted retrovirus vector particles to cells,” Gene Therapy, vol. 8, no. 14, pp. 1088–1096, 2001.
[32]  K. Guse, V. Cerullo, and A. Hemminki, “Oncolytic vaccinia virus for the treatment of cancer,” Expert Opinion on Biological Therapy, vol. 11, no. 5, pp. 595–608, 2011.
[33]  M. C. Galmiche, L. Rindisbacher, W. Wels, R. Wittek, and F. Buchegger, “Expression of a functional single chain antibody on the surface of extracellular enveloped vaccinia virus as a step towards selective tumour cell targeting,” Journal of General Virology, vol. 78, part 11, pp. 3019–3027, 1997.
[34]  S. Paul, M. Geist, K. Dott et al., “Specific tumor cell targeting by a recombinant MVA expressing a functional single chain antibody on the surface of intracellular mature virus (IMV) particles,” Viral Immunology, vol. 20, no. 4, pp. 664–671, 2007.
[35]  L. Kuo, G. J. Godeke, M. J. Raamsman, P. S. Masters, and P. J. Rottier, “Retargeting of coronavirus by substitution of the spike glycoprotein ectodomain: crossing the host cell species barrier,” Journal of Virology, vol. 74, no. 3, pp. 1393–1406, 2000.
[36]  H. P. Lesch, M. U. Kaikkonen, J. T. Pikkarainen, and S. Yla-Herttuala, “Avidin-biotin technology in targeted therapy,” Expert Opinion on Drug Delivery, vol. 7, no. 5, pp. 551–564, 2010.
[37]  L. Pereboeva, S. Komarova, J. Roth, S. Ponnazhagan, and D. T. Curiel, “Targeting EGFR with metabolically biotinylated fiber-mosaic adenovirus,” Gene Therapy, vol. 14, no. 8, pp. 627–637, 2007.
[38]  J. W. Park, H. Mok, and T. G. Park, “Epidermal growth factor (EGF) receptor targeted delivery of PEGylated adenovirus,” Biochemical and Biophysical Research Communications, vol. 366, no. 3, pp. 769–774, 2008.
[39]  P. Henning, M. K. Magnusson, E. Gunneriusson et al., “Genetic modification of adenovirus 5 tropism by a novel class of ligands based on a three-helix bundle scaffold derived from staphylococcal protein A,” Human Gene Therapy, vol. 13, no. 12, pp. 1427–1439, 2002.
[40]  P. S. Banerjee, E. S. Zuniga, I. Ojima, and I. S. Carrico, “Targeted and armed oncolytic adenovirus via chemoselective modification,” Bioorganic and Medicinal Chemistry Letters, vol. 21, no. 17, pp. 4985–4988, 2011.
[41]  L. Gigout, P. Rebollo, N. Clement et al., “Altering AAV tropism with mosaic viral capsids,” Molecular Therapy, vol. 11, no. 6, pp. 856–865, 2005.
[42]  M. D. Stachler, I. Chen, A. Y. Ting, and J. S. Bartlett, “Site-specific modification of AAV vector particles with biophysical probes and targeting ligands using biotin ligase,” Molecular Therapy, vol. 16, no. 8, pp. 1467–1473, 2008.
[43]  J. I. Quetglas, M. Ruiz-Guillen, A. Aranda, E. Casales, J. Bezunartea, and C. Smerdou, “Alphavirus vectors for cancer therapy,” Virus Research, vol. 153, no. 2, pp. 179–196, 2010.
[44]  K. Ohno, K. Sawai, Y. Lijima, B. Levin, and D. Meruelo, “Cell-specific targeting of sindbis virus vectors displaying IgG-binding domains of protein A,” Nature Biotechnology, vol. 15, no. 8, pp. 763–767, 1997.
[45]  W. B. Klimstra, J. C. Williams, K. D. Ryman, and H. W. Heidner, “Targeting Sindbis virus-based vectors to Fc receptor-positive cell types,” Virology, vol. 338, no. 1, pp. 9–21, 2005.
[46]  C. K. Tai, C. R. Logg, J. M. Park, W. F. Anderson, M. F. Press, and N. Kasahara, “Antibody-mediated targeting of replication-competent retroviral vectors,” Human Gene Therapy, vol. 14, no. 8, pp. 789–802, 2003.
[47]  J. T. Douglas, B. E. Rogers, M. E. Rosenfeld, S. I. Michael, M. Feng, and D. T. Curiel, “Targeted gene delivery by tropism-modified adenoviral vectors,” Nature Biotechnology, vol. 14, no. 11, pp. 1574–1578, 1996.
[48]  S. J. Watkins, V. V. Mesyanzhinov, L. P. Kurochkina, and R. E. Hawkins, “The “adenobody” approach to viral targeting: specific and enhanced adenoviral gene delivery,” Gene Therapy, vol. 4, no. 10, pp. 1004–1012, 1997.
[49]  C. R. Miller, D. J. Buchsbaum, P. N. Reynolds et al., “Differential susceptibility of primary and established human glioma cells to adenovirus infection: targeting via the epidermal growth factor receptor achieves fiber receptor-independent gene transfer,” Cancer Research, vol. 58, no. 24, pp. 5738–5748, 1998.
[50]  B. E. Rogers, J. T. Douglas, C. Ahlem, D. J. Buchsbaum, J. Frincke, and D. T. Curiel, “Use of a novel cross-linking method to modify adenovirus tropism,” Gene Therapy, vol. 4, no. 12, pp. 1387–1392, 1997.
[51]  C. K. Goldman, B. E. Rogers, J. T. Douglas et al., “Targeted gene delivery to Kaposi's sarcoma cells via the fibroblast growth factor receptor,” Cancer Research, vol. 57, no. 8, pp. 1447–1451, 1997.
[52]  D. L. Gu, A. M. Gonzalez, M. A. Printz et al., “Fibroblast growth factor 2 retargeted adenovirus has redirected cellular tropism: evidence for reduced toxicity and enhanced antitumor activity in mice,” Cancer Research, vol. 59, no. 11, pp. 2608–2614, 1999.
[53]  T. J. Wickham, D. M. Segal, P. W. Roelvink et al., “Targeted adenovirus gene transfer to endothelial and smooth muscle cells by using bispecific antibodies,” Journal of Virology, vol. 70, no. 10, pp. 6831–6838, 1996.
[54]  H. J. Haisma, G. K. Kamps, A. Bouma et al., “Selective targeting of adenovirus to αvβ3 integrins, VEGFR2 and Tie2 endothelial receptors by angio-adenobodies,” International Journal of Pharmaceutics, vol. 391, no. 1-2, pp. 155–161, 2010.
[55]  H. J. Haisma, H. M. Pinedo, A. Rijswijk et al., “Tumor-specific gene transfer via an adenoviral vector targeted to the pan-carcinoma antigen EpCAM,” Gene Therapy, vol. 6, no. 8, pp. 1469–1474, 1999.
[56]  D. A. Heideman, P. J. Snijders, M. E. Craanen et al., “Selective gene delivery toward gastric and esophageal adenocarcinoma cells via EpCAM-targeted adenoviral vectors,” Cancer Gene Therapy, vol. 8, no. 5, pp. 342–351, 2001.
[57]  D. M. Nettelbeck, A. A. Rivera, J. Kupsch et al., “Retargeting of adenoviral infection to melanoma: combining genetic ablation of native tropism with a recombinant bispecific single-chain diabody (scDb) adapter that binds to fiber knob and HMWMAA,” International Journal of Cancer, vol. 108, no. 1, pp. 136–145, 2004.
[58]  W. Jongmans, K. van den Oudenalder, D. M. Tiemessen et al., “Targeting of adenovirus to human renal cell carcinoma cells,” Urology, vol. 62, no. 3, pp. 559–565, 2003.
[59]  B. W. Tillman, T. D. de Gruijl, S. A. Luykx-de Bakker et al., “Maturation of dendritic cells accompanies high-efficiency gene transfer by a CD40-targeted adenoviral vector,” Journal of Immunology, vol. 162, no. 11, pp. 6378–6383, 1999.
[60]  B. W. Tillman, T. L. Hayes, T. D. DeGruijl, J. T. Douglas, and D. T. Curiel, “Adenoviral vectors targeted to CD40 enhance the efficacy of dendritic cell-based vaccination against human papillomavirus 16-induced tumor cells in a murine model,” Cancer Research, vol. 60, no. 19, pp. 5456–5463, 2000.
[61]  M. Trepel, M. Grifman, M. D. Weitzman, and R. Pasqualini, “Molecular adaptors for vascular-targeted adenoviral gene delivery,” Human Gene Therapy, vol. 11, no. 14, pp. 1971–1981, 2000.
[62]  M. Breidenbach, D. T. Rein, M. Everts et al., “Mesothelin-mediated targeting of adenoviral vectors for ovarian cancer gene therapy,” Gene Therapy, vol. 12, no. 2, pp. 187–193, 2005.
[63]  R. Kraaij, A. L. van Rijswijk, M. H. Oomen, H. J. Haisma, and C. H. Bangma, “Prostate specific membrane antigen (PSMA) is a tissue-specific target for adenoviral transduction of prostate cancer in vitro,” Prostate, vol. 62, no. 3, pp. 253–259, 2005.
[64]  H. J. van Zeeburg, V. W. van Beusechem, A. Huizenga et al., “Adenovirus retargeting to surface expressed antigens on oral mucosa,” Journal of Gene Medicine, vol. 12, no. 4, pp. 365–376, 2010.
[65]  E. Li, S. L. Brown, D. J. von Seggern, G. B. Brown, and G. R. Nemerow, “Signaling antibodies complexed with adenovirus circumvent CAR and integrin interactions and improve gene delivery,” Gene Therapy, vol. 7, no. 18, pp. 1593–1599, 2000.
[66]  S. K. Yoon, L. Mohr, C. R. O'Riordan, A. Lachapelle, D. Armentano, and J. R. Wands, “Targeting a recombinant adenovirus vector to HCC cells using a bifunctional Fab-antibody conjugate,” Biochemical and Biophysical Research Communications, vol. 272, no. 2, pp. 497–504, 2000.
[67]  I. Dmitriev, E. Kashentseva, B. E. Rogers, V. Krasnykh, and D. T. Curiel, “Ectodomain of coxsackievirus and adenovirus receptor genetically fused to epidermal growth factor mediates adenovirus targeting to epidermal growth factor receptor-positive cells,” Journal of Virology, vol. 74, no. 15, pp. 6875–6884, 2000.
[68]  J. G. Wesseling, P. J. Bosma, V. Krasnykh et al., “Improved gene transfer efficiency to primary and established human pancreatic carcinoma target cells via epidermal growth factor receptor and integrin-targeted adenoviral vectors,” Gene Therapy, vol. 8, no. 13, pp. 969–976, 2001.
[69]  C. Ebbinghaus, A. Al-Jaibaji, E. Operschall et al., “Functional and selective targeting of adenovirus to high-affinity Fcγ receptor I-positive cells by using a bispecific hybrid adapter,” Journal of Virology, vol. 75, no. 1, pp. 480–489, 2001.
[70]  E. A. Kashentseva, T. Seki, D. T. Curiel, and I. P. Dmitriev, “Adenovirus targeting to c-erbB-2 oncoprotein by single-chain antibody fused to trimeric form of adenovirus receptor ectodomain,” Cancer Research, vol. 62, no. 2, pp. 609–616, 2002.
[71]  H. J. Li, M. Everts, L. Pereboeva et al., “Adenovirus tumor targeting and hepatic untargeting by a coxsackie/adenovirus receptor ectodomain anticarcinoembryonic antigen bispecific adapter,” Cancer Research, vol. 67, no. 11, pp. 5354–5361, 2007.
[72]  N. K. Green, J. Morrison, S. Hale et al., “Retargeting polymer-coated adenovirus to the FGF receptor allows productive infection and mediates efficacy in a peritoneal model of human ovarian cancer,” Journal of Gene Medicine, vol. 10, no. 3, pp. 280–289, 2008.
[73]  J. Moselhy, S. Sarkar, M. C. Chia et al., “Evaluation of copolymers of N-isopropylacrylamide and 2-dimethyl(aminoethyl)methacrylate in nonviral and adenoviral vectors for gene delivery to nasopharyngeal carcinoma,” International Journal of Nanomedicine, vol. 2, no. 3, pp. 461–478, 2007.
[74]  J. Han, D. Zhao, Z. Zhong, Z. Zhang, T. Gong, and X. Sun, “Combination of adenovirus and cross-linked low molecular weight PEI improves efficiency of gene transduction,” Nanotechnology, vol. 21, no. 10, Article ID 105106, 2010.
[75]  H. Bachtarzi, M. Stevenson, V. Subr, K. Ulbrich, L. W. Seymour, and K. D. Fisher, “Targeting adenovirus gene delivery to activated tumour-associated vasculature via endothelial selectins,” Journal of Controlled Release, vol. 150, no. 2, pp. 196–203, 2011.
[76]  K. D. Fisher, N. K. Green, A. Hale, V. Subr, K. Ulbrich, and L. W. Seymour, “Passive tumour targeting of polymer-coated adenovirus for cancer gene therapy,” Journal of Drug Targeting, vol. 15, no. 7-8, pp. 546–551, 2007.
[77]  M. Stevenson, A. B. Hale, S. J. Hale et al., “Incorporation of a laminin-derived peptide (SIKVAV) on polymer-modified adenovirus permits tumor-specific targeting via α6-integrins,” Cancer Gene Therapy, vol. 14, no. 4, pp. 335–345, 2007.
[78]  J. Lanciotti, A. Song, J. Doukas et al., “Targeting adenoviral vectors using heterofunctional polyethylene glycol FGF2 conjugates,” Molecular Therapy, vol. 8, no. 1, pp. 99–107, 2003.
[79]  Y. Eto, J. Q. Gao, F. Sekiguchi et al., “PEGylated adenovirus vectors containing RGD peptides on the tip of PEG show high transduction efficiency and antibody evasion ability,” Journal of Gene Medicine, vol. 7, no. 5, pp. 604–612, 2005.
[80]  J. Q. Gao, Y. Eto, Y. Yoshioka et al., “Effective tumor targeted gene transfer using PEGylated adenovirus vector via systemic administration,” Journal of Controlled Release, vol. 122, no. 1, pp. 102–110, 2007.
[81]  Y. Jung, H. J. Park, P. H. Kim et al., “Retargeting of adenoviral gene delivery via Herceptin-PEG-adenovirus conjugates to breast cancer cells,” Journal of Controlled Release, vol. 123, no. 2, pp. 164–171, 2007.
[82]  C. Y. Chen, S. M. May, and M. A. Barry, “Targeting adenoviruses with factor X-single-chain antibody fusion proteins,” Human Gene Therapy, vol. 21, no. 6, pp. 739–749, 2010.
[83]  J. S. Bartlett, J. Kleinschmidt, R. C. Boucher, and R. J. Samulski, “Targeted adeno-associated virus vector transduction of nonpermissive cells mediated cells by a bispecific F(ab'γ)2 antibody,” Nature Biotechnology, vol. 17, no. 2, pp. 181–186, 1999.
[84]  K. Nakano, R. Asano, K. Tsumoto et al., “Herpes simplex virus targeting to the EGF receptor by a gD-Specific soluble bridging molecule,” Molecular Therapy, vol. 11, no. 4, pp. 617–626, 2005.
[85]  H. Bian, P. Fournier, R. Moormann, B. Peeters, and V. Schirrmacher, “Selective gene transfer in vitro to tumor cells via recombinant newcastle disease virus,” Cancer Gene Therapy, vol. 12, no. 3, pp. 295–303, 2005.
[86]  H. Bian, H. Wilden, P. Fournier, B. Peeters, and V. Schirrmacher, “In vivo efficacy of systemic tumor targeting of a viral RNA vector with oncolytic properties using a bispecific adapter protein,” International Journal of Oncology, vol. 29, no. 6, pp. 1359–1369, 2006.
[87]  B. J. Haijema, H. Volders, and P. J. Rottier, “Switching species tropism: an effective way to manipulate the feline coronavirus genome,” Journal of Virology, vol. 77, no. 8, pp. 4528–4538, 2003.
[88]  T. Wurdinger, M. H. Verheije, M. Raaben et al., “Targeting non-human coronaviruses to human cancer cells using a bispecific single-chain antibody,” Gene Therapy, vol. 12, no. 18, pp. 1394–1404, 2005.
[89]  T. Wurdinger, M. H. Verheije, K. Broen et al., “Soluble receptor-mediated targeting of mouse hepatitis coronavirus to the human epidermal growth factor receptor,” Journal of Virology, vol. 79, no. 24, pp. 15314–15322, 2005.
[90]  M. H. Verheije, M. L. Lamfers, T. Wurdinger et al., “Coronavirus genetically redirected to the epidermal growth factor receptor exhibits effective antitumor activity against a malignant glioblastoma,” Journal of Virology, vol. 83, no. 15, pp. 7507–7516, 2009.
[91]  A. Hemminki, I. Dmitriev, B. Liu, R. A. Desmond, R. Alemany, and D. T. Curiel, “Targeting oncolytic adenoviral agents to the epidermal growth factor pathway with a secretory fusion molecule,” Cancer Research, vol. 61, no. 17, pp. 6377–6381, 2001.
[92]  V. W. van Beusechem, D. C. Mastenbroek, P. B. van den Doel et al., “Conditionally replicative adenovirus expressing a targeting adapter molecule exhibits enhanced oncolytic potency on CAR-deficient tumors,” Gene Therapy, vol. 10, no. 23, pp. 1982–1991, 2003.
[93]  J. E. Carette, H. C. Graat, F. H. Schagen et al., “A conditionally replicating adenovirus with strict selectivity in killing cells expressing epidermal growth factor receptor,” Virology, vol. 361, no. 1, pp. 56–67, 2007.
[94]  D. A. Einfeld, R. Schroeder, P. W. Roelvink et al., “Reducing the native tropism of adenovirus vectors requires removal of both CAR and integrin interactions,” Journal of Virology, vol. 75, no. 23, pp. 11284–11291, 2001.
[95]  A. Hemminki, M. Wang, T. Hakkarainen, R. A. Desmond, J. Wahlfors, and D. T. Curiel, “Production of an EGFR targeting molecule from a conditionally replicating adenovirus impairs its oncolytic potential,” Cancer Gene Therapy, vol. 10, no. 8, pp. 583–588, 2003.
[96]  J. N. Glasgow, G. Mikheeva, V. Krasnykh, and D. T. Curiel, “A strategy for adenovirus vector targeting with a secreted single chain antibody,” Plos One, vol. 4, no. 12, Article ID e8355, 2009.
[97]  V. W. Van Beusechem, J. Grill, D. C. Jeroen Mastenbroek et al., “Efficient and selective gene transfer into primary human brain tumors by using single-chain antibody-targeted adenoviral vectors with native tropism abolished,” Journal of Virology, vol. 76, no. 6, pp. 2753–2762, 2002.
[98]  S. D. Barker, I. P. Dmitriev, D. M. Nettelbeck et al., “Combined transcriptional and transductional targeting improves the specificity and efficacy of adenoviral gene delivery to ovarian carcinoma,” Gene Therapy, vol. 10, no. 14, pp. 1198–1204, 2003.
[99]  M. Everts, S. A. Kim-Park, M. A. Preuss et al., “Selective induction of tumor-associated antigens in murine pulmonary vasculature using double-targeted adenoviral vectors,” Gene Therapy, vol. 12, no. 13, pp. 1042–1048, 2005.
[100]  H. J. Li, M. Everts, M. Yamamoto, D. T. Curiel, and H. R. Herschman, “Combined transductional untargeting/retargeting and transcriptional restriction enhances adenovirus gene targeting and therapy for hepatic colorectal cancer tumors,” Cancer Research, vol. 69, no. 2, pp. 554–564, 2009.
[101]  P. N. Reynolds, S. A. Nicklin, L. Kaliberova et al., “Combined transductional and transcriptional targeting improves the specificity of transgene expression in vivo,” Nature Biotechnology, vol. 19, no. 9, pp. 838–842, 2001.
[102]  P. N. Reynolds, K. R. Zinn, V. D. Gavrilyuk et al., “A targetable, injectable adenoviral vector for selective gene delivery to pulmonary endothelium in vivo,” Molecular Therapy, vol. 2, no. 6, pp. 562–578, 2000.
[103]  M. H. Verheije, T. Wurdinger, V. W. van Beusechem, C. A. de Haan, W. R. Gerritsen, and P. J. Rottier, “Redirecting coronavirus to a nonnative receptor through a virus-encoded targeting adapter,” Journal of Virology, vol. 80, no. 3, pp. 1250–1260, 2006.
[104]  C. A. de Haan, L. van Genne, J. N. Stoop, H. Volders, and P. J. Rottier, “Coronaviruses as vectors: position dependence of foreign gene expression,” Journal of Virology, vol. 77, no. 21, pp. 11312–11323, 2003.
[105]  K. J. Kimball, M. A. Preuss, M. N. Barnes et al., “A phase I study of a tropism-modified conditionally replicative adenovirus for recurrent malignant gynecologic diseases,” Clinical Cancer Research, vol. 16, no. 21, pp. 5277–5287, 2010.
[106]  D. Koppers-Lalic and R. C. Hoeben, “Non-human viruses developed as therapeutic agent for use in humans,” Reviews in Medical Virology, vol. 21, no. 4, pp. 227–239, 2011.
[107]  N. K. Green, C. W. Herbert, S. J. Hale et al., “Extended plasma circulation time and decreased toxicity of polymer-coated adenovirus,” Gene Therapy, vol. 11, no. 16, pp. 1256–1263, 2004.
[108]  H. Nakashima, B. Kaur, and E. A. Chiocca, “Directing systemic oncolytic viral delivery to tumors via carrier cells,” Cytokine and Growth Factor Reviews, vol. 21, no. 2-3, pp. 119–126, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413