Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-17T15:06:52.884Z Has data issue: false hasContentIssue false

GENE THERAPY FOR OBSTETRIC CONDITIONS

Published online by Cambridge University Press:  08 June 2015

REBECCA N. SPENCER*
Affiliation:
Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK.
DAVID J. CARR
Affiliation:
Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK.
ANNA L. DAVID
Affiliation:
Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK.
*
Rebecca N. Spencer, MRCOG, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK. Email: rebecca.spencer@ucl.ac.uk

Extract

The first clinical trials of gene therapy in the 1990s offered the promise of a new paradigm for the treatment of genetic diseases. Over the decades that followed the challenges and setbacks which gene therapy faced often overshadowed any successes. Despite this, recent years have seen cause for renewed optimism. In 2012 Glybera™, an adeno-associated viral vector expressing lipoprotein lipase, became the first gene therapy product to receive marketing authorisation in Europe, with a licence to treat familial lipoprotein lipase deficiency. This followed the earlier licensing in China of two gene therapies: Gendicine™ for head and neck squamous cell carcinoma and Oncorine™ for late-stage nasopharyngeal cancer. By this stage over 1800 clinical trials had been, or were being, conducted worldwide, and the therapeutic targets had expanded far beyond purely genetic disorders. So far no trials of gene therapy have been carried out in pregnancy, but an increasing understanding of the molecular mechanisms underlying obstetric diseases means that it is likely to have a role to play in the future. This review will discuss how gene therapy works, its potential application in obstetric conditions and the risks and limitations associated with its use in this setting. It will also address the ethical and regulatory issues that will be faced by any potential clinical trial of gene therapy during pregnancy.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2015 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

1. Wirth, T, Parker, N, Yla-Herttuala, S. History of gene therapy. Gene 2013; 525 (2): 162–9.Google Scholar
2. Ginn, SL, Alexander, IE, Edelstein, ML, Abedi, MR, Wixon, J. Gene therapy clinical trials worldwide to 2012 - an update. J Gene Med 2013; 15 (2): 6577.Google Scholar
3. Leader, B, Baca, QJ, Golan, DE. Protein therapeutics: a summary and pharmacological classification. Nat Rev Drug Discov 2008; 7 (1): 2139.Google Scholar
4. Yamamoto, S, Konishi, I, Mandai, M, Kuroda, H, Komatsu, T, Nanbu, K, et al. Expression of vascular endothelial growth factor (VEGF) in epithelial ovarian neoplasms: correlation with clinicopathology and patient survival, and analysis of serum VEGF levels. Br J Cancer 1997; 76 (9): 1221–7.Google Scholar
5. Tuppurainen, L, Sallinen, H, Kokki, E, Koponen, J, Anttila, M, Pulkkinen, K, et al. Preclinical safety, toxicology, and biodistribution study of adenoviral gene therapy with sVEGFR-2 and sVEGFR-3 combined with chemotherapy for ovarian cancer. Hum Gene Ther Clin Dev 2013; 24 (1): 2937.Google Scholar
6. Hassan, M, Zhang, D, Salama, S, Hamada, F, Arafa, H, Fouad, H, et al. Towards fibroid gene therapy: adenovirus-mediated delivery of herpes simplex virus 1 thymidine kinase gene/ganciclovir shrinks uterine leiomyoma in the Eker rat model. Gynecol Obstet Invest 2009; 68 (1): 1932.Google Scholar
7. David, AL, Peebles, D. Gene therapy for the fetus: is there a future? Best Pract Res Clin Obstet Gynaecol 2008; 22 (1): 203–18.Google Scholar
8. Coutelle, C, Waddington, SN. Vector systems for prenatal gene therapy: choosing vectors for different applications. Methods Mol Biol 2012; 891: 4153.Google Scholar
9. Al-Hendy, A, Salama, S. Gene therapy and uterine leiomyoma: a review. Hum Reprod Update 2006; 12 (4): 385400.Google Scholar
10. Stribley, JM, Rehman, KS, Niu, H, Christman, GM. Gene therapy and reproductive medicine. Fertil Steril 2002; 77 (4): 645–57.Google Scholar
11. Kaeppel, C, Beattie, SG, Fronza, R, van Logtenstein, R, Salmon, F, Schmidt, S, et al. A largely random AAV integration profile after LPLD gene therapy. Nat Med 2013; 19 (7): 889–91.Google Scholar
12. Raper, SE, Chirmule, N, Lee, FS, Wivel, NA, Bagg, A, Gao, GP, et al. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab 2003; 80 (1–2): 148–58.CrossRefGoogle Scholar
13. Wilson, JM. Lessons learned from the gene therapy trial for ornithine transcarbamylase deficiency. Mol Genet Metab 2009; 96 (4): 151–7.Google Scholar
14. Mattar, CN, Waddington, SN, Biswas, A, Davidoff, AM, Choolani, M, Chan, JKY, et al. The case for intrauterine gene therapy. Best Pract Res Clin Obstet Gynaecol 2012; 26 (5): 697709.Google Scholar
15. Wiznerowicz, M, Trono, D. Harnessing HIV for therapy, basic research and biotechnology. Trends Biotechnol 2005; 23 (1): 42–7.Google Scholar
16. Nowrouzi, A, Cheung, WT, Li, T, Zhang, X, Arens, A, Paruzynski, A, et al. The fetal mouse is a sensitive genotoxicity model that exposes lentiviral-associated mutagenesis resulting in liver oncogenesis. Mol Ther 2013; 21 (2): 324–37.Google Scholar
17. Mukherjee, S, Thrasher, AJ. Gene therapy for PIDs: progress, pitfalls and prospects. Gene 2013; 525 (2): 174–81.Google Scholar
18. Fischer, A, Cavazzana-Calvo, M. Gene therapy of inherited diseases. Lancet 2008; 371 (9629): 2044–7.Google Scholar
19. Gene Therapy Advisory Committee. Report on the potential use of gene therapy in utero. Health Departments of the United Kingdom, November 1998. Hum Gene Ther 1999; 10 (4): 689–92.Google Scholar
20. Tachibana, M, Amato, P, Sparman, M, Woodward, J, Sanchis, DM, Ma, H, et al. Towards germline gene therapy of inherited mitochondrial diseases. Nature 2013; 493 (7434): 627–31.Google Scholar
21. Human Fertilisation and Embryology Authority. Statement on mitochondrial donation. 2015. Available from: www.hfea.gov.uk/9606.html.Google Scholar
22. Touzot, F, Hacein-Bey-Abina, S, Fischer, A, Cavazzana, M. Gene therapy for inherited immunodeficiency. Expert Opin Biol Ther 2014; 14 (6): 789–98.Google Scholar
23. Pipino, C, Shangaris, P, Resca, E, Zia, S, Deprest, J, Sebire, NJ, et al. Placenta as a reservoir of stem cells: an underutilized resource? Br Med Bull 2013; 105: 4368.Google Scholar
24. Shaw, SW, Bollini, S, Nader, KA, Gastadello, A, Mehta, V, Filppi, E, et al. Autologous transplantation of amniotic fluid-derived mesenchymal stem cells into sheep fetuses. Cell Transplant 2011; 20 (7): 1015–31.Google Scholar
25. Shaw, SW, David, AL, De Coppi, P. Clinical applications of prenatal and postnatal therapy using stem cells retrieved from amniotic fluid. Curr Opin Obstet Gynecol 2011; 23 (2): 109–16.Google Scholar
26. De Coppi, P, Bartsch, G Jr., Siddiqui, MM, Xu, T, Santos, CC, Perin, L, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 2007; 25 (1): 100–6.Google Scholar
27. Ditadi, A, de Coppi, P, Picone, O, Gautreau, L, Smati, R, Six, E, et al. Human and murine amniotic fluid c-Kit+Lin- cells display hematopoietic activity. Blood 2009; 113 (17): 3953–60.Google Scholar
28. de Silva, S, Bowers, WJ. Targeting the central nervous system with herpes simplex virus/sleeping beauty hybrid amplicon vectors. Curr Gene Ther 2011; 11 (5): 332–40.Google Scholar
29. Katayama, K, Furuki, R, Yokoyama, H, Kaneko, M, Tachibana, M, Yoshida, I, et al. Enhanced in vivo gene transfer into the placenta using RGD fiber-mutant adenovirus vector. Biomaterials 2011; 32 (17): 4185–93.Google Scholar
30. Othman, E-ER, Zhu, ZB, Curiel, DT, Khatoon, N, Salem, HT, Khalifa, EA-DM, et al. Toward gene therapy of endometriosis: transductional and transcriptional targeting of adenoviral vectors to endometriosis cells. Am J Obstet Gynecol 2008; 199 (2): 117.e1–6.CrossRefGoogle ScholarPubMed
31. Nair, S, Curiel, DT, Rajaratnam, V, Thota, C, Al-Hendy, A. Targeting adenoviral vectors for enhanced gene therapy of uterine leiomyomas. Hum Reprod 2013; 28 (9): 2398–406.Google Scholar
32. Turkay, A, Saunders, T, Kurachi, K. Intrauterine gene transfer: gestational stage-specific gene delivery in mice. Gene Ther 1999; 6 (10): 1685–94.Google Scholar
33. Xing, A, Boileau, P, Cauzac, M, Challier, JC, Girard, J, Hauguel-de Mouzon, S. Comparative in vivo approaches for selective adenovirus-mediated gene delivery to the placenta. Hum Gene Ther 2000; 11 (1): 167–77.Google Scholar
34. Senoo, M, Matsubara, Y, Fujii, K, Nagasaki, Y, Hiratsuka, M, Kure, S, et al. Adenovirus-mediated in utero gene transfer in mice and guinea pigs: tissue distribution of recombinant adenovirus determined by quantitative TaqMan-polymerase chain reaction assay. Mol Genet Metab 2000; 69 (4): 269–76.Google Scholar
35. Katz, AB, Keswani, SG, Habli, M, Lim, FY, Zoltick, PW, Midrio, P, et al. Placental gene transfer: transgene screening in mice for trophic effects on the placenta. Am J Obstet Gynecol 2009; 201 (5): 499.e1–8.Google Scholar
36. Woo, YJ, Raju, GP, Swain, JL, Richmond, ME, Gardner, TJ, Balice-Gordon, RJ. In utero cardiac gene transfer via intraplacental delivery of recombinant adenovirus. Circulation 1997; 96 (10): 3561–9.Google Scholar
37. Heikkila, A, Hiltunen, MO, Turunen, MP, Keski-Nisula, L, Turunen, AM, Rasanen, H, et al. Angiographically guided utero-placental gene transfer in rabbits with adenoviruses, plasmid/liposomes and plasmid/polyethyleneimine complexes. Gene Ther 2001; 8 (10): 784–8.Google Scholar
38. Mehta, V, Peebles, DM, Boyd, M, Zachary, I, Martin, J, David, AL. Gene Targeting to the Utero-Placental Circulation of Pregnant Guinea Pigs. Society for Gynaecologic Investigation 58th Annual Meeting: Reproductive Sciences, 2011; 332A.Google Scholar
39. Buckley, SM, Rahim, AA, Chan, JK, David, AL, Peebles, DM, Coutelle, C, et al. Recent advances in fetal gene therapy. Ther Deliv 2011; 2 (4): 461–9.CrossRefGoogle ScholarPubMed
40. David, AL, Waddington, SN. Candidate diseases for prenatal gene therapy. Methods Mol Biol 2012; 891: 939.Google Scholar
41. Mehta, V, Abi Nader, K, Waddington, S, David, AL. Organ targeted prenatal gene therapy–how far are we? Prenat Diagn 2011; 31 (7): 720–34.Google Scholar
42. Waddington, SN, Nivsarkar, MS, Mistry, AR, Buckley, SM, Kemball-Cook, G, Mosley, KL, et al. Permanent phenotypic correction of hemophilia B in immunocompetent mice by prenatal gene therapy. Blood 2004; 104 (9): 2714–21.Google Scholar
43. Rucker, M, Fraites, TJ Jr., Porvasnik, SL, Lewis, MA, Zolotukhin, I, Cloutier, DA, et al. Rescue of enzyme deficiency in embryonic diaphragm in a mouse model of metabolic myopathy: pompe disease. Development 2004; 131 (12): 3007–19.Google Scholar
44. Seppen, J, van der Rijt, R, Looije, N, van Til, NP, Lamers, WH, Oude Elferink, RP. Long-term correction of bilirubin UDPglucuronyltransferase deficiency in rats by in utero lentiviral gene transfer. Mol Ther 2003; 8 (4): 593–9.Google Scholar
45. Koppanati, BM, Li, J, Reay, DP, Wang, B, Daood, M, Zheng, H, et al. Improvement of the mdx mouse dystrophic phenotype by systemic in utero AAV8 delivery of a minidystrophin gene. Gene Ther 2010; 17 (11): 1355–62.Google Scholar
46. Dejneka, NS, Surace, EM, Aleman, TS, Cideciyan, AV, Lyubarsky, A, Savchenko, A, et al. In utero gene therapy rescues vision in a murine model of congenital blindness. Mol Ther 2004; 9 (2): 182–8.Google Scholar
47. Williams, ML, Coleman, JE, Haire, SE, Aleman, TS, Cideciyan, AV, Sokal, I, et al. Lentiviral expression of retinal guanylate cyclase-1 (RetGC1) restores vision in an avian model of childhood blindness. PLoS Med 2006; 3 (6): e201.Google Scholar
48. Wu, C, Endo, M, Yang, BH, Radecki, MA, Davis, PF, Zoltick, PW, et al. Intra-amniotic transient transduction of the periderm with a viral vector encoding TGFbeta3 prevents cleft palate in Tgfbeta3(-/-) mouse embryos. Mol Ther 2013; 21 (1): 817.Google Scholar
49. David, AL, McIntosh, J, Peebles, DM, Cook, T, Waddington, S, Weisz, B, et al. Recombinant adeno-associated virus-mediated in utero gene transfer gives therapeutic transgene expression in the sheep. Hum Gene Ther 2011; 22 (4): 419–26.Google Scholar
50. Mattar, CN, Nathwani, AC, Waddington, SN, Dighe, N, Kaeppel, C, Nowrouzi, A, et al. Stable human FIX expression after 0.9G intrauterine gene transfer of self-complementary adeno-associated viral vector 5 and 8 in macaques. Mol Ther 2011; 19 (11): 1950–60.Google Scholar
51. Mattar, CN, Waddington, SN, Biswas, A, Johana, N, Ng, XW, Fisk, AS, et al. Systemic delivery of scAAV9 in fetal macaques facilitates neuronal transduction of the central and peripheral nervous systems. Gene Ther 2013; 20 (1): 6983.Google Scholar
52. U. S. National Institutes of Health. Recombinant DNA Advisory Committee. Prenatal gene tranfer: scientific, medical, and ethical issues: a report of the Recombinant DNA Advisory Committee. Hum Gene Ther 2000; 11 (8): 1211–29.Google Scholar
53. Ramachandra, DL, Shaw, SS, Shangaris, P, Loukogeorgakis, S, Guillot, PV, Coppi, PD, et al. In utero therapy for congenital disorders using amniotic fluid stem cells. Front Pharmacol 2014; 5: 270.Google Scholar
54. Tiblad, E, Westgren, M. Fetal stem-cell transplantation. Best Pract Res Clin Obstet Gynaecol 2008; 22 (1): 189201.Google Scholar
55. Shaw, SW, Blundell, MP, Pipino, C, Shangaris, P, Maghsoudlou, P, Ramachandra, DL, et al. Sheep CD34+ amniotic fluid cells have hematopoietic potential and engraft after autologous in utero transplantation. Stem Cells 2015; 33 (1): 122–32.Google Scholar
56. Flenady, V, Koopmans, L, Middleton, P, Froen, JF, Smith, GC, Gibbons, K, et al. Major risk factors for stillbirth in high-income countries: a systematic review and meta-analysis. Lancet 2011; 377 (9774): 1331–40.Google Scholar
57. Marlow, N, Wolke, D, Bracewell, MA, Samara, M. Neurologic and developmental disability at six years of age after extremely preterm birth. N Engl J Med 2005; 352 (1): 919.Google Scholar
58. Baschat, AA, Cosmi, E, Bilardo, CM, Wolf, H, Berg, C, Rigano, S, et al. Predictors of neonatal outcome in early-onset placental dysfunction. Obstet Gynecol 2007; 109 (2 Pt 1): 253–61.Google Scholar
59. Lyall, F, Robson, SC, Bulmer, JN. Spiral artery remodeling and trophoblast invasion in preeclampsia and fetal growth restriction: relationship to clinical outcome. Hypertension 2013; 62 (6): 1046–54.Google Scholar
60. Konje, JC, Howarth, ES, Kaufmann, P, Taylor, DJ. Longitudinal quantification of uterine artery blood volume flow changes during gestation in pregnancies complicated by intrauterine growth restriction. BJOG 2003; 110 (3): 301–5.Google Scholar
61. Savvidou, MD, Yu, CK, Harland, LC, Hingorani, AD, Nicolaides, KH. Maternal serum concentration of soluble fms-like tyrosine kinase 1 and vascular endothelial growth factor in women with abnormal uterine artery Doppler and in those with fetal growth restriction. Am J Obstet Gynecol 2006; 195 (6): 1668–73.Google Scholar
62. Nevo, O, Many, A, Xu, J, Kingdom, J, Piccoli, E, Zamudio, S, et al. Placental expression of soluble fms-like tyrosine kinase 1 is increased in singletons and twin pregnancies with intrauterine growth restriction. J Clin Endocrinol Metab 2008; 93 (1): 285–92.Google Scholar
63. David, AL, Torondel, B, Zachary, I, Wigley, V, Abi-Nader, K, Mehta, V, et al. Local delivery of VEGF adenovirus to the uterine artery increases vasorelaxation and uterine blood flow in the pregnant sheep. Gene Ther 2008; 15 (19): 1344–50.Google Scholar
64. Mehta, V, Abi-Nader, KN, Peebles, DM, Benjamin, E, Wigley, V, Torondel, B, et al. Long-term increase in uterine blood flow is achieved by local overexpression of VEGF-A(165) in the uterine arteries of pregnant sheep. Gene Ther 2012; 19 (9): 925–35.Google Scholar
65. Mehta, V, Abi-Nader, KN, Shangaris, P, Shaw, SW, Filippi, E, Benjamin, E, et al. Local over-expression of VEGF-DDeltaNDeltaC in the uterine arteries of pregnant sheep results in long-term changes in uterine artery contractility and angiogenesis. PloS One 2014; 9 (6): e100021.Google Scholar
66. Carr, D, Wallace, JM, Aitken, RP, Milne, JS, Mehta, V, Martin, JF, et al. Uteroplacental adenovirus VEGF gene therapy increases fetal growth velocity in growth-restricted sheep pregnancies. Hum Gene Ther 2014; 25 (4): 375–84.Google Scholar
67. Mehta, V, Boyd, M, Martin, J, Zachary, I, Peebles, DM, David, AL. Local administration of Ad.VEGF-A165 to the uteroplacental circulation enhances fetal growth and reduces brain sparing in an FGR model of guinea pig pregnancy. Reprod Sci 2012; 19 (3): 78A.Google Scholar
68. Olsson, AK, Dimberg, A, Kreuger, J, Claesson-Welsh, L. VEGF receptor signalling - in control of vascular function. Nat Rev Mol Cell Biol 2006; 7 (5): 359–71.Google Scholar
69. Rutanen, JMD, Rissanen, TTMDP, Markkanen, JEMD, Gruchala, MMD, PMD, Silvennoinen, Kivela, AMD, et al. Adenoviral catheter-mediated intramyocardial gene transfer using the mature form of vascular endothelial growth factor-D induces transmural angiogenesis in porcine heart. Circulation 2004; 109 (8): 1029–35.Google Scholar
70. Wallace, JM, Luther, JS, Milne, JS, Aitken, RP, Redmer, DA, Reynolds, LP, et al. Nutritional modulation of adolescent pregnancy outcome – a review. Placenta 2006; 27 (Suppl. A): S618.Google Scholar
71. Robinson, JS, Kingston, EJ, Jones, CT, Thorburn, GD. Studies on experimental growth retardation in sheep. The effect of removal of a endometrial caruncles on fetal size and metabolism. J Dev Physiol 1979; 1 (5): 379–98.Google Scholar
72. Wallace, JM, Aitken, RP, Milne, JS, Hay, WW Jr. Nutritionally mediated placental growth restriction in the growing adolescent: consequences for the fetus. Biol Reprod 2004; 71 (4): 1055–62.Google Scholar
73. Wallace, JM, Milne, JS, Matsuzaki, M, Aitken, RP. Serial measurement of uterine blood flow from mid to late gestation in growth restricted pregnancies induced by overnourishing adolescent sheep dams. Placenta. 2008; 29 (8): 718–24.Google Scholar
74. Wallace, JM, Bourke, DA, Aitken, RP, Palmer, RM, Da Silva, P, Cruickshank, MA. Relationship between nutritionally-mediated placental growth restriction and fetal growth, body composition and endocrine status during late gestation in adolescent sheep. Placenta 2000; 21 (1): 100–8.Google Scholar
75. Carr, DJ, Aitken, RP, Milne, JS, David, AL, Wallace, JM. Fetoplacental biometry and umbilical artery Doppler velocimetry in the overnourished adolescent model of fetal growth restriction. Am J Obstet Gynecol 2012; 207 (2): 141 e6–15.Google Scholar
76. Redmer, DA, Aitken, RP, Milne, JS, Reynolds, LP, Wallace, JM. Influence of maternal nutrition on messenger RNA expression of placental angiogenic factors and their receptors at midgestation in adolescent sheep. Biol Reprod 2005; 72 (4): 1004–9.Google Scholar
77. Redmer, DA, Luther, JS, Milne, JS, Aitken, RP, Johnson, ML, Borowicz, PP, et al. Fetoplacental growth and vascular development in overnourished adolescent sheep at day 50, 90 and 130 of gestation. Reproduction 2009; 137 (4): 749–57.Google Scholar
78. Lea, RG, Wooding, P, Stewart, I, Hannah, LT, Morton, S, Wallace, K, et al. The expression of ovine placental lactogen, StAR and progesterone-associated steroidogenic enzymes in placentae of overnourished growing adolescent ewes. Reproduction 2007; 133 (4): 785–96.Google Scholar
79. Carr, DJ, Aitken, RP, Milne, JS, Peebles, DM, Martin, JM, Zachary, IC, et al. Prenatal Ad.VEGF gene therapy - a promising new treatment for fetal growth restriction. Hum Gene Ther 2011; 22 (10): A128.Google Scholar
80. Carr, DJ, Aitken, RP, Milne, JS, Peebles, DM, Martin, JM, Zachary, IC, et al. Maternal delivery of Ad.VEGF gene therapy increases fetal growth velocity in an ovine paradigm of fetal growth restriction. Reprod Sci 2011; 18 (Suppl. 3): 269A.Google Scholar
81. Carr, DJ, Aitken, RP, Milne, JS, Peebles, DM, Martin, JM, Zachary, IC, et al. Alterations in postnatal growth and metabolism following prenatal treatment of intrauterine growth restriction with Ad.VEGF gene therapy in the sheep. Arch Dis Child Fetal Neonatal Ed 2011; 96: Fa7.Google Scholar
82. Roberts, CT, Sohlstrom, A, Kind, KL, Earl, RA, Khong, TY, Robinson, JS, et al. Maternal food restriction reduces the exchange surface area and increases the barrier thickness of the placenta in the guinea-pig. Placenta 2001; 22 (2–3): 177–85.Google Scholar
83. Carter, AM. Animal models of human placentation–a review. Placenta 2007; 28 (Suppl. A): S417.Google Scholar
84. Mess, A. The Guinea pig placenta: model of placental growth dynamics. Placenta 2007; 28 (8–9): 812–5.Google Scholar
85. EVERREST Consortium. EVERREST: maternal growth factor therapy to improve fetal growth. 2014. Available from: www.everrest-fp7.eu.Google Scholar
86. Forbes, K, Westwood, M. The IGF axis and placental function. a mini review. Horm Res 2008; 69 (3): 129–37.Google Scholar
87. Eremia, SC, de Boo, HA, Bloomfield, FH, Oliver, MH, Harding, JE. Fetal and amniotic insulin-like growth factor-I supplements improve growth rate in intrauterine growth restriction fetal sheep. Endocrinology 2007; 148 (6): 2963–72.Google Scholar
88. Wali, JA, de Boo, HA, Derraik, JG, Phua, HH, Oliver, MH, Bloomfield, FH, et al. Weekly intra-amniotic IGF-1 treatment increases growth of growth-restricted ovine fetuses and up-regulates placental amino acid transporters. PloS One 2012; 7 (5): e37899.Google Scholar
89. Miller, AG, Aplin, JD, Westwood, M. Adenovirally mediated expression of insulin-like growth factors enhances the function of first trimester placental fibroblasts. J Clin Endocrinol Metab 2005; 90 (1): 379–85.Google Scholar
90. Jones, H, Crombleholme, T, Habli, M. Regulation of amino acid transporters by adenoviral-mediated human insulin-like growth factor-1 in a mouse model of placental insufficiency in vivo and the human trophoblast line BeWo in vitro. Placenta 2014; 35 (2): 132–8.Google Scholar
91. Jones, HN, Crombleholme, T, Habli, M. Adenoviral-mediated placental gene transfer of IGF-1 corrects placental insufficiency via enhanced placental glucose transport mechanisms. PloS One 2013; 8 (9): e74632.Google Scholar
92. Keswani, SG, Balaji, S, Katz, AB, King, A, Omar, K, Habli, M, et al. Intraplacental gene therapy with Ad-IGF-1 corrects naturally occurring rabbit model of intrauterine growth restriction. Hum Gene Ther 2015; 26 (3): 172–82.Google Scholar
93. Habli, M, Jones, H, Aronow, B, Omar, K, Crombleholme, TM. Recapitulation of characteristics of human placental vascular insufficiency in a novel mouse model. Placenta 2013; 34 (12): 1150–8.Google Scholar
94. Steegers, EA, von Dadelszen, P, Duvekot, JJ, Pijnenborg, R. Pre-eclampsia. Lancet 2010; 376 (9741): 631–44.Google Scholar
95. Foidart, JM, Schaaps, JP, Chantraine, F, Munaut, C, Lorquet, S. Dysregulation of anti-angiogenic agents (sFlt-1, PLGF, and sEndoglin) in preeclampsia–a step forward but not the definitive answer. J Reprod Immunol 2009; 82 (2): 106–11.Google Scholar
96. George, EM, Granger, JP. Mechanisms and potential therapies for preeclampsia. Curr Hypertens Rep 2011; 13 (4): 269–75.Google Scholar
97. Maynard, SE, Min, JY, Merchan, J, Lim, KH, Li, J, Mondal, S, et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest 2003; 111 (5): 649–58.Google Scholar
98. Bridges, JP, Gilbert, JS, Colson, D, Gilbert, SA, Dukes, MP, Ryan, MJ, et al. Oxidative stress contributes to soluble fms-like tyrosine kinase-1 induced vascular dysfunction in pregnant rats. Am J Hypertens 2009; 22 (5): 564–8.Google Scholar
99. Bergmann, A, Ahmad, S, Cudmore, M, Gruber, AD, Wittschen, P, Lindenmaier, W, et al. Reduction of circulating soluble Flt-1 alleviates preeclampsia-like symptoms in a mouse model. J Cell Mol Med 2010; 14 (6B): 1857–67.Google Scholar
100. Koyama, S, Kimura, T, Ogita, K, Nakamura, H, Khan, MA, Yoshida, S, et al. Transient local overexpression of human vascular endothelial growth factor (VEGF) in mouse feto-maternal interface during mid-term pregnancy lowers systemic maternal blood pressure. Horm Metab Res 2006; 38 (10): 619–24.Google Scholar
101. Woods, AK, Hoffmann, DS, Weydert, CJ, Butler, SD, Zhou, Y, Sharma, RV, et al. Adenoviral delivery of VEGF121 early in pregnancy prevents spontaneous development of preeclampsia in BPH/5 mice. Hypertension 2011; 57 (1): 94102.Google Scholar
102. Zenclussen, ML, Anegon, I, Bertoja, AZ, Chauveau, C, Vogt, K, Gerlof, K, et al. Over-expression of heme oxygenase-1 by adenoviral gene transfer improves pregnancy outcome in a murine model of abortion. J Reprod Immunol 2006; 69 (1): 3552.Google Scholar
103. Marquez-Rodas, I, Longo, F, Rothlin, RP, Balfagon, G. Pathophysiology and therapeutic possibilities of calcitonin gene-related peptide in hypertension. J Physiol Biochem 2006; 62 (1): 4556.Google Scholar
104. Romero, R, Espinoza, J, Kusanovic, JP, Gotsch, F, Hassan, S, Erez, O, et al. The preterm parturition syndrome. BJOG 2006; 113 (Suppl. 3): 1742.Google Scholar
105. Rein, DT, Schmidt, T, Bauerschmitz, G, Hampl, M, Beyer, IM, Paupoo, AAV, et al. Treatment of endometriosis with a VEGF-targeted conditionally replicative adenovirus. Fertil Steril 2010; 93 (8): 2687–94.Google Scholar
106. Ghadami, M, Salama, SA, Khatoon, N, Chilvers, R, Nagamani, M, Chedrese, PJ, et al. Toward gene therapy of primary ovarian failure: adenovirus expressing human FSH receptor corrects the Finnish C566T mutation. Mol Hum Reprod 2008; 14 (1): 915.Google Scholar
107. Li, W, Li, B, Fan, W, Geng, L, Li, X, Li, L, et al. CTLA4Ig gene transfer alleviates abortion in mice by expanding CD4+CD25+ regulatory T cells and inducing indoleamine 2,3-dioxygenase. J Reprod Immunol 2009; 80 (1–2): 111.Google Scholar
108. Hassan, MH, Salama, SA, Zhang, D, Arafa, HMM, Hamada, FMA, Fouad, H, et al. Gene therapy targeting leiomyoma: adenovirus-mediated delivery of dominant-negative estrogen receptor gene shrinks uterine tumors in Eker rat model. Fertil Steril 2010; 93 (1): 239–50.Google Scholar
109. Li, W, Li, B, Li, S. Adenovirus mediated CTLA4Ig transgene therapy alleviates abortion by inhibiting spleen lymphocyte proliferation and regulating apoptosis in the feto-placental unit. J Reprod Immunol 2013; 97 (2): 167–74.Google Scholar
110. Epperly, MW, Smith, T, Zhang, X, Goff, JP, Franicola, D, Greenberger, B, et al. Modulation of in utero total body irradiation induced newborn mouse growth retardation by maternal manganese superoxide dismutase-plasmid liposome (MnSOD-PL) gene therapy. Gene Ther 2011; 18 (6): 579–83.Google Scholar
111. Khare, R, Chen, CY, Weaver, EA, Barry, MA. Advances and future challenges in adenoviral vector pharmacology and targeting. Curr Gene Ther 2011; 11 (4): 241–58.Google Scholar
112. Gonzaga, S, Henriques-Coelho, T, Davey, M, Zoltick, PW, Leite-Moreira, AF, Correia-Pinto, J, et al. Cystic adenomatoid malformations are induced by localized FGF10 overexpression in fetal rat lung. Am J Respir Cell Mol Biol 2008; 39 (3): 346–55.Google Scholar
113. Tarantal, AF, Chen, H, Shi, TT, Lu, CH, Fang, AB, Buckley, S, et al. Overexpression of transforming growth factor-beta1 in fetal monkey lung results in prenatal pulmonary fibrosis. Eur Respir J 2010; 36 (4): 907–14.Google Scholar
114. Baschat, AA, Towbin, J, Bowles, NE, Harman, CR, Weiner, CP. Prevalence of viral DNA in amniotic fluid of low-risk pregnancies in the second trimester. J Matern Fetal Neonatal Med 2003; 13 (6): 381–4.Google Scholar
115. Koyama, S, Kimura, T, Ogita, K, Nakamura, H, Tabata, C, Md Abu Hadi Noor Ali, K, et al. Simple and highly efficient method for transient in vivo gene transfer to mid-late pregnant mouse uterus. J Reprod Immunol 2006; 70 (1–2): 5969.Google Scholar
116. Desforges, M, David, AL, Greenwood, S, Sibley, CP, Brownbill, P. In Vitro Human Placental Studies To Support an Adenovirus-Mediated VEGF-DΔNΔC Gene Therapy Treatment of Severe Fetal Growth Restriction. Society for Gynecologic Investigation 59th Annual Meeting: Reproductive Sciences; 2012; 147A.Google Scholar
117. Park, PJ, Colletti, E, Ozturk, F, Wood, JA, Tellez, J, Almeida-Porada, G, et al. Factors determining the risk of inadvertent retroviral transduction of male germ cells after in utero gene transfer in sheep. Hum Gene Ther 2009; 20 (3): 201–15.Google Scholar
118. Lee, CC, Jimenez, DF, Kohn, DB, Tarantal, AF. Fetal gene transfer using lentiviral vectors and the potential for germ cell transduction in rhesus monkeys (Macaca mulatta). Hum Gene Ther 2005; 16 (4): 417–25.Google Scholar
119. McCullough, LB, Coverdale, JH, Chervenak, FA. A comprehensive ethical framework for responsibly designing and conducting pharmacologic research that involves pregnant women. Am J Obstet Gynecol 2005; 193 (3 Pt 2): 901–7.Google Scholar
120. Lyerly, AD, Little, MO, Faden, RR. Pregnancy and clinical research. Hastings Cent Rep 2008; 38 (6): c3.Google Scholar
121. Wild, V. How are pregnant women vulnerable research participants? Int J Fem Approaches Bioeth 2012; 5 (2): 82104.Google Scholar
122. Protection of Human Subjects (45 CFR 46), CFR(2009).Google Scholar
123. General Medical Council. Consent: patients and doctors making decisions together. 2008. [cited 2014] Available from: http://www.gmc-uk.org/guidance/ethical_guidance/consent_guidance_index.asp.Google Scholar
124. Council for International Organizations of Medical Sciences (CIOMS). International ethical guidelines for biomedical research involving human subjects: the world health organization. 2002 [cited 2014]. Available from: www.cioms.ch/publications/guidelines/guidelines_nov_2002_blurb.htm.Google Scholar
125. Salomon, LJ, Ortqvist, L, Aegerter, P, Bussieres, L, Staracci, S, Stirnemann, JJ, et al. Long-term developmental follow-up of infants who participated in a randomized clinical trial of amniocentesis vs laser photocoagulation for the treatment of twin-to-twin transfusion syndrome. Am J Obstet Gynecol 2010; 203 (5): 444.e1–7.Google Scholar
126. Adzick, NS, Thom, EA, Spong, CY, Brock, JW 3rd, Burrows, PK, Johnson, MP, et al. A randomized trial of prenatal versus postnatal repair of myelomeningocele. N Engl J Med 2011; 364 (11): 9931004.Google Scholar
127. Sheppard, MK, Spencer, RN, David, AL, Ashcroft, R. Evaluation of the ethics and social acceptability of a proposed clinical trial using maternal gene therapy to treat severe early-onset fetal growth restriction in pregnant women. Fetal Growth 2014;Oxford 2014.Google Scholar
128. Council of Europe. Additional protocol to the convention on human rights and biomedicine, concerning biomedical research. 2005. Available from: http://conventions.coe.int/Treaty/en/Treaties/html/195.htm.Google Scholar
129. Chervenak, FA, McCullough, LB. An ethically justified framework for clinical investigation to benefit pregnant and fetal patients. Am J Bioeth 2011; 11 (5): 3949.Google Scholar