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...

Silencing Sexually Transmitted Infections: Topical siRNA-Based Interventions for the Prevention of HIV and HSV

DOI: 10.1155/2014/125087

Full-Text   Cite this paper   Add to My Lib

Abstract:

The global impact of sexually transmitted infections (STIs) is significant. The sexual transmission of viruses such as herpes simplex virus type-2 (HSV-2) and the human immunodeficiency virus type-1 (HIV-1), has been especially difficult to control. To date, no effective vaccines have been developed to prevent the transmission of these STIs. Although antiretroviral drugs have been remarkably successful in treating the symptoms associated with these viral infections, the feasibility of their widespread use for prevention purposes may be more limited. Microbicides might provide an attractive alternative option to reduce their spread. In particular, topically applied small inhibitory RNAs (siRNAs) have been shown to not only block transmission of viral STIs to mucosal tissues both in vitro and in vivo, but also confer durable knockdown of target gene expression, thereby circumventing the need to apply a microbicide around the time of sexual encounter, when compliance is mostly difficult. Despite numerous clinical trials currently testing the efficacy of siRNA-based therapeutics, they have yet to be approved for use in the treatment of viral STIs. While several obstacles to their successful implementation in the clinic still exist, promising preclinical studies suggest that siRNAs are a viable modality for the future prevention and treatment of HSV and HIV. 1. Clinical Context Sexually transmitted infections (STIs) continue to be a major source of global morbidity and mortality [1]. Viral infections, notably herpes simplex virus type-2 (HSV-2) and the human immunodeficiency virus type-1 (HIV-1), have proven particularly problematic to control from both scientific and public health perspectives. HSV-2, the most common cause of genital ulcers, is the most widespread viral sexually transmitted infection (STI) worldwide [2]. Prevalence is high; currently estimated to be ~15–25% of sexually active adults in the US and as high as 60–80% in some developing nations [2–5]. Rates of infection are even higher among women, owing to the greater efficiency of male-to-female (MTF) transmission [6, 7]. While multiple variables factor into the high prevalence rate at the population level, the fact that most sexual transmissions occur in the absence of clinically identifiable genital lesions likely plays a major role [8, 9]. While antiviral drugs have proven effective to reduce viral transmission [6, 8, 10], they are not curative. Furthermore, due to the high costs associated with population-wide screening and maintenance therapy, their utility as public health initiatives

References

[1]  R. J. Whitley, Field’s Virology, Lippincott Williams & Wilkins, Philadelphia, Pa, USA, 2001.
[2]  Centers for Disease Control and Prevention , “Seroprevalence of herpes simplex virus type 2 among persons aged 14–49 years—United States, 2005–2008,” Morbidity and Mortality Weekly Report, vol. 59, pp. 456–459, 2010.
[3]  H. Weiss, “Epidemiology of herpes simplex virus type 2 infection in the developing world,” Herpes, vol. 11, supplement 1, pp. 24A–35A, 2004.
[4]  L. Corey, A. Wald, C. L. Celum, and T. C. Quinn, “The effects of herpes simplex virus-2 on HIV-1 acquisition and transmission: a review of two overlapping epidemics,” Journal of Acquired Immune Deficiency Syndromes, vol. 35, no. 5, pp. 435–445, 2004.
[5]  D. M. Koelle and A. Wald, “Herpes simplex virus: the importance of asymptomatic shedding,” Journal of Antimicrobial Chemotherapy, vol. 45, supplement 3, pp. 1–8, 2000.
[6]  L. Corey, A. Wald, R. Patel et al., “Once-daily valacyclovir to reduce the risk of transmission of genital herpes,” The New England Journal of Medicine, vol. 350, no. 1, pp. 11–20, 2004.
[7]  L. R. Stanberry, S. L. Spruance, A. L. Cunningham et al., “Glycoprotein-D-adjuvant vaccine to prevent genital herpes,” The New England Journal of Medicine, vol. 347, no. 21, pp. 1652–1661, 2002.
[8]  R. Gupta, A. Wald, E. Krantz et al., “Valacyclovir and acyclovir for suppression of shedding of herpes simplex virus in the genital tract,” Journal of Infectious Diseases, vol. 190, no. 8, pp. 1374–1381, 2004.
[9]  G. J. Mertz, O. Schmidt, and J. L. Jourden, “Frequency of acquisition of first-episode genital infection with herpes simplex virus from symptomatic and asymptomatic source contacts,” Sexually Transmitted Diseases, vol. 12, no. 1, pp. 33–39, 1985.
[10]  A. Wald, D. Carrell, M. Remington, E. Kexel, J. Zeh, and L. Corey, “Two-day regimen of acyclovir for treatment of recurrent genital herpes simplex virus type 2 infection,” Clinical Infectious Diseases, vol. 34, no. 7, pp. 944–948, 2002.
[11]  P. E. Bonnar, “Suppressive valacyclovir therapy to reduce genital herpes transmission: good public health policy?” McGill Journal of Medicine, vol. 12, pp. 39–46, 2009.
[12]  C. Casper and A. Wald, “Condom use and the prevention of genital herpes acquisition,” Herpes, vol. 9, no. 1, pp. 10–14, 2002.
[13]  A. Wald, A. G. M. Langenberg, K. Link et al., “Effect of condoms on reducing the transmission of herpes simplex virus type 2 from men to women,” Journal of the American Medical Association, vol. 285, no. 24, pp. 3100–3106, 2001.
[14]  D. Serwadda, R. H. Gray, N. K. Sewankambo et al., “Human immunodeficiency virus acquisition associated with genital ulcer disease and herpes simplex virus type 2 infection: a nested case-control study in Rakai, Uganda,” Journal of Infectious Diseases, vol. 188, no. 10, pp. 1492–1497, 2003.
[15]  A. Wald and K. Link, “Risk of human immunodeficiency virus infection in herpes simplex virus type 2-seropositive persons: a meta-analysis,” Journal of Infectious Diseases, vol. 185, no. 1, pp. 45–52, 2002.
[16]  E. E. Freeman, H. A. Weiss, J. R. Glynn, P. L. Cross, J. A. Whitworth, and R. J. Hayes, “Herpes simplex virus 2 infection increases HIV acquisition in men and women: systematic review and meta-analysis of longitudinal studies,” AIDS, vol. 20, no. 1, pp. 73–83, 2006.
[17]  UNAIDS, Report on the Global HIV/AIDS Epidemic 2008: Executive Summary, UNAIDS, Geneva, Switzerland, 2008.
[18]  M. S. Hirsch and J. C. Kaplan, “Prospects of therapy for infections with human T-lymphotropic virus type III,” Annals of Internal Medicine, vol. 103, no. 5, pp. 750–755, 1985.
[19]  T. Beardsley, “AIDS progress. Synthetic vaccine only a distant prospect,” Nature, vol. 314, no. 6013, p. 659, 1985.
[20]  P. S. Sarma, K. J. Cremer, and J. Gruber, “Acquired immunodeficiency syndrome: progress and prospects for vaccine development,” Journal of the National Cancer Institute, vol. 80, no. 15, pp. 1193–1197, 1988.
[21]  W. C. Koff and D. F. Hoth, “Development and testing of AIDS vaccines,” Science, vol. 241, no. 4864, pp. 426–432, 1988.
[22]  H. W. Virgin and B. D. Walker, “Immunology and the elusive AIDS vaccine,” Nature, vol. 464, no. 7286, pp. 224–231, 2010.
[23]  B. D. Walker and D. R. Burton, “Toward an AIDS vaccine,” Science, vol. 320, no. 5877, pp. 760–764, 2008.
[24]  S. W. Lagakos and A. R. Gable, “Challenges to HIV prevention—seeking effective measures in the absence of a vaccine,” The New England Journal of Medicine, vol. 358, no. 15, pp. 1543–1545, 2008.
[25]  E. L. Murphy, A. C. Collier, L. A. Kalish et al., “Highly active antiretroviral therapy decreases mortality and morbidity in patients with advanced HIV disease,” Annals of Internal Medicine, vol. 135, no. 1, pp. 17–26, 2001.
[26]  V. Simon and D. D. Ho, “HIV-1 dynamics in vivo: implications for therapy,” Nature reviews. Microbiology, vol. 1, no. 3, pp. 181–190, 2003.
[27]  R. S. Hogg, K. V. Heath, B. Yip et al., “Improved survival among HIV-infected individuals following initiation of antiretroviral therapy,” Journal of the American Medical Association, vol. 279, no. 6, pp. 450–454, 1998.
[28]  F. J. Palella Jr., K. M. Delaney, A. C. Moorman et al., “Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection,” The New England Journal of Medicine, vol. 338, no. 13, pp. 853–860, 1998.
[29]  D. C. Watson and J. J. Farley, “Efficacy of and adherence to highly active antiretroviral therapy in children infected with human immunodeficiency virus type 1,” Pediatric Infectious Disease Journal, vol. 18, no. 8, pp. 682–689, 1999.
[30]  M. de Martino, P.-A. Tovo, M. Balducci et al., “Reduction in mortality with availability of antiretroviral therapy for children with perinatal HIV-1 infection,” Journal of the American Medical Association, vol. 284, no. 2, pp. 190–197, 2000.
[31]  T.-W. Chun, L. Carruth, D. Finzi et al., “Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection,” Nature, vol. 387, no. 6629, pp. 183–188, 1997.
[32]  V. Giacomet, F. Albano, F. Starace et al., “Adherence to antiretroviral therapy and its determinants in children with human immunodeficiency virus infection: a multicentre, national study,” Acta Paediatrica, vol. 92, no. 12, pp. 1398–1402, 2003.
[33]  M. P. Carrieri, F. Raffi, C. Lewden et al., “Impact of early versus late adherence to highly active antiretroviral therapy on immuno-virological response: a 3-year follow-up study,” Antiviral Therapy, vol. 8, no. 6, pp. 585–594, 2003.
[34]  D. L. Paterson, S. Swindells, J. Mohr et al., “Adherence to protease inhibitor therapy and outcomes in patients with HIV infection,” Annals of Internal Medicine, vol. 133, no. 1, pp. 21–30, 2000.
[35]  J. A. Epping-Jordan, R. Bengoa, R. Kawar, and E. Sabaté, “The challenge of chronic conditions: WHO responds,” British Medical Journal, vol. 323, no. 7319, pp. 947–948, 2001.
[36]  E. Wood, R. S. Hogg, B. Yip, P. R. Harrigan, M. V. O'Shaughnessy, and J. S. G. Montaner, “Effect of medication adherence on survival of HIV-infected adults who start highly active antiretroviral therapy when the CD4+ cell count is 0.200 to 0.350 x 10(9) cells/L,” Annals of Internal Medicine, vol. 139, no. 10, pp. 810–816, 2003.
[37]  I. Lanièce, M. Ciss, A. Desclaux et al., “Adherence to HAART and its principal determinants in a cohort of Senegalese adults,” AIDS, vol. 17, no. 3, pp. S103–S108, 2003.
[38]  L. Weiss, T. French, R. Finkelstein, M. Waters, R. Mukherjee, and B. Agins, “HIV-related knowledge and adherence to HAART,” AIDS Care, vol. 15, no. 5, pp. 673–679, 2003.
[39]  D. A. Murphy, M. Sarr, S. J. Durako, A.-B. Moscicki, C. M. Wilson, and L. R. Muenz, “Barriers to HAART adherence among human immunodeficiency virus—infected adolescents,” Archives of Pediatrics and Adolescent Medicine, vol. 157, no. 3, pp. 249–255, 2003.
[40]  B. S. Peters and K. Conway, “Therapy for HIV: past, present, and future,” Advances in Dental Research, vol. 23, no. 1, pp. 23–27, 2011.
[41]  A. Krüsi, E. Wood, J. Montaner, and T. Kerr, “Social and structural determinants of HAART access and adherence among injection drug users,” International Journal of Drug Policy, vol. 21, no. 1, pp. 4–9, 2010.
[42]  F. Orsi and C. D'Almeida, “Soaring antiretroviral prices, TRIPS and TRIPS flexibilities: a burning issue for antiretroviral treatment scale-up in developing countries,” Current Opinion in HIV and AIDS, vol. 5, no. 3, pp. 237–241, 2010.
[43]  V. Simon, J. Vanderhoeven, A. Hurley et al., “Evolving patterns of HIV-1 resistance to antiretroviral agents in newly infected individuals,” AIDS, vol. 16, no. 11, pp. 1511–1519, 2002.
[44]  D. Finzi, M. Hermankova, T. Pierson et al., “Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy,” Science, vol. 278, no. 5341, pp. 1295–1300, 1997.
[45]  L. Zhang, B. Ramratnam, K. Tenner-Racz et al., “Quantifying residual HIV-1 replication in patients receiving combination antiretroviral therapy,” The New England Journal of Medicine, vol. 340, no. 21, pp. 1605–1613, 1999.
[46]  O. Turriziani, M. Andreoni, and G. Antonelli, “Resistant viral variants in cellular reservoirs of human immunodeficiency virus infection,” Clinical Microbiology and Infection, vol. 16, no. 10, pp. 1518–1524, 2010.
[47]  D. D. Ho and L. Zhang, “HIV-1 rebound after anti-retroviral therapy,” Nature Medicine, vol. 6, no. 7, pp. 736–737, 2000.
[48]  B. Ramratnam, J. E. Mittler, L. Zhang et al., “The decay of the latent reservoir of replication-competent HIV-1 is inversely correlated with the extent of residual viral replication during prolonged anti-retroviral therapy,” Nature Medicine, vol. 6, no. 1, pp. 82–85, 2000.
[49]  F. Hladik and G. F. Doncel, “Preventing mucosal HIV transmission with topical microbicides: challenges and opportunities,” Antiviral Research, vol. 88, supplement 1, pp. S3–S9, 2010.
[50]  N. S. Padian, A. Buvé, J. Balkus, D. Serwadda, and W. Cates Jr., “Biomedical interventions to prevent HIV infection: evidence, challenges, and way forward,” The Lancet, vol. 372, no. 9638, pp. 585–599, 2008.
[51]  L. A. Paxton, T. Hope, and H. W. Jaffe, “Pre-exposure prophylaxis for HIV infection: what if it works?” The Lancet, vol. 370, no. 9581, pp. 89–93, 2007.
[52]  K. Underhill, D. Operario, M. J. Mimiaga, M. R. Skeer, and K. H. Mayer, “Implementation science of pre-exposure prophylaxis: preparing for public Use,” Current HIV/AIDS Reports, vol. 7, no. 4, pp. 210–219, 2010.
[53]  A. K. Sethi, D. D. Celentano, S. J. Gange, R. D. Moore, and J. E. Gallant, “Association between adherence to antiretroviral therapy and human immunodeficiency virus drug resistance,” Clinical Infectious Diseases, vol. 37, no. 8, pp. 1112–1118, 2003.
[54]  UNAIDS, “World AIDS day report 2011,” in World AIDS Day Report, UNAIDS, Geneva, Switzerland, 2011.
[55]  M. J. McElrath, S. C. de Rosa, Z. Moodie et al., “HIV-1 vaccine-induced immunity in the test-of-concept step study: a case-cohort analysis,” The Lancet, vol. 372, no. 9653, pp. 1894–1905, 2008.
[56]  S. P. Buchbinder, D. V. Mehrotra, A. Duerr et al., “Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the step study): a double-blind, randomised, placebo-controlled, test-of-concept trial,” The Lancet, vol. 372, no. 9653, pp. 1881–1893, 2008.
[57]  J. Cohen, “Beyond thailand: making sense of a qualified AIDS vaccine ‘success’,” Science, vol. 326, no. 5953, pp. 652–653, 2009.
[58]  N. Karasavvas, E. Billings, M. Rao et al., “The Thai Phase III HIV Type 1 Vaccine trial (RV144) regimen induces antibodies that target conserved regions within the V2 loop of gp120,” AIDS Research and Human Retroviruses, vol. 28, pp. 1444–1457, 2012.
[59]  A. Sabchareon, D. Wallace, C. Sirivichayakul et al., “Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial,” The Lancet, vol. 380, pp. 1559–1567, 2012.
[60]  T. Dudek and D. M. Knipe, “Replication-defective viruses as vaccines and vaccine vectors,” Virology, vol. 344, no. 1, pp. 230–239, 2006.
[61]  Y. Hoshino, S. K. Dalai, K. Wang et al., “Comparative efficacy and immunogenicity of replication-defective, recombinant glycoprotein, and DNA vaccines for herpes simplex virus 2 infections in mice and guinea pigs,” Journal of Virology, vol. 79, pp. 410–418, 2005.
[62]  J. T. Bryan, “Developing an HPV vaccine to prevent cervical cancer and genital warts,” Vaccine, vol. 25, no. 16, pp. 3001–3006, 2007.
[63]  R. K. Zimmerman, “HPV vaccine and its recommendations, 2007,” Journal of Family Practice, vol. 56, no. 2, pp. S1–S5, 2007.
[64]  E. A. Joura, S. Leodolter, M. Hernandez-Avila et al., “Efficacy of a quadrivalent prophylactic human papillomavirus (types 6, 11, 16, and 18) L1 virus-like-particle vaccine against high-grade vulval and vaginal lesions: a combined analysis of three randomised clinical trials,” The Lancet, vol. 369, no. 9574, pp. 1693–1702, 2007.
[65]  S. M. Garland, M. Hernandez-Avila, C. M. Wheeler et al., “Quadrivalent vaccine against human papillomavirus to prevent anogenital diseases,” The New England Journal of Medicine, vol. 356, no. 19, pp. 1928–1943, 2007.
[66]  L. R. Minces and I. McGowan, “Advances in the development of microbicides for the prevention of HIV infection,” Current Infectious Disease Reports, vol. 12, no. 1, pp. 56–62, 2010.
[67]  J. M. Lange, M. Karam, and P. Piot, “Boost for vaginal microbicides against HIV,” The Lancet, vol. 342, no. 8883, p. 1356, 1993.
[68]  D. S. Nikolic, E. Garcia, and V. Piguet, “Microbicides and other topical agents in the prevention of HIV and sexually transmitted infections,” Expert Review of Anti-Infective Therapy, vol. 5, no. 1, pp. 77–88, 2007.
[69]  A. S. Bourinbaiar and E. C. Fruhstorfer, “The efficacy of nonoxynol-9 from an in vitro point of view,” AIDS, vol. 10, no. 5, pp. 558–559, 1996.
[70]  M. Malkovsky, A. Newell, and A. G. Dalgleish, “Inactivation of HIV by nonoxynol-9,” The Lancet, vol. 1, no. 8586, p. 645, 1988.
[71]  B. C. Herold, R. Kirkpatrick, D. Marcellino et al., “Bile salts: natural detergents for the prevention of sexually transmitted diseases,” Antimicrobial Agents and Chemotherapy, vol. 43, no. 4, pp. 745–751, 1999.
[72]  M. K. Stafford, H. Ward, A. Flanagan et al., “Safety study of nonoxynol-9 as a vaginal microbicide: evidence of adverse effects,” Journal of Acquired Immune Deficiency Syndromes and Human Retrovirology, vol. 17, no. 4, pp. 327–331, 1998.
[73]  R. E. Roddy, L. Zekeng, K. A. Ryan, U. Tamoufé, S. S. Weir, and E. L. Wong, “A controlled trial of nonoxynol 9 film to reduce male-to-female transmission of sexually transmitted diseases,” The New England Journal of Medicine, vol. 339, no. 8, pp. 504–510, 1998.
[74]  L. van Damme, G. Ramjee, M. Alary et al., “Effectiveness of COL-1492, a nonoxynol-9 vaginal gel, on HIV-1 transmission in female sex workers: a randomised controlled trial,” The Lancet, vol. 360, no. 9338, pp. 971–977, 2002.
[75]  S. L. Hillier, T. Moench, R. Shattock, R. Black, P. Reichelderfer, and F. Veronese, “In vitro and in vivo: the story of nonoxynol 9,” Journal of Acquired Immune Deficiency Syndromes, vol. 39, no. 1, pp. 1–8, 2005.
[76]  K. H. Mayer, J. Peipert, T. Fleming et al., “Safety and tolerability of BufferGel, a novel vaginal microbicide, in women in the United States,” Clinical Infectious Diseases, vol. 32, no. 3, pp. 476–482, 2001.
[77]  L. van Damme, R. Govinden, F. M. Mirembe et al., “Lack of effectiveness of cellulose sulfate gel for the prevention of vaginal HIV transmission,” The New England Journal of Medicine, vol. 359, no. 5, pp. 463–472, 2008.
[78]  M. J. Keller, A. Tuyama, M. J. Carlucci, and B. C. Herold, “Topical microbicides for the prevention of genital herpes infection,” Journal of Antimicrobial Chemotherapy, vol. 55, no. 4, pp. 420–423, 2005.
[79]  I. McGowan, “Microbicides for HIV prevention: reality or hope?” Current Opinion in Infectious Diseases, vol. 23, no. 1, pp. 26–31, 2010.
[80]  Q. A. Karim, S. S. A. Karim, J. A. Frohlich et al., “Effectiveness and safety of tenofovir gel, an antiretroviral microbicide, for the prevention of HIV infection in women,” Science, vol. 329, no. 5996, pp. 1168–1174, 2010.
[81]  I. McGowan, “Rectal microbicide development,” Current Opinion in HIV and AIDS, vol. 7, pp. 526–533, 2012.
[82]  P. A. Anton, R. D. Cranston, A. Kashuba et al., “RMP-02/MTN-006: a phase 1 rectal safety, acceptability, pharmacokinetic, and pharmacodynamic study of tenofovir 1% gel compared with oral tenofovir disoproxil fumarate,” AIDS Research and Human Retroviruses, vol. 28, pp. 1412–1421, 2012.
[83]  C. Celum and J. M. Baeten, “Tenofovir-based pre-exposure prophylaxis for HIV prevention: evolving evidence,” Current Opinion in Infectious Diseases, vol. 25, no. 1, pp. 51–57, 2012.
[84]  S. M. Elbashir, J. Harborth, W. Lendeckel, A. Yalcin, K. Weber, and T. Tuschl, “Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells,” Nature, vol. 411, no. 6836, pp. 494–498, 2001.
[85]  A. Fire, S. Xu, M. K. Montgomery, S. A. Kostas, S. E. Driver, and C. C. Mello, “Potent and specific genetic interference by double-stranded RNA in caenorhabditis elegans,” Nature, vol. 391, no. 6669, pp. 806–811, 1998.
[86]  R. Jorgensen, “Altered gene expression in plants due to trans interactions between homologous genes,” Trends in Biotechnology, vol. 8, no. 12, pp. 340–344, 1990.
[87]  B. Wightman, I. Ha, and G. Ruvkun, “Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans,” Cell, vol. 75, no. 5, pp. 855–862, 1993.
[88]  R. C. Lee, R. L. Feinbaum, and V. Ambros, “The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14,” Cell, vol. 75, no. 5, pp. 843–854, 1993.
[89]  C. D. Novina, M. F. Murray, D. M. Dykxhoorn et al., “siRNA-directed inhibition of HIV-1 infection,” Nature Medicine, vol. 8, pp. 681–686, 2002.
[90]  S.-K. Lee, D. M. Dykxhoorn, P. Kumar et al., “Lentiviral delivery of short hairpin RNAs protects CD4 T cells from multiple clades and primary isolates of HIV,” Blood, vol. 106, no. 3, pp. 818–826, 2005.
[91]  P. Kumar, H.-S. Ban, S.-S. Kim et al., “T cell-specific siRNA delivery suppresses HIV-1 infection in humanized mice,” Cell, vol. 134, no. 4, pp. 577–586, 2008.
[92]  E. Song, S.-K. Lee, D. M. Dykxhoorn et al., “Sustained small interfering RNA-mediated human immunodeficiency virus type 1 inhibition in primary macrophages,” Journal of Virology, vol. 77, no. 13, pp. 7174–7181, 2003.
[93]  E. Song, P. Zhu, S.-K. Lee et al., “Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors,” Nature Biotechnology, vol. 23, no. 6, pp. 709–717, 2005.
[94]  V. Bitko, A. Musiyenko, O. Shulyayeva, and S. Barik, “Inhibition of respiratory viruses by nasally administered siRNA,” Nature Medicine, vol. 11, no. 1, pp. 50–55, 2005.
[95]  D. Palliser, D. Chowdhury, Q.-Y. Wang et al., “An siRNA-based microbicide protects mice from lethal herpes simplex virus 2 infection,” Nature, vol. 439, no. 7072, pp. 89–94, 2006.
[96]  Y. Wu, F. Navarro, A. Lal et al., “Durable protection from herpes simplex virus-2 transmission following intravaginal application of siRNAs targeting both a viral and host gene,” Cell Host and Microbe, vol. 5, no. 1, pp. 84–94, 2009.
[97]  D. Peer, J. P. Eun, Y. Morishita, C. V. Carman, and M. Shimaoka, “Systemic leukocyte-directed siRNA delivery revealing cyclin D1 as an anti-inflammatory target,” Science, vol. 319, no. 5863, pp. 627–630, 2008.
[98]  D. Peer, P. Zhu, C. V. Carman, J. Lieberman, and M. Shimaoka, “Selective gene silencing in activated leukocytes by targeting siRNAs to the integrin lymphocyte function-associated antigen-1,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 10, pp. 4095–4100, 2007.
[99]  E. Song, S.-K. Lee, J. Wang et al., “RNA interference targeting Fas protects mice from fulminant hepatitis,” Nature Medicine, vol. 9, no. 3, pp. 347–351, 2003.
[100]  P. Hamar, E. Song, G. K?keny, A. Chen, N. Ouyang, and J. Lieberman, “Small interfering RNA targeting Fas protects mice against renal ischemia-reperfusion injury,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 41, pp. 14883–14888, 2004.
[101]  D. M. Dykxhoorn, D. Chowdhury, and J. Lieberman, “RNA interference and cancer: endogenous pathways and therapeutic approaches,” Advances in Experimental Medicine and Biology, vol. 615, pp. 299–329, 2008.
[102]  S. I. Pai, Y.-Y. Lin, B. Macaes, A. Meneshian, C.-F. Hung, and T.-C. Wu, “Prospects of RNA interference therapy for cancer,” Gene Therapy, vol. 13, no. 6, pp. 464–477, 2006.
[103]  T. R. Brummelkamp, R. Bernards, and R. Agami, “Stable suppression of tumorigenicity by virus-mediated RNA interference,” Cancer Cell, vol. 2, no. 3, pp. 243–247, 2002.
[104]  D. H. Kim and J. J. Rossi, “RNAi mechanisms and applications,” BioTechniques, vol. 44, no. 5, pp. 613–616, 2008.
[105]  L. A. Wheeler and D. M. Dykxhoorn, “RNAi as a potential new therapy for HIV infection,” Future HIV Therapy, vol. 2, no. 6, pp. 567–577, 2008.
[106]  D. S. Schwarz, G. Hutvágner, T. Du, Z. Xu, N. Aronin, and P. D. Zamore, “Asymmetry in the assembly of the RNAi enzyme complex,” Cell, vol. 115, no. 2, pp. 199–208, 2003.
[107]  C. Matranga, Y. Tomari, C. Shin, D. P. Bartel, and P. D. Zamore, “Passenger-strand cleavage facilitates assembly of siRNA into Ago2-containing RNAi enzyme complexes,” Cell, vol. 123, no. 4, pp. 607–620, 2005.
[108]  D. M. Dykxhoorn and J. Lieberman, “Running interference: prospects and obstacles to using small interfering RNAs as small molecule drugs,” Annual Review of Biomedical Engineering, vol. 8, pp. 377–402, 2006.
[109]  D. P. Bartel, “MicroRNAs: genomics, biogenesis, mechanism, and function,” Cell, vol. 116, no. 2, pp. 281–297, 2004.
[110]  G. Stefani and F. J. Slack, “Small non-coding RNAs in animal development,” Nature Reviews Molecular Cell Biology, vol. 9, no. 3, pp. 219–230, 2008.
[111]  L. Wu and J. G. Belasco, “Let Me count the ways: mechanisms of gene regulation by miRNAs and siRNAs,” Molecular Cell, vol. 29, no. 1, pp. 1–7, 2008.
[112]  A. de Fougerolles, H.-P. Vornlocher, J. Maraganore, and J. Lieberman, “Interfering with disease: a progress report on siRNA-based therapeutics,” Nature Reviews Drug Discovery, vol. 6, no. 6, pp. 443–453, 2007.
[113]  B. Berkhout and O. Ter Brake, “Towards a durable RNAi gene therapy for HIV-AIDS,” Expert Opinion on Biological Therapy, vol. 9, no. 2, pp. 161–170, 2009.
[114]  J. Capodici, K. Karikó, and D. Weissman, “Inhibition of HIV-1 infection by small interfering RNA-mediated RNA interference,” Journal of Immunology, vol. 169, no. 9, pp. 5196–5201, 2002.
[115]  G. A. Coburn and B. R. Cullen, “Potent and specific inhibition of human immunodeficiency virus type 1 replication by RNA interference,” Journal of Virology, vol. 76, no. 18, pp. 9225–9231, 2002.
[116]  J.-M. Jacque, K. Triques, and M. Stevenson, “Modulation of HIV-1 replication by RNA interference,” Nature, vol. 418, no. 6896, pp. 435–438, 2002.
[117]  N. S. Lee, T. Dohjima, G. Bauer et al., “Expression of small interfering RNAs targeted against HIV-1 rev transcripts in human cells,” Nature Biotechnology, vol. 20, no. 5, pp. 500–505, 2002.
[118]  A. L. Brass, D. M. Dykxhoorn, Y. Benita et al., “Identification of host proteins required for HIV infection through a functional genomic screen,” Science, vol. 319, no. 5865, pp. 921–926, 2008.
[119]  Y. Gao, M. A. Lobritz, J. Roth et al., “Targets of small interfering RNA restriction during human immunodeficiency virus type 1 replication,” Journal of Virology, vol. 82, no. 6, pp. 2938–2951, 2008.
[120]  R. Alvarez, S. Elbashir, T. Borland et al., “RNA interference-mediated silencing of the respiratory syncytial virus nucleocapsid defines a potent antiviral strategy,” Antimicrobial Agents and Chemotherapy, vol. 53, no. 9, pp. 3952–3962, 2009.
[121]  W. Zhang, H. Yang, X. Kong et al., “Inhibition of respiratory syncytial virus infection with intranasal siRNA nanoparticles targeting the viral NS1 gene,” Nature Medicine, vol. 11, no. 1, pp. 56–62, 2005.
[122]  L. A. Wheeler, V. Vrbanac, R. Trifonova et al., “Durable knockdown and protection from HIV transmission in humanized mice treated with gel-formulated CD4 aptamer-siRNA chimeras,” Molecular Therapy, vol. 21, pp. 1378–1389, 2013.
[123]  L. A. Wheeler, R. Trifonova, V. Vrbanac et al., “Inhibition of HIV transmission in human cervicovaginal explants and humanized mice using CD4 aptamer-siRNA chimeras,” Journal of Clinical Investigation, vol. 121, no. 6, pp. 2401–2412, 2011.
[124]  R. M. Grant, D. Hamer, T. Hope et al., “Whither or wither microbicides?” Science, vol. 321, no. 5888, pp. 532–534, 2008.
[125]  J. Yano, K. Hirabayashi, S.-I. Nakagawa et al., “Antitumor activity of small interfering RNA/cationic liposome complex in mouse models of cancer,” Clinical Cancer Research, vol. 10, no. 22, pp. 7721–7726, 2004.
[126]  J. Anderson, A. Banerjea, V. Planelles, and R. Akkina, “Potent suppression of HIV type 1 infection by a short hairpin anti-CXCR4 siRNA,” AIDS Research and Human Retroviruses, vol. 19, no. 8, pp. 699–706, 2003.
[127]  E. Devroe and P. A. Silver, “Therapeutic potential of retroviral RNAi vectors,” Expert Opinion on Biological Therapy, vol. 4, no. 3, pp. 319–327, 2004.
[128]  D. L. Lewis and J. A. Wolff, “Delivery of siRNA and siRNA expression constructs to adult mammals by hydrodynamic intravascular injection,” Methods in Enzymology, vol. 392, pp. 336–350, 2005.
[129]  C. Lorenz, P. Hadwiger, M. John, H.-P. Vornlocher, and C. Unverzagt, “Steroid and lipid conjugates of siRNAs to enhance cellular uptake and gene silencing in liver cells,” Bioorganic and Medicinal Chemistry Letters, vol. 14, no. 19, pp. 4975–4977, 2004.
[130]  Y. Minakuchi, F. Takeshita, N. Kosaka et al., “Atelocollagen-mediated synthetic small interfering RNA delivery for effective gene silencing in vitro and in vivo,” Nucleic Acids Research, vol. 32, no. 13, p. e109, 2004.
[131]  J. Soutschek, A. Akinc, B. Bramlage et al., “Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs,” Nature, vol. 432, no. 7014, pp. 173–178, 2004.
[132]  B. Urban-Klein, S. Werth, S. Abuharbeid, F. Czubayko, and A. Aigner, “RNAi-mediated gene-targeting through systemic application of polyethylenimine (PEI)-complexed siRNA in vivo,” Gene Therapy, vol. 12, no. 5, pp. 461–466, 2005.
[133]  R. A. Stull and F. C. Szoka Jr., “Antigene, ribozyme and aptamer nucleic acid drugs: progress and prospects,” Pharmaceutical Research, vol. 12, no. 4, pp. 465–483, 1995.
[134]  L. Scherer, J. J. Rossi, and M. S. Weinberg, “Progress and prospects: RNA-based therapies for treatment of HIV infection,” Gene Therapy, vol. 14, no. 14, pp. 1057–1064, 2007.
[135]  D. M. Held, J. D. Kissel, J. T. Patterson, D. G. Nickens, and D. H. Burke, “HIV-1 inactivation by nucleic acid aptamers,” Frontiers in Bioscience, vol. 11, no. 1, pp. 89–112, 2006.
[136]  J. O. McNamara II, E. R. Andrechek, Y. Wang et al., “Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras,” Nature Biotechnology, vol. 24, no. 8, pp. 1005–1015, 2006.
[137]  J. P. Dassie, X.-Y. Liu, G. S. Thomas et al., “Systemic administration of optimized aptamer-siRNA chimeras promotes regression of PSMA-expressing tumors,” Nature Biotechnology, vol. 27, no. 9, pp. 839–846, 2009.
[138]  J. Zhou, H. Li, S. Li, J. Zaia, and J. J. Rossi, “Novel dual inhibitory function aptamer-siRNA delivery system for HIV-1 therapy,” Molecular Therapy, vol. 16, no. 8, pp. 1481–1489, 2008.
[139]  J. Zhou, P. Swiderski, H. Li et al., “Selection, characterization and application of new RNA HIV gp 120 aptamers for facile delivery of Dicer substrate siRNAs into HIV infected cells,” Nucleic Acids Research, vol. 37, no. 9, pp. 3094–3109, 2009.
[140]  C. P. Neff, J. Zhou, L. Remling et al., “An aptamer-siRNA chimera suppresses HIV-1 viral loads and protects from helper CD4+ T cell decline in humanized mice,” Science Translational Medicine, vol. 3, no. 66, Article ID 66ra6, 2011.
[141]  D. M. Brainard, E. Seung, N. Frahm et al., “Induction of robust cellular and humoral virus-specific adaptive immune responses in human immunodeficiency virus-infected humanized BLT mice,” Journal of Virology, vol. 83, no. 14, pp. 7305–7321, 2009.
[142]  J. Zhou, H. Li, J. Zhang, S. Piotr, and J. Rossi, “Development of cell-type specific anti-HIV gp120 aptamers for siRNA delivery,” Journal of Visualized Experiments, no. 52, Article ID e2954, 2011.
[143]  OPKO Health, “OPKO Health announces update on Phase III clinical trial of bevasiranib,” Press release.
[144]  M. Robbins, A. Judge, and I. MacLachlan, “SiRNA and innate immunity,” Oligonucleotides, vol. 19, no. 2, pp. 89–102, 2009.
[145]  J. A. Katakowski and D. Palliser, “SiRNA-based topical microbicides targeting sexually transmitted infections,” Current Opinion in Molecular Therapeutics, vol. 12, no. 2, pp. 192–202, 2010.
[146]  V. Hornung, M. Guenthner-Biller, C. Bourquin et al., “Sequence-specific potent induction of IFN-α by short interfering RNA in plasmacytoid dendritic cells through TLR7,” Nature Medicine, vol. 11, no. 3, pp. 263–270, 2005.
[147]  A. D. Judge, V. Sood, J. R. Shaw, D. Fang, K. McClintock, and I. MacLachlan, “Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA,” Nature Biotechnology, vol. 23, no. 4, pp. 457–462, 2005.
[148]  M. Robbins, A. Judge, E. Ambegia et al., “Misinterpreting the therapeutic effects of small interfering RNA caused by immune stimulation,” Human Gene Therapy, vol. 19, no. 10, pp. 991–999, 2008.
[149]  M. Schlee, V. Hornung, and G. Hartmann, “siRNA and isRNA: two edges of one sword,” Molecular Therapy, vol. 14, no. 4, pp. 463–470, 2006.
[150]  H. Poeck, R. Besch, C. Maihoefer et al., “5′-triphosphate-siRNA: turning gene silencing and Rig-I activation against melanoma,” Nature Medicine, vol. 14, no. 11, pp. 1256–1263, 2008.
[151]  A. L. Jackson, J. Burchard, D. Leake et al., “Position-specific chemical modification of siRNAs reduces “off-target” transcript silencing,” RNA, vol. 12, no. 7, pp. 1197–1205, 2006.
[152]  K. Ui-Tei, Y. Naito, S. Zenno et al., “Functional dissection of siRNA sequence by systematic DNA substitution: modified siRNA with a DNA seed arm is a powerful tool for mammalian gene silencing with significantly reduced off-target effect,” Nucleic Acids Research, vol. 36, no. 7, pp. 2136–2151, 2008.
[153]  M. B. Laursen, M. M. Pakula, S. Gao et al., “Utilization of unlocked nucleic acid (UNA) to enhance siRNA performance in vitro and in vivo,” Molecular BioSystems, vol. 6, no. 5, pp. 862–870, 2010.
[154]  Y.-L. Chiu and T. M. Rana, “RNAi in human cells: basic structural and functional features of small interfering RNA,” Molecular Cell, vol. 10, no. 3, pp. 549–561, 2002.
[155]  M. E. Kleinman, K. Yamada, A. Takeda et al., “Sequence- and target-independent angiogenesis suppression by siRNA via TLR3,” Nature, vol. 452, no. 7187, pp. 591–597, 2008.
[156]  T. S. Zimmermann, A. C. H. Lee, A. Akinc et al., “RNAi-mediated gene silencing in non-human primates,” Nature, vol. 441, no. 1, pp. 111–114, 2006.
[157]  A. A. Khan, D. Betel, M. L. Miller, C. Sander, C. S. Leslie, and D. S. Marks, “Transfection of small RNAs globally perturbs gene regulation by endogenous microRNAs,” Nature Biotechnology, vol. 27, no. 6, pp. 549–555, 2009.
[158]  D. Grimm, K. L. Streetz, C. L. Jopling et al., “Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways,” Nature, vol. 441, no. 7092, pp. 537–541, 2006.
[159]  M. John, R. Constien, A. Akinc et al., “Effective RNAi-mediated gene silencing without interruption of the endogenous microRNA pathway,” Nature, vol. 449, no. 7163, pp. 745–747, 2007.
[160]  J. L. McBride, R. L. Boudreau, S. Q. Harper et al., “Artificial miRNAs mitigate shRNA-mediated toxicity in the brain: implications for the therapeutic development of RNAi,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 15, pp. 5868–5873, 2008.
[161]  A. D. Judge, G. Bola, A. C. H. Lee, and I. MacLachlan, “Design of noninflammatory synthetic siRNA mediating potent gene silencing in vivo,” Molecular Therapy, vol. 13, no. 3, pp. 494–505, 2006.
[162]  D. Castanotto, K. Sakurai, R. Lingeman et al., “Combinatorial delivery of small interfering RNAs reduces RNAi efficacy by selective incorporation into RISC,” Nucleic Acids Research, vol. 35, no. 15, pp. 5154–5164, 2007.
[163]  A. R. de Fougerolles, “Delivery vehicles for small interfering RNA in vivo,” Human Gene Therapy, vol. 19, no. 2, pp. 125–132, 2008.
[164]  B. L. Davidson and P. B. McCray Jr., “Current prospects for RNA interference-based therapies,” Nature Reviews Genetics, vol. 12, no. 5, pp. 329–340, 2011.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133