Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-22dnz Total loading time: 0 Render date: 2024-04-27T16:06:36.391Z Has data issue: false hasContentIssue false

Section 2: - Fetal Disease: Pathogenesis and Treatment

Published online by Cambridge University Press:  21 October 2019

Mark D. Kilby
Affiliation:
University of Birmingham
Anthony Johnson
Affiliation:
University of Texas Medical School at Houston
Dick Oepkes
Affiliation:
Leids Universitair Medisch Centrum
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Fetal Therapy
Scientific Basis and Critical Appraisal of Clinical Benefits
, pp. 91 - 560
Publisher: Cambridge University Press
Print publication year: 2020

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

References

Bowman, JM, Pollock, JM, Penston, LE. Fetomaternal transplacental hemorrhage during pregnancy and after delivery. Vox Sang. 1986; 51: 117–21.CrossRefGoogle ScholarPubMed
Nederlandse Vereniging voor Obstetrie & Gynaecologie (NVOG) (2009). Erytrocytenimmunisatie en zwangerschap: Versie 2.1. https://www.nvog.nl/wp-content/uploads/2018/03/Erytrocytenimmunisatie-en-zwangerschap_.pdfGoogle Scholar
Committee on Practice Bulletins-Obstetrics. Practice Bulletin No. 181: Prevention of Rh D Alloimmunization. Obstet Gynecol. 2017; 130: e57–70.Google Scholar
Qureshi, H, Massey, E, Kirwan, D, Davies, T, Robson, S, White, J, Jones, J, Allard, S, British Society for Haematology. BCSH guideline for the use of anti-D immunoglobulin for the prevention of haemolytic disease of the fetus and newborn. Transfus Med. 2014; 24: 820.Google Scholar
Levine, P. Serological factors as possible causes in spontaneous abortions. J Hered. 1943; 34: 7180.Google Scholar
Zwiers, C, Koelewijn, JM, Vermij, L, van Sambeeck, J, Oepkes, D, de Haas, M, van der Schoot, CE. ABO incompatibility and RhIG immunoprophylaxis protect against non-D alloimmunization by pregnancy. Transfusion. 2018; 58: 1611–17.CrossRefGoogle ScholarPubMed
Royal College of Obstetricians and Gynaecologists (2014). Green-top Guideline 65: The Management of Women with Red Cell Antibodies during Pregnancy. https://www.rcog.org.uk/en/guidelines-research-services/guidelines/gtg65Google Scholar
ACOG. Practice Bulletin No. 192: Management of alloimmunization during pregnancy. Obstet Gynecol. 2018; 131: e82–90.Google Scholar
Rijksinstituut voor Volksgezondheid en Milieu (RIVM) (2018). Draaiboek prenatale screening infectieziekten en erytrocytenimmunisatie: Versie 6.0. https://www.rivm.nl/documenten/draaiboek-prenatale-screening-infectieziekten-en-erytrocytenimmunisatieGoogle Scholar
National Blood Authority Australia (2015). Patient Blood Management Guidelines: Module 5 Obstetrics and Maternity. https://www.blood.gov.au/pbm-module-5Google Scholar
Koelewijn, JM, de Haas, M, Vrijkotte, TG, Bonsel, GJ, van der Schoot, CE. One single dose of 200 microg of antenatal RhIG halves the risk of anti-D immunization and hemolytic disease of the fetus and newborn in the next pregnancy. Transfusion. 2008; 48: 1721–9.Google Scholar
Mayne, S, Parker, JH, Harden, TA, Dodds, SD, Beale, JA. Rate of RhD sensitisation before and after implementation of a community based antenatal prophylaxis programme. BMJ. 1997; 315: 1588.CrossRefGoogle ScholarPubMed
van der Ploeg, CPB, Schönbeck, Y, Oomen, P, Vos, K. Prenatale Screening Infectieziekten en Erytrocytenimmunisatie (PSIE) Procesmonitor 2015. Bilthoven: RIVM, TNO, 2017.Google Scholar
Hendrickson, JE, Delaney, M. Hemolytic Disease of the fetus and newborn: modern practice and future Investigations. Transfus Med Rev. 2016; 30: 159–64.Google Scholar
Scheffer, PG, van der Schoot, CE, Page-Christiaens, GC, de Haas, M. Noninvasive fetal blood group genotyping of rhesus D, c, E and of K in alloimmunised pregnant women: evaluation of a 7-year clinical experience. BJOG. 2011; 118: 1340–8.Google Scholar
Slootweg, YM, Lindenburg, IT, Koelewijn, JM, van Kamp, IL, Oepkes, D, de Haas, M. Predicting anti-Kell-mediated hemolytic disease of the fetus and newborn: diagnostic accuracy of laboratory management. Am J Obstet Gynecol. 2018; 219: 393.e1393.e8.Google Scholar
van Dijk, BA, Dooren, MC, Overbeeke, MA. Red cell antibodies in pregnancy: there is no ‘critical titre’. Transfus Med. 1995; 5: 199202.CrossRefGoogle ScholarPubMed
Oepkes, D, van Kamp, IL, Simon, MJ, Mesman, J, Overbeeke, MA, Kanhai, HH. Clinical value of an antibody-dependent cell-mediated cytotoxicity assay in the management of Rh D alloimmunization. Am J Obstet Gynecol. 2001; 184: 1015–20.Google Scholar
Zwiers, C, van Kamp, I, Oepkes, D, Lopriore, E. Intrauterine transfusion and non-invasive treatment options for hemolytic disease of the fetus and newborn – review on current management and outcome. Expert Rev Hematol. 2017; 10: 337–44.Google Scholar
Mari, G, Deter, RL, Carpenter, RL, Rahman, F, Zimmerman, R, Moise, KJ Jr., et al. Noninvasive diagnosis by Doppler ultrasonography of fetal anemia due to maternal red-cell alloimmunization. Collaborative group for Doppler assessment of the blood velocity in anemic fetuses. N Engl J Med. 2000; 342: 914.Google Scholar
Zimmerman, R, Carpenter, RJ Jr., Durig, P, Mari, G. Longitudinal measurement of peak systolic velocity in the fetal middle cerebral artery for monitoring pregnancies complicated by red cell alloimmunisation: a prospective multicentre trial with intention-to-treat. BJOG. 2002; 109: 746–52.Google ScholarPubMed
Oepkes, D, Brand, R, Vandenbussche, FP, Meerman, RH, Kanhai, HH. The use of ultrasonography and Doppler in the prediction of fetal haemolytic anaemia: a multivariate analysis. Br J Obstet Gynaecol. 1994; 101: 680–4.Google Scholar
Chitkara, U, Wilkins, I, Lynch, L, Mehalek, K, Berkowitz, RL. The role of sonography in assessing severity of fetal anemia in Rh- and Kell-isoimmunized pregnancies. Obstet Gynecol. 1988; 71: 393–8.Google ScholarPubMed
Dukler, D, Oepkes, D, Seaward, G, Windrim, R, Ryan, G. Noninvasive tests to predict fetal anemia: a study comparing Doppler and ultrasound parameters. Am J Obstet Gynecol. 2003; 188: 1310–14.Google Scholar
Oepkes, D, Seaward, PG, Vandenbussche, FP, Windrim, R, Kingdom, J, Beyene, J, Kanhai, HH, Ohlsson, A, Ryan, G, DIAMOND Study Group. Doppler ultrasonography versus amniocentesis to predict fetal anemia. N Engl J Med. 2006; 355: 156–64.Google Scholar
Bang, J, Bock, JE, Trolle, D. Ultrasound-guided fetal intravenous transfusion for severe rhesus haemolytic disease. BMJ (Clin Res Ed). 1982; 284: 373–4.Google Scholar
Moise, KJ Jr. Management of rhesus alloimmunization in pregnancy. Obstet Gynecol. 2008; 112: 164–76.Google Scholar
Nicolaides, KH, Soothill, PW, Clewell, WH, Rodeck, CH, Mibashan, RS, Campbell, S. Fetal haemoglobin measurement in the assessment of red cell isoimmunisation. Lancet. 1988; 1: 1073–5.Google Scholar
Society for Maternal-Fetal Medicine, Mari, G, Norton, ME, Stone, J, Berghella, V, Sciscione, AC, Tate, D, Schenone, MH. Society for Maternal-Fetal Medicine (SMFM) Clinical Guideline #8: the fetus at risk for anemia – diagnosis and management. Am J Obstet Gynecol. 2015; 212: 697710.Google Scholar
Adama van Scheltema, PN, Borkent, S, Sikkel, E, Oepkes, D, Vandenbussche, FP. Fetal brain hemodynamic changes in intrauterine transfusion: influence of needle puncture site. Fetal Diagn Ther. 2009; 26: 131–3.CrossRefGoogle ScholarPubMed
Moise, KJ Jr., Carpenter, RJ Jr., Kirshon, B, Deter, RL, Sala, JD, Cano, LE. Comparison of four types of intrauterine transfusion: effect on fetal hematocrit. Fetal Ther. 1989; 4: 126–37.CrossRefGoogle ScholarPubMed
Zwiers, C, Lindenburg, ITM, Klumper, FJ, de Haas, M, Oepkes, D, Van Kamp, IL. Complications of intrauterine intravascular blood transfusion: lessons learned after 1678 procedures. Ultrasound Obstet Gynecol. 2017; 50: 180–6.Google Scholar
Detti, L, Oz, U, Guney, I, Ferguson, JE, Bahado-Singh, RO, Mari, G. Doppler ultrasound velocimetry for timing the second intrauterine transfusion in fetuses with anemia from red cell alloimmunization. Am J Obstet Gynecol. 2001; 185: 1048–51.Google Scholar
Weiner, CP, Williamson, RA, Wenstrom, KD, Sipes, SL, Widness, JA, Grant, SS, Estle, L. Management of fetal hemolytic disease by cordocentesis. II. Outcome of treatment. Am J Obstet Gynecol. 1991; 165: 1302–7.Google Scholar
Scheier, M, Hernandez-Andrade, E, Fonseca, EB, Nicolaides, KH. Prediction of severe fetal anemia in red blood cell alloimmunization after previous intrauterine transfusions. Am J Obstet Gynecol. 2006; 195: 1550–6.CrossRefGoogle ScholarPubMed
Tiblad, E, Kublickas, M, Ajne, G, Bui, TH, Ek, S, Karlsson, A, Wikman, A, Westgren, M. Procedure-related complications and perinatal outcome after intrauterine transfusions in red cell alloimmunization in Stockholm. Fetal Diagn Ther. 2011; 30: 266–73.Google Scholar
Pasman, SA, Claes, L, Lewi, L, Van Schoubroeck, D, Debeer, A, Emonds, M, Geuten, E, De Catte, L, Devlieger, R. Intrauterine transfusion for fetal anemia due to red blood cell alloimmunization: 14 years experience in Leuven. Facts Views Vis Obgyn. 2015; 7: 129–36.Google Scholar
Sainio, S, Nupponen, I, Kuosmanen, M, Aitokallio-Tallberg, A, Ekholm, E, Halmesmäki, E, Orden, MR, Palo, P, Raudaskoski, T, Tekay, A, Tuimala, J, Uotila, J, Stefanovic, V. Diagnosis and treatment of severe hemolytic disease of the fetus and newborn: a 10-year nationwide retrospective study. Acta Obstet Gynecol Scand. 2015; 94: 383–90.CrossRefGoogle Scholar
Lindenburg, IT, Wolterbeek, R, Oepkes, D, Klumper, FJ, Vandenbussche, FP, van Kamp, IL. Quality control for intravascular intrauterine transfusion using cumulative sum (CUSUM) analysis for the monitoring of individual performance. Fetal Diagn Ther. 2011; 29: 307–14.Google Scholar
Zwiers, C, Oepkes, D, Lopriore, E, Klumper, FJ, De Haas, M, van Kamp, IL. The near disappearance of fetal immune hydrops in relation to current state‐of‐the‐art management of red cell alloimmunization. Prenat Diagn. 2018; 38: 943–50.Google Scholar
van Kamp, IL, Klumper, FJ, Bakkum, RS, Oepkes, D, Meerman, RH, Scherjon, SA, Kanhai, HH. The severity of immune fetal hydrops is predictive of fetal outcome after intrauterine treatment. Am J Obstet Gynecol. 2001; 185: 668–73.Google Scholar
Ree, IMC, Smits-Wintjens, V, van der Bom, JG, van Klink, JMM, Oepkes, D, Lopriore, E. Neonatal management and outcome in alloimmune hemolytic disease. Expert Rev Hematol. 2017; 10: 607–16.Google Scholar
Lindenburg, IT, Smits-Wintjens, VE, van Klink, JM, Verduin, E, van Kamp, IL, Walther, FJ, et al. Long-term neurodevelopmental outcome after intrauterine transfusion for hemolytic disease of the fetus/newborn: the LOTUS study. Am J Obstet Gynecol. 2012; 206: 141. e1–8.CrossRefGoogle ScholarPubMed
Zwiers, C, van der Bom, JG, van Kamp, IL, van Geloven, N, Lopriore, E, Smoleniec, J, et al. Postponing Early Intrauterine Transfusion with Intravenous immunoglobulin Treatment; the PETIT study on severe hemolytic disease of the fetus and newborn. Am J Obstet Gynecol. 2018; 219: e1291. e9.Google Scholar
Bowman, JM. Antenatal suppression of Rh alloimmunization. Clin Obstet Gynecol. 1991; 34: 296303.Google Scholar
Giannina, G, Moise, KJ Jr., Dorman, K. A simple method to estimate volume for fetal intravascular transfusions. Fetal Diagn Ther. 1998; 13: 94–7.Google Scholar

References

Dreyfus, M, Kaplan, C, Verdy, E, Schlegel, N, Durand-Zaleski, I, Tchernia, G. Frequency of immune thrombocytopenia in newborns: a prospective study. Immune Thrombocytopenia Working Group. Blood. 1997; 89: 4402–6.Google Scholar
Sola-Visner, M, Saxonhouse, MA, Brown, RE. Neonatal thrombocytopenia: what we do and don’t know. Early Hum Dev. 2008; 84: 499506.Google Scholar
Winkelhorst, D, Kamphuis, MM, de Kloet, LC, Zwaginga, JJ, Oepkes, D, Lopriore, E. Severe bleeding complications other than intracranial hemorrhage in neonatal alloimmune thrombocytopenia: a case series and review of the literature. Transfusion. 2016; 56: 1230–5.Google Scholar
Tiller, H, Kamphuis, MM, Flodmark, O, Papadogiannakis, N, David, AL, Sainio, S, et al. Fetal intracranial haemorrhages caused by fetal and neonatal alloimmune thrombocytopenia: an observational cohort study of 43 cases from an international multicentre registry. BMJ Open. 2013; 3: e002490.Google Scholar
Campbell, S, Swann, HR, Seif, MW, Kimber, SJ, Aplin, JD. Cell adhesion molecules on the oocyte and preimplantation human embryo. Hum Reprod. 1995; 10: 1571–8.CrossRefGoogle ScholarPubMed
Ohto, H. [Neonatal alloimmune thrombocytopenia]. Nihon Rinsho. 1997; 55: 2310–14.Google Scholar
Ohto, H, Miura, S, Ariga, H, Ishii, T, Fujimori, K, Morita, S. The natural history of maternal immunization against foetal platelet alloantigens. Transfus Med. 2004; 14: 399408.Google Scholar
Kunishima, S, Hayakawa, A, Fujita, K, Saito, H. Transient macrothrombocytopenia associated with maternal-neonatal HPA-21bw incompatibility. Thromb Res. 2013; 131: e286–8.CrossRefGoogle ScholarPubMed
Shivdasani, RA, Rosenblatt, MF, Zucker-Franklin, D, Jackson, CW, Hunt, P, Saris, CJ, Orkin, SH. Transcription factor NF-E2 is required for platelet formation independent of the actions of thrombopoietin/MGDF in megakaryocyte development. Cell. 1995; 81: 695704.Google Scholar
van Gils, JM, Stutterheim, J, van Duijn, TJ, Zwaginga, JJ, Porcelijn, L, de Haas, M, Hordijk, PL. HPA-1a alloantibodies reduce endothelial cell spreading and monolayer integrity. Mol Immunol. 2009; 46: 406–15.Google Scholar
Yougbaré, I, Lang, S, Yang, H, Chen, P, Zhao, X, Tai, WS, et al. Maternal anti-platelet beta3 integrins impair angiogenesis and cause intracranial hemorrhage. J Clin Invest. 2015; 125: 1545–56.Google Scholar
Santoso, S, Wihadmadyatami, H, Bakchoul, T, Werth, S, Al-Fakhri, N, Bein, G, et al. Antiendothelial alphavbeta3 antibodies are a major cause of intracranial bleeding in fetal/neonatal alloimmune thrombocytopenia. Arterioscler Thromb Vasc Biol. 2016; 36: 1517–24.Google Scholar
Tiller, H, Killie, MK, Husebekk, A, Skogen, B, Ni, H, Kjeldsen-Kragh, J, Øian, P. Platelet antibodies and fetal growth: maternal antibodies against fetal platelet antigen 1a are strongly associated with reduced birthweight in boys. Acta Obstet Gynecol Scand. 2012; 91: 7986.Google Scholar
Murphy, MF, Hambley, H, Nicolaides, K, Waters, AH. Severe fetomaternal alloimmune thrombocytopenia presenting with fetal hydrocephalus. Prenat Diagn. 1996; 16: 1152–5.3.0.CO;2-J>CrossRefGoogle ScholarPubMed
Althaus, J, Weir, EG, Askin, F, Kickler, TS, Blakemore, K. Chronic villitis in untreated neonatal alloimmune thrombocytopenia: an etiology for severe early intrauterine growth restriction and the effect of intravenous immunoglobulin therapy. Am J Obstet Gynecol. 2005; 193: 1100–4.Google Scholar
Pereira, J, Cretney, C, Aster, RH. Variation of class I HLA antigen expression among platelet density cohorts: a possible index of platelet age? Blood. 1988; 71: 516–19.CrossRefGoogle ScholarPubMed
Masson, E, Vidal, C, Deschamps, M, Bongain, S, Thevenin, C, Dupont, I, et al. Incidence and risk factors of anti-HLA immunization after pregnancy. Hum Immunol. 2013; 74: 946–51.CrossRefGoogle Scholar
Sharon, R, Amar, A. Maternal anit-HLA antibodies and neonatal thrombocytopenia. Lancet. 1981; 1: 1313.Google Scholar
Vilches, M, Nieto, A. Analysis of Pregnancy-Induced Anti-HLA Antibodies Using Luminex Platform. Transplant Proc. 2015; 47: 2608–10.Google Scholar
Chow, MP, Sun, KJ, Yung, CH, Hu, HY, Tzeng, JL, Lee, TD. Neonatal alloimmune thrombocytopenia due to HLA-A2 antibody. Acta Haematol. 1992; 87: 153–5.CrossRefGoogle ScholarPubMed
De Tar, MW, Klohe, E, Grosset, A, Rau, T. Neonatal alloimmune thrombocytopenia with HLA alloimmunization: case report with immunohematologic and placental findings. Pediatr Dev Pathol. 2002; 5: 200–5.Google ScholarPubMed
del Rosario, ML, Fox, ER, Kickler, TS, Kao, KJ. Neonatal alloimmune thrombocytopenia associated with maternal anti-HLA antibody: a case report. J Pediatr Hematol Oncol. 1998; 20: 252–6.Google Scholar
Gramatges, MM, Fani, P, Nadeau, K, Pereira, S, Jeng, MR. Neonatal alloimmune thrombocytopenia and neutropenia associated with maternal human leukocyte antigen antibodies. Pediatr Blood Cancer. 2009; 53: 97–9.Google Scholar
Hutchinson, AL, Dennington, PM, Holdsworth, R, Downe, L. Recurrent HLA-B56 mediated neonatal alloimmune thrombocytopenia with fatal outcomes. Transfus Apher Sci. 2015; 52: 311–13.CrossRefGoogle ScholarPubMed
Moncharmont, P, Dubois, V, Obegi, C, Vignal, M, Mérieux, Y, Gebuhrer, L, Rigal, D. HLA antibodies and neonatal alloimmune thrombocytopenia. Acta Haematol. 2004; 111: 215–20.Google Scholar
Onishi, S, Okubo, S, Matsuzaki, T, Ishida, T, Yasunaga, K. [Report of two cases of neonatal alloimmune thrombocytopenia caused by anti-HLA antibody, and their screening using umbilical cord blood]. Rinsho Ketsueki. 1992; 33: 42–7.Google Scholar
Saito, S, Ota, M, Komatsu, Y, Ota, S, Aoki, S, Koike, K, et al. Serologic analysis of three cases of neonatal alloimmune thrombocytopenia associated with HLA antibodies. Transfusion. 2003; 43: 908–17.CrossRefGoogle ScholarPubMed
Sasaki, M, Yagihashi, A, Kobayashi, D, Watanabe, N, Fujikawa, T, Chiba, S, et al. Neonatal alloimmune thrombocytopenia due to anti-human leukocyte antigen antibody: a case report. Pediatr Hematol Oncol. 2001; 18: 519–24.Google Scholar
Thude, H, Schorner, U, Helfricht, C, Loth, M, Maak, B, Barz, D. Neonatal alloimmune thrombocytopenia caused by human leucocyte antigen-B27 antibody. Transfus Med. 2006; 16: 143–9.Google Scholar
Starcevic, M, Tomicic, M, Malenica, M, Zah-Matakovic, V. Neonatal alloimmune thrombocytopenia caused by anti-HLA-A24 alloantibodies. Acta Paediatr. 2010; 99: 630–2.Google Scholar
Winkelhorst, D, Porcelijn, L, van de Weerd, JME, Huiskes, E, Muizelaar, E, Lardy, NM, et al. HLA class I antibodies in FNAIT. (Poster). 14th European Symposium on Platelet and Granulocyte Immunobiology. Stockholm, 2016.Google Scholar
Wu, S, Maslanka, K, Gorski, J. An integrin polymorphism that defines reactivity with alloantibodies generates an anchor for MHC class II peptide binding: a model for unidirectional alloimmune responses. J Immunol. 1997; 158: 3221–6.CrossRefGoogle Scholar
Burrows, RF, Kelton, JG. Fetal thrombocytopenia and its relation to maternal thrombocytopenia. New Engl J Med. 1993; 329: 1463–6.Google Scholar
Kamphuis, MM, Paridaans, NP, Porcelijn, L, Lopriore, E, Oepkes, D. Incidence and consequences of neonatal alloimmune thrombocytopenia: a systematic review. Pediatrics. 2014; 133: 715–21.CrossRefGoogle ScholarPubMed
Kamphuis, MM, Paridaans, N, Porcelijn, L, De Haas, M, Van Der Schoot, CE, Brand, A, Bonsel, GJ, Oepkes, D. Screening in pregnancy for fetal or neonatal alloimmune thrombocytopenia: systematic review. BJOG. 2010; 117: 1335–43.Google Scholar
Davoren, A, McParland, P, Barnes, CA, Murphy, WG. Neonatal alloimmune thrombocytopenia in the Irish population: a discrepancy between observed and expected cases. J Clin Pathol. 2002; 55: 289–92.Google Scholar
Spencer, JA, Burrows, RF. Feto-maternal alloimmune thrombocytopenia: a literature review and statistical analysis. Aust N Z J Obstet Gynaecol. 2001; 41: 4555.Google Scholar
Kiefel, V, Santoso, S, Weisheit, M, Mueller-Eckhardt, C. Monoclonal antibody--specific immobilization of platelet antigens (MAIPA): a new tool for the identification of platelet-reactive antibodies. Blood. 1987; 70: 1722–6.Google Scholar
Scheffer, PG, Ait Soussan, A, Verhagen, OJ, Page-Christiaens, GC, Oepkes, D, de Haas, M, Van Der Schoot, CE. Noninvasive fetal genotyping of human platelet antigen-1a. BJOG. 2011; 118: 1392–5.Google Scholar
Wienzek-Lischka, S, Krautwurst, A, Fröhner, V, Hackstein, H, Gattenlöhner, S, Bräuninger, A, et al. Noninvasive fetal genotyping of human platelet antigen-1a using targeted massively parallel sequencing. Transfusion. 2015; 55: 1538–44.Google Scholar
Bessos, H, Turner, M, Urbaniak, SJ. Is there a relationship between anti-HPA-1a concentration and severity of neonatal alloimmune thrombocytopenia? Immunohematology. 2005; 21: 102–9.Google Scholar
Sonneveld, ME, Natunen, S, Sainio, S, Koeleman, CA, Holst, S, Dekkers, G, et al. Glycosylation pattern of anti-platelet IgG is stable during pregnancy and predicts clinical outcome in alloimmune thrombocytopenia. Br J Haematol. 2016; 174: 310–20.CrossRefGoogle ScholarPubMed
Radder, CM, Brand, A, Kanhai, HH. Will it ever be possible to balance the risk of intracranial haemorrhage in fetal or neonatal alloimmune thrombocytopenia against the risk of treatment strategies to prevent it? Vox Sang. 2003; 84: 318–25.Google Scholar
Winkelhorst, D, Murphy, MF, Greinacher, A, Shehata, N, Bakchoul, T, Massey, E, et al. Antenatal management in fetal and neonatal alloimmune thrombocytopenia: a systematic review. Blood. 2017; 129: 1538–47.Google Scholar
Bussel, JB, Berkowitz, RL, McFarland, JG, Lynch, L, Chitkara, U. Antenatal treatment of neonatal alloimmune thrombocytopenia. New Engl J Med. 1988; 319: 1374–8.Google Scholar
Daffos, F, Forestier, F, Muller, JY, Reznikoff-Etievant, M, Habibi, B, Capella-Pavlovsky, M, et al. Prenatal treatment of alloimmune thrombocytopenia. Lancet. 1984; 2: 632.Google Scholar
Sainio, S, Teramo, K, Kekomaki, R. Prenatal treatment of severe fetomaternal alloimmune thrombocytopenia. Transfus Med. 1999; 9: 321–30.Google Scholar
Kamphuis, M, Paridaans, N, Winkelhorst, D, Wikman, A, Tiblad, E, Lopriore, E, et al. Lower-dose intravenous immunoglobulins for the treatment of fetal and neonatal alloimmune thrombocytopenia: a cohort study. Transfusion. 2016; 56: 2308–13.Google Scholar
Bussel, JB, Berkowitz, RL, Hung, C, Kolb, EA, Wissert, M, Primiani, A, et al. Intracranial hemorrhage in alloimmune thrombocytopenia: stratified management to prevent recurrence in the subsequent affected fetus. Am J Obstet Gynecol. 2010; 203: 135.e1–14.Google Scholar
Berkowitz, RL, Lesser, ML, McFarland, JG, Wissert, M, Primiani, A, Hung, C, Bussel, JB. Antepartum treatment without early cordocentesis for standard-risk alloimmune thrombocytopenia: a randomized controlled trial. Obstet Gynecol. 2007; 110: 249–55.CrossRefGoogle ScholarPubMed
Radder, CM, de Haan, MJ, Brand, A, Stoelhorst, GM, Veen, S, Kanhai, HH. Follow up of children after antenatal treatment for alloimmune thrombocytopenia. Early Hum Dev. 2004; 80: 6576.Google Scholar
Kaplan, C, Murphy, MF, Kroll, H, Waters, AH. Feto-maternal alloimmune thrombocytopenia: antenatal therapy with IvIgG and steroids – more questions than answers. European Working Group on FMAIT. Br J Haematol. 1998; 100: 62–5.Google Scholar
Bertrand, G, Drame, M, Martageix, C, Kaplan, C. Prediction of the fetal status in noninvasive management of alloimmune thrombocytopenia. Blood. 2011; 117: 3209–13.Google Scholar
Berkowitz, RL, Kolb, EA, McFarland, JG, Wissert, M, Primani, A, Lesser, M, Bussel, JB. Parallel randomized trials of risk-based therapy for fetal alloimmune thrombocytopenia. Obstet Gynecol. 2006; 107: 91–6.Google Scholar
Bussel, JB, Berkowitz, RL, Lynch, L, Lesser, ML, Paidas, MJ, Huang, CL, McFarland, JG. Antenatal management of alloimmune thrombocytopenia with intravenous gamma-globulin: a randomized trial of the addition of low-dose steroid to intravenous gamma-globulin. Am J Obstet Gynecol. 1996; 174: 1414–23.Google Scholar
Wenstrom, KD, Weiner, CP, Williamson, RA. Antenatal treatment of fetal alloimmune thrombocytopenia. Obstet Gynecol. 1992; 80: 433–5.Google Scholar
Lynch, L, Bussel, JB, McFarland, JG, Chitkara, U, Berkowitz, RL. Antenatal treatment of alloimmune thrombocytopenia. Obstet Gynecol. 1992; 80: 6771.Google Scholar
de Haas, M, Thurik, FF, Koelewijn, JM, van der Schoot, CE. Haemolytic disease of the fetus and newborn. Vox Sang. 2015; 109: 99113.Google Scholar
Zwiers, C, Lindenburg, ITM, Klumper, FJ, de Haas, M, Oepkes, D, Van Kamp, IL. Complications of intrauterine intravascular blood transfusion: lessons learned after 1678 procedures. Ultrasound Obstet Gynecol. 2017; 50: 180–6.Google Scholar
Bowman, J. Thirty-five years of Rh prophylaxis. Transfusion. 2003; 43: 1661–6.Google Scholar
Tiller, H, Killie, MK, Chen, P, Eksteen, M, Husebekk, A, Skogen, B, Kjeldsen-Kragh, J, Ni, H. Toward a prophylaxis against fetal and neonatal alloimmune thrombocytopenia: induction of antibody-mediated immune suppression and prevention of severe clinical complications in a murine model. Transfusion. 2012; 52: 1446–57.Google Scholar
Ghevaert, C, Wilcox, DA, Fang, J, Armour, KL, Clark, MR, Ouwehand, WH, Williamson, LM. Developing recombinant HPA-1a-specific antibodies with abrogated Fcgamma receptor binding for the treatment of fetomaternal alloimmune thrombocytopenia. J Clin Invest. 2008; 118: 2929–38.Google Scholar
Ghevaert, C, Herbert, N, Hawkins, L, Grehan, N, Cookson, P, Garner, SF, et al. Recombinant HPA-1a antibody therapy for treatment of fetomaternal alloimmune thrombocytopenia: proof of principle in human volunteers. Blood. 2013; 122: 313–20.Google Scholar
Kjeldsen-Kragh, J, Killie, MK, Tomter, G, Golebiowska, E, Randen, I, Hauge, R, et al. A screening and intervention program aimed to reduce mortality and serious morbidity associated with severe neonatal alloimmune thrombocytopenia. Blood. 2007; 110: 833–9.Google Scholar
van den Akker, ESA, Oepkes, D, Brand, A, Kanhai, HHH. Vaginal delivery for fetuses at risk of alloimmune thrombocytopenia? BJOG. 2006; 113: 781–3.Google Scholar
Roberts, I, Murray, NA. Neonatal thrombocytopenia: causes and management. Arch Dis Child Fetal Neonatal Ed. 2003; 88: F359–64.Google Scholar
Gunnink, SF, Vlug, R, Fijnvandraat, K, van der Bom, JG, Stanworth, SJ, Lopriore, E. Neonatal thrombocytopenia: etiology, management and outcome. Expert Rev Hematol. 2014; 7: 387–95.Google Scholar
Ghevaert, C, Campbell, K, Walton, J, Smith, GA, Allen, D, Williamson, LM, et al. Management and outcome of 200 cases of fetomaternal alloimmune thrombocytopenia. Transfusion. 2007; 47: 901–10.Google Scholar
Inder, TE, Perlman, M, Volpe, JJ. Intracranial Hemorrhage: Subdural, Subarachnoid, Intraventricular (Term Infant), Miscellaneous. In Volpe, JJ, Neurology of the Newborn. 6th Edition ed. Philadelphia: Elsevier, 2018.Google Scholar
Vinekar, A, Hegde, K, Gilbert, C, Braganza, S, Pradeep, M, Shetty, R, Shetty, KB. Do platelets have a role in the pathogenesis of aggressive posterior retinopathy of prematurity? Retina. 2010; 30: S20–3.Google Scholar
Stanworth, SJ, Clarke, P, Watts, T, Ballard, S, Choo, L, Morris, T, Murphy, MF, Roberts, I, Platelets and Neonatal Transfusion Study Group. Prospective, observational study of outcomes in neonates with severe thrombocytopenia. Pediatrics. 2009; 124: e826–34.CrossRefGoogle ScholarPubMed
Murray, NA, Roberts, IA. Circulating megakaryocytes and their progenitors in early thrombocytopenia in preterm neonates. Pediatr Res. 1996; 40: 112–19.Google Scholar
Blajchman, MA, Ali, AM, Richardson, HL. Bacterial contamination of cellular blood components. Vox Sang. 1994; 67 (Suppl. 3): 2533.Google Scholar
Murphy, MF, Williamson, LM. Antenatal screening for fetomaternal alloimmune thrombocytopenia: an evaluation using the criteria of the UK National Screening Committee. Br J Haematol. 2000; 111: 726–32.Google Scholar
Kiefel, V, Bassler, D, Kroll, H, Paes, B, Giers, G, Ditomasso, J, et al. Antigen-positive platelet transfusion in neonatal alloimmune thrombocytopenia (NAIT). Blood. 2006; 107: 3761–3.Google Scholar
Chen, P, Li, C, Lang, S, Zhu, G, Reheman, A, Spring, CM, Freedman, J, Ni, H. Animal model of fetal and neonatal immune thrombocytopenia: role of neonatal Fc receptor in the pathogenesis and therapy. Blood. 2010; 116: 3660–8.Google Scholar
Sidiropoulos, D, Straume, B. The treatment of neonatal isoimmune thrombocytopenia with intravenous immunoglobin (IgG i.v.). Blut. 1984; 48: 383–6.Google Scholar
Derycke, M, Dreyfus, M, Ropert, JC, Tchernia, G. Intravenous immunoglobulin for neonatal isoimmune thrombocytopenia. Arch Dis Child. 1985; 60: 667–9.Google Scholar
Winkelhorst, D, Kamphuis, MM, Steggerda, SJ, Rijken, M, Oepkes, D, Lopriore, E, van Klink, JMM. Perinatal outcome and long-term neurodevelopment after intracranial haemorrhage due to fetal and neonatal alloimmune thrombocytopenia. Fetal Diagn Ther. 2019; 45: 184–91.Google Scholar
Winkelhorst, D, Oostweegel, M, Porcelijn, L, Middelburg, RA, Zwaginga, JJ, Oepkes, D, et al. Treatment and outcomes of fetal/neonatal alloimmune thrombocytopenia: a nationwide cohort study in newly detected cases. Br J Haematol. 2019; 184: 1026–9.Google Scholar

References

Poelmann, RE, Gittenberger-de Groot, AC, Biermans, MWM, Dolfing, AI, Jagessar, A, van Hattum, S, et al. Outflow tract septation and the aortic arch system in reptiles: lessons for understanding the mammalian heart. Evodevo; 2017; 8: 9.CrossRefGoogle ScholarPubMed
DeRuiter, MC, Poelmann, RE, VanderPlas-de Vries, I, Mentink, MM, Gittenberger-de Groot, AC. The development of the myocardium and endocardium in mouse embryos. Fusion of two heart tubes? Anat Embryol (Berl). 1992; 185: 461–73.Google Scholar
Buckingham, M, Meilhac, S, Zaffran, S. Building the mammalian heart from two sources of myocardial cells. Nat Rev Genet. 2005; 6: 826–35.Google Scholar
Cai, CL, Liang, X, Shi, Y, Chu, PH, Pfaff, SL, Chen, J, Evans, S. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev Cell. 2003; 5: 877–89.Google Scholar
de la Cruz, M, Sanchez-Gomez, C, Palomino, MA. The primitive cardiac regions in the straight tube heart (Stage 9-) and their anatomical expression in the mature heart: an experimental study in the chick heart. J Anat. 1989; 165: 121–31.Google Scholar
Miquerol, L, Kelly, RG. Organogenesis of the vertebrate heart. Wiley Interdiscip Rev Dev Biol. 2013; 2: 1729.Google Scholar
Kirby, ML, Gale, TF, Stewart, DE. Neural crest cells contribute to normal aorticopulmonary septation. Science. 1983; 220: 1059–61.Google Scholar
Bergwerff, M, Verberne, ME, DeRuiter, MC, Poelmann, RE, Gittenberger-de Groot, AC. Neural crest cell contribution to the developing circulatory system: implications for vascular morphology? Circ Res. 1998; 82: 221–31.Google Scholar
Farrell, MJ, Burch, JL, Wallis, K, Rowley, L, Kumiski, D, Stadt, H, Godt, RE, Creazzo, TL, Kirby, ML. FGF-8 in the ventral pharynx alters development of myocardial calcium transients after neural crest ablation. J Clin Invest. 2001; 107: 1509–17.Google Scholar
Lindsay, EA, Vitelli, F, Su, H, Morishima, M, Huynh, T, Pramparo, T, Jurecic, V, Ogunrinu, G, Sutherland, HF, Scambler, PJ, Bradley, A, Baldini, A. Tbx1 haploinsufficiency in the DiGeorge syndrome region causes aortic arch defects in mice. Nature. 2001; 410: 97101.Google Scholar
Poelmann, RE, Mikawa, T, Gittenberger-de Groot, AC. Neural crest cells in outflow tract septation of the embryonic chicken heart: differentiation and apoptosis. Dev Dyn. 1998; 212: 373–84.Google Scholar
Poelmann, RE, Gittenberger-de Groot, AC. A dual pathway to the heart links neural crest to in- and outflow tract septation and to differentiation of the conduction system. Anat Embryol. 2000; 231–5.Google Scholar
Gurjarpadhye, A, Hewett, KW, Justus, C, Wen, X, Stadt, H, Kirby, ML, Sedmera, D, Gourdie, RG. Cardiac neural crest ablation inhibits compaction and electrical function of conduction system bundles. Am J Physiol Heart Circ Physiol. 2007; 292: H1291–300.Google Scholar
Bax, NA, Bleyl, SB, Gallini, R, Wisse, LJ, Hunter, J, van Oorschot, AAM, Mahtab, EAF, Lie-Venema, H, Goumans, M-J, Betsholtz, C, Gittenberger-de Groot, AC. Cardiac malformations in Pdgfralpha mutant embryos are associated with increased expression of WT1 and Nkx2.5 in the second heart field. Dev Dyn. 2010; 239: 2307–17.Google Scholar
Kruithof, BP, van Wijk, B, Somi, S, Kruithof-de Julio, M, Pérez Pomares, JM, Weesie, F, Wessels, A, Moorman, AF, van den Hoff, MJ. BMP and FGF regulate the differentiation of multipotential pericardial mesoderm into the myocardial or epicardial lineage. Dev Biol. 2006; 295: 507–22.Google Scholar
Pérez-Pomares, JM, Phelps, A, Sedmerova, M, Carmona, R, González-Iriarte, M, Muñoz-Chápuli, R, Wessels, A. Experimental studies on the spatiotemporal expression of WT1 and RALDH2 in the embryonic avian heart: a model for the regulation of myocardial and valvuloseptal development by epicardially derived cells (EPDCs). Dev Biol. 2002; 247: 307–26.Google Scholar
Gittenberger-de Groot, AC, Winter, EM, Bartelings, MM, Goumans, MJ, DeRuiter, MC, Poelmann, RE. The arterial and cardiac epicardium in development, disease and repair. Differentiation. 2012 ; 84: 4153.Google Scholar
Gittenberger-de Groot, AC, Vrancken Peeters, MP, Mentink, MM, Gourdie, RG, Poelmann, RE. Epicardium-derived cells contribute a novel population to the myocardial wall and the atrioventricular cushions. Circ Res. 1998; 82: 1043–52.Google Scholar
Lie-Venema, H, van den Akker, NM, Bax, NA, Winter, EM, Maas, S, Kekarainen, T, Hoeben, RC, deRuiter, MC, Poelmann, RE, Gittenberger-de Groot, AC. Origin, fate, and function of epicardium-derived cells (EPCDs) in normal and abnormal cardiac development. ScientificWorldJournal. 2007; 7: 1777–98.Google Scholar
Red-Horse, K, Ueno, H, Weissman, IL, Krasnow, MA. Coronary arteries form by developmental reprogramming of venous cells. Nature. 2010; 464: 549–53.Google Scholar
Palmquist-Gomes, P, Guadix, JA, Pérez-Pomares, JM. Avian embryonic coronary arterio-venous patterning involves the contribution of different endothelial and endocardial cell populations. Dev Dyn. 2018; 247: 686–98.Google Scholar
Tian, X, Hu, T, He, L, Zhang, H, Huang, X, Poelmann, RE, Liu, W, Yang, Z, Yan, Y, Pu, WT, Zhou, B. Peritruncal coronary endothelial cells contribute to proximal coronary artery stems and their aortic orifices in the mouse heart. PLoS One. 2013; 8: e80857.Google Scholar
Zhou, B, Ma, Q, Rajagopal, S, Wu, SM, Domian, I, Rivera-Feliciano, J, Jiang, D, von Gise, A, Ikeda, S, Chien, KR, Pu, WT. Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature. 2008; 454: 109–13.Google Scholar
Gittenberger-de Groot, AC, Winter, EM, Poelmann, RE. Epicardium-derived cells (EPDCs) in development, cardiac disease and repair of ischemia. J Cell Mol Med. 2010; 14: 1056–60.Google Scholar
Goumans, MJ, de Boer, TP, Smits, AM, van Laake, LW, van Vliet, P, Metz, CH, et al. TGF-beta1 induces efficient differentiation of human cardiomyocyte progenitor cells into functional cardiomyocytes in vitro. Stem Cell Res. 2007; 1: 138–49.Google Scholar
Winter, EM, Van Oorschot, AA, Hogers, B, van der Graaf, LM, Doevendans, PA, Poelmann, RE, Atsma, DE, Gittenberger-de Groot, AC, Goumans, MJ. A new direction for cardiac regeneration therapy: application of synergistically acting epicardium-derived cells and cardiomyocyte progenitor cells. Circ Heart Fail. 2009; 2: 643–53.Google Scholar
van Vliet, P, Smits, AM, de Boer, TP, Korfage, TH, Metz, CH, Roccio, M, van der Heyden, MA, van Veen, TA, Sluijter, JP, Doevendans, PA, Goumans, MJ. Foetal and adult cardiomyocyte progenitor cells have different developmental potential. J Cell Mol Med. 2010; 14: 861–70.Google Scholar
Poelmann, RE, Jongbloed, MR, Gittenberger-de Groot, AC. Pitx2: a challenging teenager. Circ Res. 2008; 102: 749–51.Google Scholar
Franco, D, Campione, M. The role of Pitx2 during cardiac development. Linking left-right signaling and congenital heart diseases. Trends Cardiovasc Med. 2003; 13: 157–63.CrossRefGoogle ScholarPubMed
Manasek, FJ, Monroe, RG. Early cardiac morphogenesis is independent of function. Dev Biol. 1972; 27: 584–8.Google Scholar
Bouman, HG, Broekhuizen, ML, Baasten, AM, Gittenberger-de Groot, AC, Wenink, AC. Spectrum of looping disturbances in stage 34 chicken hearts after retinoic acid treatment. Anat Rec. 1995; 243: 101–8.Google Scholar
Blom, NA, Gittenberger-de Groot, AC, DeRuiter, MC, Poelmann, RE, Mentink, MM, Ottenkamp, J. Development of the cardiac conduction tissue in human embryos using HNK-1 antigen expression: possible relevance for understanding of abnormal atrial automaticity. Circulation. 1999; 99: 800–6.Google Scholar
Gittenberger-de Groot, AC, Calkoen, EE, Poelmann, RE, Bartelings, MM, Jongbloed, MR. Morphogenesis and molecular considerations on congenital cardiac septal defects. Ann Med. 2014; 46: 640–52.Google Scholar
Scherptong, RW, Jongbloed, MR, Wisse, LJ, Vicente-Steijn, R, Bartelings, MM, Poelmann, RE, Schalij, MJ, Gittenberger-de Groot, AC. Morphogenesis of outflow tract rotation during cardiac development: the pulmonary push concept. Dev Dyn. 2012; 241: 1413–22.Google Scholar
Bartelings, MM, Gittenberger-de Groot, AC, Wenink, ACG, et al. The morphogenesis of common arterial trunk reconsidered. Recent and classical views. Cardia Selectief. 1992; 5: 10-10.Google Scholar
Conway, SJ, Bundy, J, Chen, J, Dickman, E, Rogers, R, Will, BM. Decreased neural crest stem cell expansion is responsible for the conotruncal heart defects within the splotch (Sp(2H))/Pax3 mouse mutant. Cardiovasc Res. 2000; 47: 314–28.Google Scholar
Kirby, ML, Waldo, KL. Role of neural crest in congenital heart disease. Circulation. 1990; 82: 332–40.Google Scholar
Baardman, ME, Zwier, MV, Wisse, LJ, Gittenberger-de Groot, AC, Kerstjens-Frederikse, WS, Hofstra, RM, et al. Common arterial trunk and ventricular non-compaction in Lrp2 knockout mice indicate a crucial role of LRP2 in cardiac development. Dis Model Mech. 2016; 9: 413–25.Google Scholar
Bogers, AJ, Bartelings, MM, Bökenkamp, R, Stijnen, T, van Suylen, RJ, Poelmann, RE, Gittenberger-de Groot, AC. Common arterial trunk, uncommon coronary arterial anatomy. J Thorac Cardiovasc Surg. 1993; 106: 1133–7.Google Scholar
Gittenberger-de Groot, AC, Bartelings, MM, Bogers, AJJC, Boot, MJ, Poelmann, RE. The embryology of the common arterial trunk. Progr Pediatr Cardiol. 2002; 15: 18.Google Scholar
Van Den Akker, NM, Molin, DG, Peters, PP, Maas, S, Wisse, LJ, van Brempt, R, et al. Tetralogy of Fallot and alterations in vascular endothelial growth factor-A signaling and notch signaling in mouse embryos solely expressing the VEGF120 isoform. Circ Res. 2007; 100: 842–9.Google Scholar
Molin, DG, Roest, PA, Nordstrand, H, Wisse, LJ, Poelmann, RE, Eriksson, UJ, Gittenberger-de Groot, AC. Disturbed morphogenesis of cardiac outflow tract and increased rate of aortic arch anomalies in the offspring of diabetic rats. Birth Defects Res A Clin Mol Teratol. 2004; 70: 927–38.Google Scholar
Bartram, U, Molin, DG, Wisse, LJ, Mohamad, A, Sanford, LP, Doetschman, T, Speer, CP, Poelmann, RE, Gittenberger-de Groot, AC. Double-outlet right ventricle and overriding tricuspid valve reflect disturbances of looping, myocardialization, endocardial cushion differentiation, and apoptosis in TGFß2-knockout mice. Circulation. 2001; 103: 2745–52.Google Scholar
Jenkins, SJ, Hutson, DR, Kubalak, SW. Analysis of the proepicardium-epicardium transition during the malformation of the RXRalpha-/- epicardium. Dev Dyn. 2005; 233: 1091–101.Google Scholar
Hogers, B, DeRuiter, MC, Gittenberger-de Groot, AC, Poelmann, RE. Unilateral vitelline vein ligation alters intracardiac blood flow patterns and morphogenesis in the chick embryo. Circ Res. 1997; 80: 473–81.Google Scholar
Van Loo, PF, Mahtab, EA, Wisse, LJ, Hou, J, Grosveld, F, Suske, G, Philipsen, S, Gittenberger-de Groot, AC. Transcription Factor Sp3 knockout mice display serious cardiac malformations. Mol Cell Biol. 2007; 27: 8571–82.Google Scholar
Gittenberger-de Groot, AC, Vrancken Peeters, MP, Bergwerff, M, Mentink, MM, Poelmann, RE. Epicardial outgrowth inhibition leads to compensatory mesothelial outflow tract collar and abnormal cardiac septation and coronary formation. Circ Res. 2000; 87: 969–71.Google Scholar
Nakajima, Y, Morishima, M, Nakazawa, M, Momma, K. Inhibition of outflow cushion mesenchyme formation in retinoic acid-induced complete transposition of the great arteries. Cardiovasc Res. 1996; 31: E77–85.CrossRefGoogle ScholarPubMed
Moazzen, H, Lu, X, Ma, NL, Velenosi, TJ, Urquhart, BL, Wisse, LJ, Gittenberger-de Groot, AC, Feng, Q. N-Acetylcysteine prevents congenital heart defects induced by pregestational diabetes. Cardiovasc Diabetol. 2014; 18: 1346.Google Scholar
Blom, NA, Ottenkamp, J, Jongeneel, TH, DeRuiter, MC, Gittenberger-de Groot, AC. Morphogenetic differences of secundum atrial septal defects. Pediatr Cardiol. 2005; 26: 338–43.Google Scholar
Benson, DW, Silberbach, GM, Kavanaugh-McHugh, A, Cottrill, C, Zhang, Y, Riggs, S, et al. Mutations in the cardiac transcription factor NKX2.5 affect diverse cardiac developmental pathways. J Clin Invest. 1999; 104: 1567–73.Google Scholar
Moskowitz, IP, Kim, JB, Moore, ML, Wolf, CM, Peterson, MA, Shendure, J, Nobrega, MA, Yokota, Y, Berul, C, Izumo, S, Seidman, JG, Seidman, CE. A molecular pathway including Id2, Tbx5, and Nkx2–5 required for cardiac conduction system development. Cell. 2007; 129: 1365–76.Google Scholar
Blaschke, RJ, Hahurij, ND, Kuijper, S, Just, S, Wisse, LJ, Deissler, K, et al. Targeted mutation reveals essential functions of the homeodomain transcription factor Shox2 in sinoatrial and pacemaking development. Circulation 2007; 115: 1830–8.Google Scholar
Barlow, GM, Chen, X-N, Shi, ZY, Lyons, GE, Kurnit, DM, Celle, L, et al. Down syndrome congenital heart disease: a narrowed region and a candidate gene. Genet Med. 2001; 3: 91101.Google Scholar
Blom, NA, Ottenkamp, J, Wenink, AG, Gittenberger-de Groot, AC. Deficiency of the vestibular spine in atrioventricular septal defects in human fetuses with down syndrome. Am J Cardiol. 2003; 91: 180–4.Google Scholar
Mahtab, EA, Wijffels, MC, van den Akker, NM, Hahurij, ND, Lie-Venema, H, Wisse, LJ, et al. Cardiac malformations and myocardial abnormalities in podoplanin knockout mouse embryos: correlation with abnormal epicardial development. Dev Dyn. 2008; 237: 847–57.Google Scholar
Steimle, JD, Moskowitz, IP. TBX5: A Key Regulator of Heart Development. Curr Top Dev Biol. 2017; 122: 195221.Google Scholar
Hinton, RB Jr., Martin, LJ, Tabangin, ME, Mazwi, ML, Cripe, LH, Benson, DW. Hypoplastic left heart syndrome is heritable. J Am Coll Cardiol. 2007; 50: 1590–5.Google Scholar
Wenink, AC, Gittenberger-de Groot, AC, Brom, AG. Developmental considerations of mitral valve anomalies. Int J Cardiol. 1986; 11: 8598.Google Scholar
Elzenga, N, Gittenberger-de Groot, AC. Coarctation and related aortic arch anomalies in hypoplastic left heart syndrome. Int J Cardiol. 1985; 8: 379–89.Google Scholar
Sedmera, D, Pexieder, T, Rychterova, V, Hu, N, Clark, EB. Remodeling of chick embryonic ventricular myoarchitecture under experimentally changed loading conditions. Anat Rec. 1999; 254: 238–52.Google Scholar
Sizarov, A, Boudjemline, Y. Valve Interventions in utero: understanding the timing, indications, and approaches. Can J Cardiol. 2017; 33: 1150–8.Google Scholar
Bartram, U, Bartelings, MM, Kramer, HH, Gittenberger-de Groot, AC. Congenital polyvalvular disease: a review. Pediatr Cardiol. 2001; 22: 93101.CrossRefGoogle ScholarPubMed
Garg, V, Muth, AN, Ransom, JF, Schluterman, MK, Barnes, R, King, IN, Grossfeld, PD, Srivastava, D. Mutations in NOTCH1 cause aortic valve disease. Nature. 2005; 437: 270–4.Google Scholar
Grewal, N, DeRuiter, MC, Jongbloed, MR, Goumans, MJ, Klautz, RJ, Poelmann, RE, Gittenberger-de Groot, AC. Normal and abnormal development of the aortic wall and valve: correlation with clinical entities. Neth Heart J. 2014; 22: 363–9.Google Scholar
Fernández, B, Durán, AC, Fernández-Gallego, T, Fernández, MC, Such, M, Arqué, JM, Sans-Coma, V. Bicuspid aortic valves with different spatial orientations of the leaflets are distinct etiological entities. J Am Coll Cardiol. 2009; 54: 2312–18.Google Scholar
Gittenberger-de Groot, AC, Tennstedt, C, Chaoui, R, Lie-Venema, H, Sauer, U, Poelmann, RE. Ventriculo coronary arterial communications (VCAC) and myocardial sinusoids in hearts with pulmonary artresia with intact ventricular septum: two different diseases. Progr Pediatr Cardiol. 2001; 13: 157–64.Google Scholar
Chaoui, R, Tennstedt, C, Göldner, B, Bollmann, R. Prenatal diagnosis of ventriculo-coronary communications in a second-trimester fetus using transvaginal and transabdominal color Doppler sonography. Ultrasound Obstet Gynecol. 1997; 9: 194–7.Google Scholar
Oosthoek, PW, Wenink, ACG, Macedo, AJ, Gittenberger-de Groot, AC. The parachute-like asymmetric mitral valve and its two papillary muscles. J Thorac Cardiovasc Surg. 1997; 114: 915.Google Scholar
Wu, B, Wang, Y, Lui, W, Langworthy, M, Tompkins, KL, Hatzopoulos, AK, Baldwin, HS, Zhou, B. Nfatc1 coordinates valve endocardial cell lineage development required for heart valve formation. Circ Res. 2011; 109: 183–92.Google Scholar
Lie-Venema, H, Eralp, I, Markwald, RR, van den Akker, NM, Wijffels, MC, Kolditz, DP, et al. Periostin expression by epicardium-derived cells (EPDCs) is involved in the development of the atrioventricular valves and fibrous heart skeleton. Differentiation. 2008; 76: 809–19.Google Scholar
Jongbloed, MR, Vicente Steijn, R, Hahurij, ND, Kelder, TP, Schalij, MJ, Gittenberger-de Groot, AC, Blom, NA. Normal and abnormal development of the cardiac conduction system; implications for conduction and rhythm disorders in the child and adult. Differentiation. 2012; 84: 131–48.Google Scholar
Haïssaguerre, M, Jaïs, P, Shah, DC, Takahashi, A, Hocini, M, Quiniou, G, Garrigue, S, Le Mouroux, A, Le Métayer, P, Clémenty, J. Spontaneous initiation of atrial fibrillation by ectopic beats originating in the pulmonary veins. New Engl J Med. 1998; 339: 659–66.Google Scholar
Syeda, F, Kirchhof, P, Fabritz, L. PITX2-dependent gene regulation in atrial fibrillation and rhythm control. J Physiol. 2017; 595: 4019–26.Google Scholar

References

van der Linde, D, Konings, EE, Slager, MA, Witsenburg, M, Helbing, WA, Takkenberg, JJ, Roos-Hesselink, JW. Birth prevalence of congenital heart disease worldwide: a systematic review and meta-analysis. J Am Coll Cardiol. 2011; 58: 2241–7.Google Scholar
Ransom, J, Srivastava, D. The genetics of cardiac birth defects. Semin Cell Dev Biol. 2007; 18: 132–9.Google Scholar
Hoffman, JI, Kaplan, S. The incidence of congenital heart disease. J Am Coll Cardiol. 2002; 39: 1890–900.Google Scholar
Triedman, JK, Newburger, JW. Trends in Congenital Heart Disease, the next decade. Circulation. 2016; 133: 2716–33.Google Scholar
Huang, JB, Liu, YL, Sun, PW, Lv, XD, Du, M, Fan, XM. Molecular mechanisms of congenital heart disease. Cardiovasc Pathol. 2010; 19: e183–93.Google Scholar
Cai, GJ, Sun, XX, Zhang, L, Hong, Q. Association between maternal body mass index and congenital heart defects in offspring: a systematic review. Am J Obstet Gynecol. 2014; 211: 91117.Google Scholar
Botto, LD, Panichello, JD, Brown, ML, Krikov, S, Feldkamp, ML, Lammer, E, et al. Congenital heart defects after maternal fever. Am J Obstet Gynecol. 2014; 210: e1–359. e11.Google Scholar
Jenkins, KJ, Correa, A, Feinstein, JA, Botto, L, Britt, AE, Daniels, SR, et al. Noninherited risk factors and congenital cardiovascular defects: current knowledge: a scientific statement from the American Heart Association Council on Cardiovascular Disease in the Young: endorsed by the American Academy of Pediatrics. Circulation. 2007; 115: 29953014.Google Scholar
Zhu, H, Kartiko, S, Finnell, RH. Importance of gene-environment interactions in the etiology of selected birth defects. Clin Genet. 2009; 75: 409–23.Google Scholar
Nora, JJ. Multifactorial inheritance hypothesis for the etiology of congenital heart diseases. The genetic-environmental interaction. Circulation. 1968; 38: 604–17.Google Scholar
Schott, JJ, Benson, DW, Basson, CT, Pease, W, Silberbach, GM, Moak, JP, et al. Congenital heart disease caused by mutations in the transcription factor NKX2–5. Science. 1998; 281: 108–11.Google Scholar
Gebbia, M, Ferrero, GB, Pilia, G, Bassi, MT, Aylsworth, A, Penman-Splitt, M, et al. X-linked situs abnormalities result from mutations in ZIC3. Nat Genet. 1997; 17: 305–8.Google Scholar
Gong, W, Gottlieb, S, Collins, J, Blescia, A, Dietz, H, Goldmuntz, E, et al. Mutation analysis of TBX1 in non-deleted patients with features of DGS/VCFS or isolated cardiovascular defects. J Med Genet. 2001; 38: E45.Google Scholar
Garg, V, Kathiriya, IS, Barnes, R, Schluterman, MK, King, IN, Butler, CA, et al. GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5. Nature. 2003; 424: 443–7.Google Scholar
Pizzuti, A, Sarkozy, A, Newton, AL, Conti, E, Flex, E, Digilio, MC, et al. Mutations of ZFPM2/FOG2 gene in sporadic cases of tetralogy of Fallot. Hum Mutat. 2003; 22: 372–7.Google Scholar
Sperling, S, Grimm, CH, Dunkel, I, Mebus, S, Sperling, HP, Ebner, A, et al. Identification and functional analysis of CITED2 mutations in patients with congenital heart defects. Hum Mutat. 2005; 26: 575–82.Google Scholar
Reamon-Buettner, SM, Ciribilli, Y, Inga, A, Borlak, J. A loss-of-function mutation in the binding domain of HAND1 predicts hypoplasia of the human hearts. Hum Mol Genet. 2008; 17: 1397–405.Google Scholar
Wang, B, Yan, J, Peng, Z, Wang, J, Liu, S, Xie, X, Ma, X. Teratocarcinoma-derived growth factor 1 (TDGF1) sequence variants in patients with congenital heart defect. Int J Cardiol. 2011; 146: 225–7.CrossRefGoogle ScholarPubMed
Kosaki, R, Gebbia, M, Kosaki, K, Lewin, M, Bowers, P, Towbin, JA, Casey, B. Left-right axis malformations associated with mutations in ACVR2B, the gene for human activin receptor type IIB. Am J Med Genet. 1999; 82: 70–6.Google Scholar
Kosaki, K, Bassi, MT, Kosaki, R, Lewin, M, Belmont, J, Schauer, G, Casey, B. Characterization and mutation analysis of human LEFTY A and LEFTY B, homologues of murine genes implicated in left-right axis development. Am J Hum Genet. 1999; 64: 712–21.Google Scholar
Bamford, RN, Roessler, E, Burdine, RD, Saplakoğlu, U, dela Cruz, J, Splitt, M, et al. Loss-of-function mutations in the EGF-CFC gene CFC1 are associated with human left-right laterality defects. Nat Genet. 2000; 26: 365–9.Google Scholar
Garg, V, Muth, AN, Ransom, JF, Schluterman, MK, Barnes, R, King, IN, et al. Mutations in NOTCH1 cause aortic valve disease. Nature. 2005; 437: 270–4.Google Scholar
Robinson, SW, Morris, CD, Goldmuntz, E, Reller, MD, Jones, MA, Steiner, RD, Maslen, CL. Missense mutations in CRELD1 are associated with cardiac atrioventricular septal defects. Am J Hum Genet. 2003; 72: 1047–52.Google Scholar
Karkera, JD, Lee, JS, Roessler, E, Banerjee-Basu, S, Ouspenskaia, MV, Mez, J, et al. Loss-of-function mutations in growth differentiation factor-1 (GDF1) are associated with congenital heart defects in humans. Am J Hum Genet. 2007; 81: 987–94.Google Scholar
Mohapatra, B, Casey, B, Li, H, Ho-Dawson, T, Smith, L, Fernbach, SD, et al. Identification and functional characterization of NODAL rare variants in heterotaxy and isolated cardiovascular malformations. Hum Mol Genet. 2009; 18: 861–71.Google Scholar
Britz-Cunningham, SH, Shah, MM, Zuppan, CW, Fletcher, WH. Mutations of the Connexin43 gap-junction gene in patients with heart malformations and defects of laterality. N Engl J Med. 1995; 332: 1323–9.Google Scholar
Li, DY, Toland, AE, Boak, BB, Atkinson, DL, Ensing, GJ, Morris, CA, Keating, MT. Elastin point mutations cause an obstructive vascular disease, supravalvular aortic stenosis. Hum Mol Genet. 1997; 6: 1021–8.Google Scholar
Muncke, N, Jung, C, Rüdiger, H, Ulmer, H, Roeth, R, Hubert, A, et al. Missense mutations and gene interruption in PROSIT240, a novel TRAP240-like gene, in patients with congenital heart defect (transposition of the great arteries). Circulation. 2003; 108: 2843–50.Google Scholar
Thienpont, B, Zhang, L, Postma, AV, Breckpot, J, Tranchevent, LC, Van Loo, P, et al. Haploinsufficiency of TAB2 causes congenital heart defects in humans. Am J Hum Genet. 2010; 86: 839–49.Google Scholar
Burn, J, Brennan, P, Little, J, Holloway, S, Coffey, R, Somerville, J, et al. Recurrence risks in offspring of adults with major heart defects: results from first cohort of British collaborative study. Lancet. 1998; 351: 311–16.Google Scholar
Grobman, W, Pergament, E. Isolated hypoplastic left heart syndrome in three siblings. Obstet Gynecol. 1996; 88: 673–5.Google Scholar
Pease, WE, Nordenberg, A, Ladda, RL. Familial atrial septal defect with prolonged atrioventricular conduction. Circulation. 1976; 53: 759–62.Google Scholar
Ferencz, C, Boughman, JA, Neill, CA, Brenner, JI, Perry, LW. Congenital cardiovascular malformations: questions on inheritance. Baltimore-Washington Infant Study Group. J Am Coll Cardiol. 1989; 14: 756–63.Google Scholar
Corone, P, Bonaiti, C, Feingold, J, Fromont, S, Berthet-Bondet, D. Familial congenital heart disease: how are the various types related? Am J Cardiol. 1983; 51: 942–5.Google Scholar
Wessels, MW, Berger, RM, Frohn-Mulder, IM, Roos-Hesselink, JW, Hoogeboom, JJ, Mancini, GS, et al. Autosomal dominant inheritance of left ventricular outflow tract obstruction. Am J Med Genet A. 2005; 134A: 171–9.Google Scholar
Musewe, NN, Alexander, DJ, Teshima, I, Smallhorn, JF, Freedom, RM. Echocardiographic evaluation of the spectrum of cardiac anomalies associated with Trisomy 13 and Trisomy 18. J Am Coll Cardiol. 1990; 15: 673–7.Google Scholar
van Egmond, H, Orye, E, Praet, M, Coppens, M, Devloo-Blancquaert, A. Hypoplastic left heart syndrome and 45X karyotype. Br Heart J. 1988; 60: 6971.Google Scholar
van Bon, BW, Mefford, HC, Menten, B, Koolen, DA, Sharp, AJ, Nillesen, WM, et al. Further delineation of the 15q13 microdeletion and duplication syndromes: a clinical spectrum varying from non-pathogenic to a severe outcome. J Med Genet. 2009; 46: 511–23.Google Scholar
Tartaglia, M, Mehler, EL, Goldberg, R, Zampino, G, Brunner, HG, Kremer, H, et al. Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome. Nat Genet. 2001; 29: 465–8.Google Scholar
Zhao, Y, Ransom, JF, Li, A, Vedantham, V, von Drehle, M, Muth, AN, et al. Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA-1-2. Cell. 2007; 129: 303–17.Google Scholar
Hearn, T, Renforth, GL, Spalluto, C, Hanley, NA, Piper, K, Brickwood, S, et al. Mutation of ALMS1, a large gene with a tandem repeat encoding 47 amino acids, causes Alstrom syndrome. Nat Genet. 2002; 31: 7983.Google Scholar
Oda, T, Elkahloun, AG, Pike, BL, Okajima, K, Krantz, ID, Genin, A, et al. Mutations in the human Jagged1 gene are responsible for Alagille syndrome. Nat Genet. 1997; 16: 235–42.Google Scholar
Newbury-Ecob, RA, Leanage, R, Raeburn, JA, Young, ID. Holt-Oram syndrome: a clinical genetic study. J Med Genet. 1996; 33: 300–7.Google Scholar
Brassington, AM, Sung, SS, Toydemir, RM, Le, T, Roeder, AD, Rutherford, AE, et al. Expressivity of Holt-Oram syndrome is not predicted by TBX5 genotype. Am J Hum Genet. 2003; 73: 7485.Google Scholar
McElhinney, DB, Geiger, E, Blinder, J, Benson, DW, Goldmuntz, E. NKX2.5 mutations in patients with congenital heart disease. J Am Coll Cardiol. 2003; 42: 1650–5.Google Scholar
Carey, AH, Kelly, D, Halford, S, Wadey, R, Wilson, D, Goodship, J, et al. Molecular genetic study of the frequency of monosomy 22q11 in DiGeorge syndrome. Am J Hum Genet. 1992; 51: 964–70.Google Scholar
Mefford, HC, Sharp, AJ, Baker, C, Itsara, A, Jiang, Z, Buysse, K, et al. Recurrent rearrangements of chromosome 1q21.1 and variable pediatric phenotypes. N Engl J Med. 2008; 359: 1685–99.Google Scholar
Hillman, K, DeVita, M, Bellomo, R, Chen, J. Meta-analysis for rapid response teams. Arch Intern Med. 2010; 170: 996–7; author reply 997.Google Scholar
D’Amours, G, Kibar, Z, Mathonnet, G, Fetni, R, Tihy, F, Désilets, V, et al. Whole-genome array CGH identifies pathogenic copy number variations in fetuses with major malformations and a normal karyotype. Clin Genet. 2011; 81: 128–41.Google Scholar
Lazier, J, Fruitman, D, Lauzon, J, Bernier, F, Argiropoulos, B, Chernos, J, et al. Prenatal Array Comparative Genomic Hybridization in Fetuses With Structural Cardiac Anomalies. J Obstet Gynaecol Can. 2016; 38: 619–26.Google Scholar
Lander, ES, Linton, LM, Birren, B, Nusbaum, C, Zody, MC, Baldwin, J, et al. Initial sequencing and analysis of the human genome. Nature. 2001; 409: 860921.Google Scholar
Snyder, M, Du, J, Gerstein, M. Personal genome sequencing: current approaches and challenges. Genes Dev. 2010; 24: 423–31.Google Scholar

References

McGovern, E, Sands, AJ. Perinatal management of major congenital heart disease. Ulster Med J. 2014; 83: 135–9.Google Scholar
Jorgensen, M, McPherson, E, Zaleski, C, Shivaram, P, Cold, C. Stillbirth: the heart of the matter. Am J Med Genet A. 2014; 164A: 691–9.Google Scholar
Dolk, H, Loane, M, Garne, E, European Surveillance of Congenital Anomalies (EUROCAT) Working Group. Congenital heart defects in Europe: prevalence and perinatal mortality, 2000 to 2005. Circulation. 2011; 123: 841–9.Google Scholar
Hoffman, JI, Kaplan, S. The incidence of congenital heart disease. J Am Coll Cardiol. 2002; 39: 1890–900.Google Scholar
Chaix, MA, Andelfinger, G, Khairy, P. Genetic testing in congenital heart disease: a clinical approach. World J Cardiol. 2016; 8: 180–91.Google Scholar
Russell, MW, Chung, WK, Kaltman, JR, Miller, TA. Advances in the Understanding of the Genetic Determinants of Congenital Heart Disease and Their Impact on Clinical Outcomes. J Am Heart Assoc. 2018; 7: e006906.Google Scholar
Weissberg, P (ed.). Children and Young People Statistics 2013. London: British Heart Foundation, 2013.Google Scholar
Fahed, AC, Nemer, GM. Genetic Causes of Syndromic and Non-syndromic Congenital Heart Disease. In Cooper, D and Chen, J-M, eds., Mutations in Human Genetic Disease. London: IntechOpen, 2012.Google Scholar
Fahed, AC, Gelb, BD, Seidman, JG, Seidman, CE. Genetics of congenital heart disease: the glass half empty. Circ Res. 2013; 112: 707–20.Google Scholar
Leatherbury, L, Berul, CI. Genetics of congenital heart disease: is the glass now half-full? Circ Cardiovasc Genet. 2017; 10: e001746.Google Scholar
Waardenberg, AJ, Ramialison, M, Bouveret, R, Harvey, RP. Genetic networks governing heart development. Cold Spring Harb Perspect Med. 2014; 4: a013839.Google Scholar
Moon, A. Mouse models of congenital cardiovascular disease. Curr Top Dev Biol. 2008; 84: 171248.Google Scholar
Dickinson, ME, Flenniken, AM, Ji, X, Teboul, L, Wong, MD, White, JK, et al. High-throughput discovery of novel developmental phenotypes. Nature. 2016; 537: 508514.Google Scholar
Wilson, R, Geyer, SH, Reissig, L, Rose, J, Szumska, D, Hardman, E, et al. Highly variable penetrance of abnormal phenotypes in embryonic lethal knockout mice. Wellcome Open Res. 2016; 1: 1.Google Scholar
Bellmann, K, Perrot, A, Rickert-Sperling, S. Human Genetics of Ventricular Septal Defect. In Rickert-Sperling, S, Kelly, R, Driscoll, D, eds., Congenital Heart Diseases: The Broken Heart. Vienna: Springer, 2016.Google Scholar
Penny, DJ, Vick, GW 3rd. Ventricular septal defect. Lancet. 2011; 377: 1103–12.Google Scholar
Schoenwolf, GC, Bleyl, S, Brauer, P, Francis-West, P. Larsen’s Human Embryology, 5th edn. Philadelphia: Churchill-Livingstone, 2014.Google Scholar
Geyer, SH, Reissig, L, Rose, J, Wilson, R, Prin, F, Szumska, D, et al. A staging system for correct phenotype interpretation of mouse embryos harvested on embryonic day 14 (E14.5). J Anat. 2017; 230: 710–19.Google Scholar
Geyer, SH, Reissig, LF, Hüsemann, M, Höfle, C, Wilson, R, Prin, F, et al. Morphology, topology and dimensions of the heart and arteries of genetically normal and mutant mouse embryos at stages S21-S23. J Anat. 2017; 231: 600614.Google Scholar
Webb, G, Gatzoulis, MA. Atrial septal defects in the adult: recent progress and overview. Circulation. 2006; 114: 1645–53.Google Scholar
Khan, R, Jay, PY. Human Genetics of Atrial Septal Defect. In Rickert-Sperling, S, Kelly, R, Driscoll, D, eds., Congenital Heart Diseases: The Broken Heart. Vienna: Springer, 2016.Google Scholar
Lin, CJ, Lin, CY, Chen, CH, Zhou, B, Chang, CP. Partitioning the heart: mechanisms of cardiac septation and valve development. Development. 2012; 139: 3277–99.Google Scholar
Anderson, RH, Webb, S, Brown, NA, Lamers, W, Moorman, A. Development of the heart: (3) formation of the ventricular outflow tracts, arterial valves, and intrapericardial arterial trunks. Heart. 2003; 89: 1110–18.Google Scholar
Shaheen, R, Rahbeeni, Z, Alhashem, A, Faqeih, E, Zhao, Q, Xiong, Y, et al. Neu-Laxova syndrome, an inborn error of serine metabolism, is caused by mutations in PHGDH. Am J Hum Genet. 2014; 94: 898904.Google Scholar
Acuna-Hidalgo, R, Schanze, D, Kariminejad, A, Nordgren, A, Kariminejad, MH, Conner, P, et al. Neu-Laxova syndrome is a heterogeneous metabolic disorder caused by defects in enzymes of the L-serine biosynthesis pathway. Am J Hum Genet. 2014; 95: 285–93.Google Scholar
Martin, PS, Kloesel, B, Norris, RA, Lindsay, M, Milan, D, Body, SC. Embryonic Development of the Bicuspid Aortic Valve. J Cardiovasc Dev Dis. 2015; 2: 248–72.Google Scholar
Mathieu, P, Bossé, Y, Huggins, GS, Della Corte, A, Pibarot, P, Michelena, HI, et al. The pathology and pathobiology of bicuspid aortic valve: State of the art and novel research perspectives. J Pathol Clin Res. 2015; 1: 195206.Google Scholar
Combs, MD, Yutzey, KE. Heart valve development: regulatory networks in development and disease. Circ Res. 2009; 105: 408–21.Google Scholar
Freeze, SL, Landis, BJ, Ware, SM, Helm, BM. Bicuspid aortic valve: a review with recommendations for genetic counseling. J Genet Couns. 2016; 25: 1171–8.Google Scholar
Hinton, RB, Martin, LJ, Rame-Gowda, S, Tabangin, ME, Cripe, LH, Benson, DW. Hypoplastic left heart syndrome links to chromosomes 10q and 6q and is genetically related to bicuspid aortic valve. J Am Coll Cardiol. 2009; 53: 1065–71.Google Scholar

References

Senat, MV, Deprest, J, Boulvain, M, Paupe, A, Winer, N, Ville, Y. Endoscopic laser surgery versus serial amnioreduction for severe twin-to-twin transfusion syndrome. N Engl J Med. 2004; 351: 136144.Google Scholar
Szwast, A, Tian, Z, McCann, M, Donaghue, D, Rychik, J. Vasoreactive response to maternal hyperoxygenation in the fetus with hypoplastic left heart syndrome. Circ Cardiovasc Imaging. 2010; 3: 172–8.Google Scholar
Schidlow, DN, Donofrio, MT. Prenatal maternal hyperoxygenation testing and implications for critical care delivery planning among fetuses with congenital heart disease: early experience. Am J Perinatol. 2018; 35: 1623.Google Scholar
Kohl, T. Chronic intermittent materno-fetal hyperoxygenation in late gestation may improve on hypoplastic cardiovascular structures associated with cardiac malformations in human fetuses. Pediatr Cardiol. 2010; 31: 250–63.Google Scholar
Zeng, S, Zhou, J, Peng, Q, Deng, W, Zhang, M, Zhao, Y, et al. Sustained maternal hyperoxygenation improves aortic arch dimensions in fetuses with coarctation. Sci Rep. 2016; 6: 39304.Google Scholar
Zeng, S, Zhou, J, Peng, Q, Deng, W, Zang, M, Wang, T, Zhou, Q. Sustained chronic maternal hyperoxygenation increases myocardial deformation in fetuses with a small aortic isthmus at risk for coarctation. J Am Soc Echocardiogr. 2017; 30; 9921000.Google Scholar
Maxwell, D, Allan, L, Tynan, MJ. Balloon dilatation of the aortic valve in the fetus: a report of two cases. Br Heart J. 1991; 65: 256–8.Google Scholar
Delius, RE, Rademecker, MA, de Leval, MR, Elliott, MJ, Stark, J. Is a high-risk biventricular repair always preferable to conversion to a single ventricle repair? J Thorac Cardiovasc Surg. 1996; 112: 1561–8; discussion 1568–9.Google Scholar
Burch, M, Kaufman, L, Archer, N, Sullivan, I. Persistent pulmonary hypertension late after neonatal aortic valvotomy: a consequence of an expanded surgical cohort. Heart. 2004; 90: 918920.Google Scholar
Emani, SM, Bacha, EA, McElhinney, DB, Marx, GR, Tworetzky, W, Pigula, FA, del Nido, PJ. Primary left ventricular rehabilitation is effective in maintaining two-ventricle physiology in the borderline left heart. J Thorac Cardiovasc Surg. 2009; 138: 1276–82.Google Scholar
Tulzer, G, Arzt, W, Franklin, RC, Loughna, PV, Mair, R, Gardiner, HM. Pulmonary valvuloplasty for critical pulmonary stenosis or atresia with intact septum. Lancet. 2002; 360: 1567–8.Google Scholar
Tworetzky, W, McElhinney, DB, Marx, GR, Benson, CB, Brusseau, R, Morash, D, et al. In utero valvuloplasty for pulmonary atresia with hypoplastic right ventricle: techniques and outcomes. Pediatrics. 2009; 124; e510–18.Google Scholar
Marshall, AC, van der Velde, ME, Tworetzky, W, Gomez, CA, Wilkins-Haug, L, Benson, CB, et al. Creation of an atrial septal defect in utero for fetuses with hypoplastic left heart syndrome and intact or highly restrictive atrial septum. Circulation. 2004; 110: 253–8.Google Scholar
Rychik, J, Rome, JJ, Collins, MH, DeCampli, WM, Spray, TL. The hypoplastic left heart syndrome with intact atrial septum: atrial morphology, pulmonary vascular histopathology and outcome. J Am Coll Cardiol. 1999; 34: 554–60.Google Scholar
Taylor, PV, Scott, JS, Gerlis, LM, Esscher, E, Scott, O. Maternal antibodies against fetal cardiac antigens in congenital complete heart block. N Engl J Med. 1986; 315: 667–72.Google Scholar
Eliasson, H, Sonesson, SE, Sharland, G, Granath, F, Simpson, JM, Carvalho, JS, et al. Isolated atrioventricular block in the fetus: a retrospective, multinational, multicenter study of 175 patients. Circulation. 2011; 124: 1919–26.Google Scholar
Lopes, LM, Tavares, GM, Damiano, AP, Lopes, MA, Aiello, VD, Schultz, R, Zugaib, M. Perinatal outcome of fetal atrioventricular block: one-hundred-sixteen cases from a single institution. Circulation. 2008; 118: 1268–75.Google Scholar
Jaeggi, ET, Fouron, JC, Silverman, ED, Ryan, G, Smallhorn, J, Hornberger, LK. Transplacental fetal treatment improves the outcome of prenatally diagnosed complete atrioventricular block without structural heart disease. Circulation. 2004; 110: 1542–8.Google Scholar
Carpenter, RJ Jr., Strasburger, JF, Garson, A Jr., Smith, RT, Deter, RL, Engelhardt, HT Jr. Fetal ventricular pacing for hydrops secondary to complete atrioventricular block. J Am Coll Cardiol. 1986; 8: 1434–6.Google Scholar
Walkinshaw, SA, Welch, CR, McCormack, J, Walsh, K. In utero pacing for fetal congenital heart block. Fetal Diagn Ther. 1994; 9: 183–5.Google Scholar
Assad, RS, Zielinsky, P, Kalil, R, Lima, G, Aramayo, A, Santos, A, et al. New lead for in utero pacing for fetal congenital heart block. J Thorac Cardiovasc Surg. 2003; 126: 300–2.Google Scholar
Bar-Cohen, Y, Loeb, GE, Pruetz, JD, Silka, MJ, Guerra, C, Vest, AN, Zhou, L, Chmait, RH. Preclinical testing and optimization of a novel fetal micropacemaker. Heart Rhythm. 2015; 12: 1683–90.Google Scholar
Co-Vu, J, Lopez-Colon, D, Vyas, HV, Weiner, N, DeGroff, C. Maternal hyperoxygenation: a potential therapy for congenital heart disease in the fetuses? A systematic review of the current literature. Echocardiography. 2017; 34: 1822–33.Google Scholar
Kovacevic, A, Öhman, A, Tulzer, G, Herberg, U, Dangel, J, Carvalho, JS, et al. Fetal hemodynamic response to aortic valvuloplasty and postnatal outcome: a European multicenter study. Ultrasound Obstet Gynecol. 2018; 52: 221–9.Google Scholar
Gardiner, HM, Kovacevic, A, Tulzer, G, Sarkola, T, Herberg, U, Dangel, J, et al. Natural history of 107 cases of fetal aortic stenosis from a European multicenter retrospective study. Ultrasound Obstet Gynecol. 2016; 48: 373–81.Google Scholar
Di Donato, RM, Jonas, RA, Lang, P, Rome, JJ, Mayer, JE Jr., Castaneda, AR. Neonatal repair of tetralogy of Fallot with and without pulmonary atresia. J Thorac Cardiovasc Surg. 1991; 101: 126–37.Google Scholar
Freud, LR, McElhinney, DB, Marshall, AC, Marx, GR, Friedman, KG, del Nido, PJ, et al. Fetal aortic valvuloplasty for evolving hypoplastic left heart syndrome: postnatal outcomes of the first 100 patients. Circulation. 2014; 130: 638–45.Google Scholar
Axt-Fliedner, R, Kreiselmaier, P, Schwarze, A, Krapp, M, Gembruch, U. Development of hypoplastic left heart syndrome after diagnosis of aortic stenosis in the first trimester by early echocardiography. Ultrasound Obstet Gynecol. 2006; 28: 106–9.Google Scholar
Freud, LR, Moon-Grady, A, Escobar-Diaz, MC, Gotteiner, NL, Young, LT, McElhinney, DB, Tworetzky, W. Low rate of prenatal diagnosis among neonates with critical aortic stenosis: insight into the natural history in utero. Ultrasound Obstet Gynecol. 2015; 45: 326–32.Google Scholar
Matsui, H, Gardiner, HM. Fetal intervention for cardiac disease: the cutting edge of perinatal care. Semin Fetal Neonatal Med. 2007; 12: 482–9.Google Scholar
Reich, O, Tax, P, Marek, J, Rázek, V, Gilík, J, Tomek, V, et al. Long term results of percutaneous balloon valvuloplasty of congenital aortic stenosis: independent predictors of outcome. Heart. 2004; 90: 7076.Google Scholar
McElhinney, DB, Lock, JE, Keane, JF, Moran, AM, Colan, SD. Left heart growth, function, and reintervention after balloon aortic valvuloplasty for neonatal aortic stenosis. Circulation. 2005; 111: 451–8.Google Scholar
Ashburn, DA, McCrindle, BW, Tchervenkov, CI, Jacobs, ML, Lofland, GK, Bove, EL, et al. Outcomes after the Norwood operation in neonates with critical aortic stenosis or aortic valve atresia. J Thorac Cardiovasc Surg. 2003; 125: 1070–82.Google Scholar
Jacobs, JP, O’Brien, SM, Pasquali, SK, Jacobs, ML, Lacour-Gayet, FG, Tchervenkov, CI, et al. Variation in outcomes for benchmark operations: an analysis of the Society of Thoracic Surgeons Congenital Heart Surgery Database. Ann Thorac Surg. 2011; 92: 2184–91; discussion 2191–2.Google Scholar
Rasiah, SV, Ewer, AK, Miller, P, Wright, JG, Barron, DJ, Brawn, WJ, Kilby, MD. Antenatal perspective of hypoplastic left heart syndrome: 5 years on. Arch Dis Child Fetal Neonatal Ed. 2008; 93: F192–7.Google Scholar
Daubeney, PE, Wang, D, Delany, DJ, Keeton, BR, Anderson, RH, Slavik, Z, Flather, M, Webber, SA, UK and Ireland Collaborative Study of Pulmonary Atresia with Intact Ventricular Septum. UK and Ireland collaborative study of pulmonary atresia with intact ventricular septum. J Thorac Cardiovasc Surg. 2005; 130: 1071.Google Scholar
Baba, K, Kotani, Y, Chetan, D, Chaturvedi, RR, Lee, KJ, Benson, LN, et al. Hybrid versus Norwood strategies for single-ventricle palliation. Circulation. 2012; 126 (Suppl. 1): S123–31.Google Scholar
Kovacevic, A, Roughton, M, Mellander, M, Öhman, A, Tulzer, G, Dangel, J, et al. Fetal aortic valvuloplasty: investigating institutional bias in surgical decision-making. Ultrasound Obstet Gynecol. 2014; 44: 538–44.Google Scholar
Arzt, W, Wertaschnigg, D, Veit, I, Klement, F, Gitter, R, Tulzer, G. Intrauterine aortic valvuloplasty in fetuses with critical aortic stenosis: Experience and results of 24 procedures. Ultrasound Obstet Gynecol. 2011; 37: 689–95.Google Scholar
McElhinney, DB, Marshall, AC, Wilkins-Haug, LE, Brown, DW, Benson, CB, Silva, V, et al. Predictors of technical success and postnatal biventricular outcome after in utero aortic valvuloplasty for aortic stenosis with evolving hypoplastic left heart syndrome. Circulation. 2009; 120: 1482–90.Google Scholar
Jaeggi, E, Renaud, C, Ryan, G, Chaturvedi, R. Intrauterine therapy for structural congenital heart disease: Contemporary results and Canadian experience. Trends Cardiovasc Med. 2016; 26: 639–46.Google Scholar
Pedra, SR, Peralta, CF, Crema, L, Jatene, IB, da Costa, RN, Pedra, CA. Fetal interventions for congenital heart disease in Brazil. Pediatr Cardiol. 2014; 35: 399405.Google Scholar
Galindo, A, Gómez-Montes, E, Gómez, O, Bennasar, M, Crispi, F, Herraiz, I, et al. Fetal aortic valvuloplasty: experience and results of two tertiary centers in Spain. Fetal Diagn Ther. 2017; 42: 262–70.Google Scholar
Moon-Grady, AJ, Morris, SA, Belfort, M, Chmait, R, Dangel, J, Devlieger, R, et al. International Fetal Cardiac Intervention Registry: A Worldwide Collaborative Description and Preliminary Outcomes. J Am Coll Cardiol. 2015; 66: 388–99.Google Scholar
Hunter, LE, Chubb, H, Miller, O, Sharland, G, Simpson, JM. Fetal aortic valve stenosis: a critique of case selection criteria for fetal intervention. Prenat Diagn. 2015; 35: 1176–81.Google Scholar
Roman, KS, Fouron, JC, Nii, M, Smallhorn, JF, Chaturvedi, R, Jaeggi, ET. Determinants of outcome in fetal pulmonary valve stenosis or atresia with intact ventricular septum. Am J Cardiol. 2007; 99: 699703.Google Scholar
Gardiner, HM, Belmar, C, Tulzer, G, Barlow, A, Pasquini, L, Carvalho, JS, et al. Morphological and functional predictors of eventual circulation in the fetus with pulmonary atresia or critical pulmonary stenosis with intact septum. J Am Coll Cardiol. 2008; 51: 1299–308.Google Scholar
Tulzer, A, Arzt, W, Gitter, R, Prandstetter, C, Grohmann, E, Mair, R, Tulzer, G. Immediate effects and outcomes after in-utero pulmonary valvuloplasty in fetuses with pulmonary atresia with intact septum or critical pulmonary stenosis. Ultrasound Obstet Gynecol. 2018; 52: 230–7.Google Scholar
Tworetzky, W, McElhinney, DB, Marx, GR, Benson, CB, Brusseau, R, Morash, D, et al. In utero valvuloplasty for pulmonary atresia with hypoplastic right ventricle: techniques and outcomes. Pediatrics. 2009; 124: e510–18.Google Scholar
Lara, DA, Morris, SA, Maskatia, SA, Challman, M, Nguyen, M, Feagin, DK, et al. Pilot study of chronic maternal hyperoxygenation and effect on aortic and mitral valve annular dimensions in fetuses with left heart hypoplasia. Ultrasound Obstet Gynecol. 2016; 48: 365–72.Google Scholar
Wohlmuth, C, Wertaschnigg, D, Wieser, I, Arzt, W, Tulzer, G. Tissue Doppler imaging in fetuses with aortic stenosis and evolving hypoplastic left heart syndrome before and after fetal aortic valvuloplasty. Ultrasound Obstet Gynecol. 2016; 47: 608–15.Google Scholar
Tworetzky, W, Wilkins-Haug, L, Jennings, RW, van der Velde, ME, Marshall, AC, Marx, GR, et al. Balloon dilation of severe aortic stenosis in the fetus: potential for prevention of hypoplastic left heart syndrome: candidate selection, technique, and results of successful intervention. Circulation. 2004; 110: 2125–31.Google Scholar
Donofrio, MT, Moon-Grady, AJ, Hornberger, LK, Copel, JA, Sklansky, MS, Abuhamad, A, et al. Diagnosis and treatment of fetal cardiac disease: a scientific statement from the American Heart Association. Circulation. 2014; 129: 2183–242.Google Scholar
Jouannic, JM, Boudjemline, Y, Benifla, JL, Bonnet, D. Transhepatic ultrasound-guided cardiac catheterization in the fetal lamb. Circulation. 2005; 111: 736–41.Google Scholar
Kohl, T, Müller, A, Tchatcheva, K, Achenbach, S, Gembruch, U. Fetal transesophageal echocardiography: clinical introduction as a monitoring tool during cardiac intervention in a human fetus. Ultrasound Obstet Gynecol. 2005; 26: 780–5.Google Scholar

References

Gaynor, JW, Nord, AS, Wernovsky, G, Bernbaum, J, Solot, CB, Burnham, N, Zackai, E, Heagerty, PJ, Clancy, RR, Nicolson, SC, Jarvik, GP, Gerdes, M. Apolipoprotein E genotype modifies the risk of behavior problems after infant cardiac surgery. Pediatrics. 2009; 124: 241250.Google Scholar
Bellinger, DC, Newburger, JW, Wypij, D, Kuban, KC, duPlessis, AJ, Rappaport, LA. Behaviour at eight years in children with surgically corrected transposition: the Boston Circulatory Arrest Trial. Cardiol Young. 2009; 19: 8697.Google Scholar
Bellinger, DC, Wypij, D, duPlessis, AJ, Rappaport, LA, Jonas, RA, Wernovsky, G, Newburger, JW. Neurodevelopmental status at eight years in children with dextro-transposition of the great arteries: the Boston Circulatory Arrest Trial. J Thorac Cardiovasc Surg. 2003; 126: 1385–96.Google Scholar
Wernovsky, G. Current insights regarding neurological and developmental abnormalities in children and young adults with complex congenital cardiac disease. Cardiol Young. 2006; 16 (Suppl. 1): 92104.Google Scholar
Yi, JJ, Tang, SX, McDonald-McGinn, DM, Calkins, ME, Whinaa, DA, Souders, MC, Zackai, EH, Goldmuntz, E, Gaynor, JW, Gur, RC, Emanuel, BS, Gur, RE. Contribution of congenital heart disease to neuropsychiatric outcome in school-age children with 22q11.2 deletion syndrome. Am J Med Genet. 2013; 165: 137–47.Google Scholar
Alsaied, T, Marino, BS, Esbensen, AJ, Anixt, JS, Epstein, JN, Cnota, JF. Does congenital heart disease affect neurodevelopmental outcomes in children with Down syndrome? Congenit Heart Dis. 2016; 11: 2633.Google Scholar
Visootsak, J, Mahle, WT, Kirshbom, PM, Huddleston, L, Caron-Besch, M, Ransom, M, Sherman, SL. Neurodevelopmental outcomes in children with Down syndrome and congenital heart defects. Am J Med Genet. 2011; 155: 2688–91.Google Scholar
Takashima, S, Becker, LE, Armstrong, DL, Chan, F. Abnormal neuronal development in the visual cortex of the human fetus and infant with Down’s syndrome. A quantitative and qualitative Golgi study. Brain Res. 1981; 225: 121.Google Scholar
Robin, NH, Taylor, CJ, McDonald-McGinn, DM, Zackai, EH, Bingham, P, Collins, KJ, et al. Polymicrogyria and deletion 22q11.2 sundrome: window to the etiology of a common cortical malformation. Am J Med Genet. 2006; 140: 2416–25.Google Scholar
Newburger, JW, Jonas, RA, Wernovsky, G, Wypij, D, Hickey, PR, Kuban, K, Farrell, DM, Holmes, GL, Helmers, SL, Constantinou, J, Carrazana, E. A comparison of the perioperative neurologic effects of hypothermic circulatory arrest versus low-flow cardiopulmonary bypass in infant heart surgery. New Engl J Med. 1993; 329: 1057–64.Google Scholar
Bellinger, DC, Jonas, RA, Rappaport, LA, Wypij, D, Wernovsky, G, Kuban, KC, Barnes, PD, Holmes, GL, Hickey, PR, Strand, RD, Walsh, AZ. Developmental and neurologic status of children after heart surgery with hypothermic circulatory arrest or low-flow cardiopulmonary bypassNew Engl J Med. 1995; 332: 549–55.Google Scholar
Bellinger, DC, Wypij, D, Rivkin, MJ, DeMaso, DR, Robertson, RL, Dunbar-Masterson, C, Rappaport, LA, Wernovsky, G, Jonas, RA, Newburger, JW. Adolescents with d-transposition of the great arteries corrected with the arterial switch procedure: neuropsychological assessment and structural brain imaging. Circulation. 2011; 124: 1361–69.Google Scholar
Wypij, D, Jonas, RA, Bellinger, DC, Del Nido, P, Mayer, JE, Bacha, EA, Forbess, JM, Pigula, F, Laussen, PC, Newburger, JW. The effect of hematocrit during hypothermic cardiopulmonary bypass in infant heart surgery: results from the combined Boston hematocrit trials. J Thorac Cardiovasc Surg. 2008; 135: 355–60.Google Scholar
Gaynor, JW, Stopp, C, Wypij, D, Andropoulos, DB, Atallah, J, Atz, AM, et al. Neurodevelopmental outcomes after cardiac surgery in infancy. Pediatrics. 2015; 135: 816–25.Google Scholar
Marino, BS, Lipkin, PH, Newburger, JW, Peacock, G, Gerdes, M, Gaynor, JW, et al. Neurodevelopmental outcomes in children with congenital heart disease: evaluation and management: a scientific statement from the American Heart Association. Circulation. 2012; 126: 1143–72.Google Scholar
Glauer, TA, Rorke, LB, Weinberg, PM, Clancy, RR. Acquired neuropathologic lesions associated with the hypoplastic left heart syndrome. Pediatrics. 1990; 85: 9911000.Google Scholar
Kinney, HC, Panigrahy, A, Newburger, JW, Jonas, RA, Sleeper, LA. Hypoxic-ischemic brain injury in infants with congenital heart disease dying after cardiac surgery. Acta Neuropathol. 2005; 110: 563–78.Google Scholar
Mahle, WT, Tavani, F, Zimmerman, RA, Nicolson, SC, Galli, KK, Gaynor, JW, et al. An MRI study of neurological injury before and after congenital heart surgery. Circulation. 2002; 106: I109–14.Google Scholar
Miller, SP, McQuillen, PS, Hamrick, S, Xu, D, Glidden, DV, Charlton, N, Karl, T, Azakie, A, Ferriero, DM, Barkovich, AJ, Vigneron, DB. Abnormal brain development in newborns with congenital heart disease. New Engl J Med. 2007; 357: 1928–38.Google Scholar
Peyvandi, S, Chau, V, Guo, T, Xu, D, Glass, HC, Synnes, A, Poskitt, K, Barkovich, AJ, Miller, SP, McQuillen, PS. Neonatal brain injury and timing of neurodevelopmental assessment in patients with congenital heart disease. J Am Coll Cardiol. 2018; 71: 1986–96.Google Scholar
Licht, DJ, Shera, DM, Clancy, RR, Wernovsky, G, Montenegro, LM, Nicolson, SC, Zimmerman, RA, Spray, TL, Gaynor, JW, Vossough, A. Brain maturation is delayed in infants with complex congenital heart defects. J Thorac Cardiovasc Surg. 2009; 137: 529–37.Google Scholar
Limperopoulos, C, Majnemer, A, Shevell, MI, Rosenblatt, B, Rohlicek, C, Tchervenkov, C. Neurologic status of newborns with congenital heart defects before open heart surgery. Pediatrics. 1999; 103: 402–8.Google Scholar
Birca, A, Vakorin, VA, Porayette, P, Madathil, S, Chau, V, Seed, M, et al. Interplay of brain structure and function in neonatal congenital heart disease. Ann Clin Transl Neurol. 2016; 3: 708–22.Google Scholar
Dimitropoulos, A, McQuillen, PS, Sethi, V, Moosa, A, Chau, V, Xu, D, Brant, R, Azakie, A, Campbell, A, Barkovich, AJ, Poskitt, KJ. Brain injury and development in newborns with critical congenital heart disease. Neurology. 2013; 81: 241–8.Google Scholar
Volpe, JJ. Encephalopathy of congenital heart disease – destructive and developmental effects intertwined. J Pediatr. 2014; 164: 962–5.Google Scholar
Hinton, RB, Andelfinger, G, Sekar, P, Hinton, AC, Gendron, RL, Michelfelder, EC, Robitaille, Y, Benson, DW. Prenatal head growth and white matter injury in hypoplastic left heart syndrome. Pediatr Res. 2008; 64: 364.Google Scholar
Limperopoulos, C, Tworetzky, W, McElhinney, DB, Newburger, JW, Brown, DW, Robertson, RL, et al. Brain volume and metabolism in fetuses with congenital heart disease: evaluation with quantitative magnetic resonance imaging and spectroscopy. Circulation. 2010; 121: 2633.Google Scholar
Schellen, C, Ernst, S, Gruber, GM, Mlczoch, E, Weber, M, Brugger, PC, Ulm, B, Langs, G, Salzer-Muhar, U, Prayer, D, Kasprian, G. Fetal MRI detects early alterations of brain development in Tetralogy of Fallot. Am J Obstet Gynecol. 2015; 213: 392.e1–7.Google Scholar
Jorgensen, DS, Tabor, A, Rode, L, Dyre, L, Ekelund, CK, Helmuth, SG, et al. Longitudinal brain and body growth in normal fetuses and fetuses with transposition of the great arteries – a quantitative volumetric magnetic resonance imaging study. Circulation. 2018; 138: 1368–70.Google Scholar
Rudolph, AM. Congenital Diseases of the Heart: Clinical-Physiologic Considerations in Diagnosis and Management. Chicago: Year Book Medical Publishers, 1974.Google Scholar
Prsa, M, Sun, L, van Amerom, J, Yoo, SJ, Grosse-Wortmann, L, Jaeggi, E, Macgowan, C, Seed, M. Reference ranges of blood flow in the major vessels of the normal human fetal circulation at term by phase contrast magnetic resonance imaging. Circ Cardiovasc Imaging. 2014; 7: 663–70.Google Scholar
Sun, L, Macgowan, CK, Portnoy, S, Sled, JG, Yoo, SJ, Grosse‐Wortmann, L, Jaeggi, E, Kingdom, J, Seed, M. New advances in fetal cardiovascular magnetic resonance imaging for quantifying the distribution of blood flow and oxygen transport: potential applications in fetal cardiovascular disease diagnosis and therapy. Echocardiography. 2017; 34: 1799–803.Google Scholar
Sun, L, Macgowan, CK, Sled, JG, Yoo, SJ, Manlhiot, C, Porayette, P, Grosse-Wortmann, L, Jaeggi, E, McCrindle, BW, Hickey, E, Miller, S, Seed, M. Reduced fetal cerebral oxygen consumption is associated with smaller brain size in fetuses with congenital heart disease. Circulation. 2015; 131: 1313–23.Google Scholar
Jones, HN, Olbrych, SK, Smith, KL, Cnota, JF, Habli, M, Ramos-Gonzales, O, Owens, KJ, Hinton, AC, Polzin, WJ, Muglia, LJ, Hinton, RB. Hypoplastic left heart syndrome is associated with structural and vascular placental abnormalities and leptin dysregulation. Placenta. 2015; 36: 1078–86.Google Scholar
Donofrio, MT, Bremer, YA, Schieken, RM, Gennings, C, Morton, LD, Eidem, BW, Cetta, F, Falkensammer, CB, Huhta, JC, Kleinman, CS. Autoregulation of cerebral blood flow in fetuses with congenital heart disease: the brain sparing effect. Pediatr Cardiol. 2003; 24: 436–43.Google Scholar
Cohn, HE, Sacks, EJ, Heymann, MA, Rudolph, AM. Cardiovascular responses to hypoxemia and acidemia in fetal lambs. Am J Obstet Gynecol. 1974; 120: 817–24.Google Scholar
Wladimiroff, JW, Tonge, HM, Stewart, PA. Doppler ultrasound assessment of cerebral blood flow in the human fetus. BJOG. 1986; 93: 471–5.Google Scholar
Pearce, W. Hypoxic regulation of the fetal cerebral circulation. J App Physiol. 2006; 100: 731–8.Google Scholar
Wheaton, WW, Chandel, NS. Hypoxia. 2. Hypoxia regulates cellular metabolism. Am J Physiol-Cell Physiol. 2010; 300: C385–93.Google Scholar
Yuen, TJ, Silbereis, JC, Griveau, A, Chang, SM, Daneman, R, Fancy, SP, Zahed, H, Maltepe, E, Rowitch, DH. Oligodendrocyte-encoded HIF function couples postnatal myelination and white matter angiogenesis. Cell. 2014; 158: 383–96.Google Scholar
Tolcos, M, Bateman, E, O’Dowd, R, Markwick, R, Vrijsen, K, Rehn, A, Rees, S. Intrauterine growth restriction affects the maturation of myelin. Exp Neurol. 2011; 232: 5365.Google Scholar
Morton, PD, Korotcova, L, Lewis, BK, Bhuvanendran, S, Ramachandra, SD, Zurakowski, D, Zhang, J, Mori, S, Frank, JA, Jonas, RA, Gallo, V, Ishibashi, N. Abnormal neurogenesis and cortical growth in congenital heart disease. Sci Transl Med. 2017; 9: 7029.Google Scholar
Fowden, AL, Giussani, DA, Forhead, AJ. Endocrine and metabolic programming during intrauterine development. Early Hum Dev. 2005; 81: 723–34.Google Scholar
Rees, S, Harding, R, Walker, D. The biological basis of injury and neuroprotection in the fetal and neonatal brain. Int J Dev Neurosci. 2011; 29: 551–63.Google Scholar
Rollins, CK, Asaro, LA, Akhondi-Asl, A, Kussman, BD, Rivkin, MJ, Bellinger, DC, Warfield, SK, Wypij, D, Newburger, JW, Soul, JS. White matter volume predicts language development in congenital heart disease. J Pediatr. 2017; 181: 42–8.Google Scholar
von Rhein, M, Buchmann, A, Hagmann, C, Huber, R, Klaver, P, Knirsch, W, Latal, B. Brain volumes predict neurodevelopment in adolescents after surgery for congenital heart disease. Brain. 2013; 137: 268–76.Google Scholar
Rivkin, MJ, Watson, CG, Scoppettuolo, LA, Wypij, D, Vajapeyam, S, Bellinger, DC, DeMaso, DR, Robertson, RL Jr., Newburger, JW. Adolescents with D-transposition of the great arteries repaired in early infancy demonstrate reduced white matter microstructure associated with clinical risk factors. J Thorac Cardiovasc Surg. 2013; 146: 543–9.Google Scholar
Panigrahy, A, Schmithorst, VJ, Wisnowski, JL, Watson, CG, Bellinger, DC, Newburger, JW, Rivkin, MJ. Relationship of white matter network topology and cognitive outcome in adolescents with d-transposition of the great arteries. NeuroImage: Clinical. 2015; 7: 438–48.Google Scholar
Ibuki, K, Watanabe, K, Yoshimura, N, Kakimoto, T, Matsui, M, Yoshida, T, Origasa, H, Ichida, F. The improvement of hypoxia correlates with neuroanatomic and developmental outcomes: comparison of midterm outcomes in infants with transposition of the great arteries or single-ventricle physiology. J Thorac Cardiovasc Surg. 2012; 143: 1077–85.Google Scholar
Homsy, J, Zaidi, S, Shen, Y, Ware, JS, Samocha, KE, Karczewski, KJ, et al. De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies. Science. 2015; 350: 1262–6.Google Scholar
Forbess, JM, Visconti, KJ, Hancock-Friesen, C, Howe, RC, Bellinger, DC, Jonas, RA. Neurodevelopmental outcome after congenital heart surgery: results from an institutional registry. Circulation. 2002; 106: I95102.Google Scholar
Laraja, K, Sadhwani, A, Tworetzky, W, Marshall, AC, Gauvreau, K, Freud, L, Hass, C, Dunbar-Masterson, C, Ware, J, Lafranchi, T, Wilkins-Haug, L. Neurodevelopmental outcome in children after fetal cardiac intervention for aortic stenosis with evolving hypoplastic left heart syndrome. J Pediatr. 2017; 184: 130–6.Google Scholar
Porayette, P, Madathil, S, Sun, L, Jaeggi, E, Grosse‐Wortmann, L, Yoo, SJ, Hickey, E, Miller, SP, Macgowan, CK, Seed, M. MRI reveals hemodynamic changes with acute maternal hyperoxygenation in human fetuses with and without congenital heart disease. Prenat Diag. 2016 ; 36: 274–81.Google Scholar
Kohl, T. Chronic intermittent materno-fetal hyperoxygenation in late gestation may improve on hypoplastic cardiovascular structures associated with cardiac malformations in human fetuses. Ped Cardiol. 2010; 31: 250–63.Google Scholar
Lara, DA, Morris, SA, Maskatia, SA, Challman, M, Nguyen, M, Feagin, DK, Schoppe, L, Zhang, J, Bhatt, A, Sexson‐Tejtel, SK, Lopez, KN. Pilot study of chronic maternal hyperoxygenation and effect on aortic and mitral valve annular dimensions in fetuses with left heart hypoplasia. Ultrasound Obstet Gynecol. 2016; 48: 365–72.Google Scholar
Zeng, S, Zhou, J, Peng, Q, Deng, W, Zhang, M, Zhao, Y, Wang, T, Zhou, Q. Sustained maternal hyperoxygenation improves aortic arch dimensions in fetuses with coarctation. Sci Rep. 2016; 6: 39304.Google Scholar
Lara, DA, Morris, SA, Maskatia, SA, Karlsten, M, Nguyen, MJ, Schoppe, L, et al. The effect of maternal hyperoxygenation on cerebral and placental vasoregulation in the fetus with left heart hypoplasia. J Am Soc Echo. 2015; 28: B92.Google Scholar
Accurso, FJ, Alpert, B, Wilkening, RB, Petersen, RG, Meshia, G. Time-dependent response of fetal pulmonary blood flow to an increase in fetal oxygen tension. Resp Physiol. 1986; 63: 4352.Google Scholar
Szwast, A, Putt, M, Gaynor, JW, Licht, D, Rychik, J. Cerebrovascular response to maternal hyperoxygenation (MH) in fetuses with hypoplastic left heart syndrome (HLHS) depends upon gestational age (GA) and baseline cerebrovascular resistance. Ultrasound Obstet Gynecol. 2017; 52: 473–8.Google Scholar
da Fonseca, EB, Bittar, RE, Carvalho, MH, Zugaib, M. Prophylactic administration of progesterone by vaginal suppository to reduce the incidence of spontaneous preterm birth in women at increased risk: a randomized placebo-controlled double-blind study. Am J Obstet Gynecol. 2003; 188: 419–24.Google Scholar
Stein, DG, Wright, DW, Kellermann, AL. Does progesterone have neuroprotective properties? Ann Emerg Med. 2008; 51: 164–72.Google Scholar
Partridge, EA, Davey, MG, Hornick, MA, McGovern, PE, Mejaddam, AY, Vrecenak, JD, et al. An extra-uterine system to physiologically support the extreme premature lamb. Nat Comm. 2017; 8: 15112.Google Scholar
Usuda, H, Watanabe, S, Miura, Y, Saito, M, Musk, GC, Rittenschober-Böhm, J, Ikeda, H, Sato, S, Hanita, T, Matsuda, T, Jobe, AH. Successful maintenance of key physiological parameters in preterm lambs treated with ex vivo uterine environment therapy for a period of 1 week. Am J Obstet Gynecol. 2017; 217: 457–e1.Google Scholar
Jenkins, KJ, Gauvreau, K, Newburger, JW, Spray, TL, Moller, JH, Iezzoni, LI. Consensus-based method for risk adjustment for surgery for congenital heart disease. J Thoracic Cardiovasc Surg. 2002; 123: 110–18.Google Scholar
Peyvandi, S, De Santiago, V, Chakkarapani, E, Chau, V, Campbell, A, Poskitt, KJ, Xu, D, Barkovich, AJ, Miller, S, McQuillen, P. Association of prenatal diagnosis of critical congenital heart disease with postnatal brain development and the risk of brain injury. JAMA Pediatr. 2016; 170: e154450.Google Scholar

References

Naheed, ZJ, Strasburger, JF, Deal, BJ, Benson, DW Jr., Gidding, SS. Fetal tachycardia: mechanisms and predictors of hydrops fetalis. J Am Coll Cardiol. 1996; 27: 1736–40.Google Scholar
Artman, M, Coetzee, W. Developmental regulation of cardiac ion channels. In Zipes, D, Jalife, J, eds., Cardiac Electrophysiology: From Cell to Bedside. Philadelphia, PA: Saunders Elsevier, 2009, pp. 157–68.Google Scholar
Creazzo, T. Functional developmental biology of the myocardium. In Loewy Kirby, M, ed., Cardiac Development. New York: Oxford University Press, 2007, pp. 5368.Google Scholar
Friedman, WF. The intrinsic physiologic properties of the developing heart. Prog Cardiovasc Dis. 1972; 15: 87111.Google Scholar
Romero, T, Covell, J, Friedman, WF. A comparison of pressure-volume relations of the fetal, newborn, and adult heart. Am J Physiol. 1972; 222: 1285–90.Google Scholar
Schmidt, MR, Kristiansen, SB, White, P, Smerup, M, Bøtker, HE, Vogel, M, Hjortdal, V, Sørensen, K, Redington, A. Glucose-insulin infusion improves cardiac function during fetal tachycardia. J Am Coll Cardiol. 2004; 43: 445–52.Google Scholar
Vanoli, E, Cerati, D, Pedretti, RF. Autonomic control of heart rate: pharmacological and nonpharmacological modulation. Basic Res Cardiol. 1998; 93 (Suppl. 1): 133–42.Google Scholar
Rudolph, AM, Heymann, MA. Cardiac output in the fetal lamb: the effects of spontaneous and induced changes of heart rate on right and left ventricular output. Am J Obstet Gynecol. 1976; 124: 183–92.Google Scholar
Reed, KL, Appleton, CP, Anderson, CF, Shenker, L, Sahn, DJ. Doppler studies of vena cava flows in human fetuses. Insights into normal and abnormal cardiac physiology. Circulation. 1990; 81: 498505.Google Scholar
Brace, RA. Effects of outflow pressure on fetal lymph flow. Am J Obstet Gynecol. 1989; 160: 494–7.Google Scholar
Rudolph, A. The fetal circulation and postnatal adaptation. In Rudolph, A, ed., Congenital Diseases of the Heart. Armonk, NY: Future Publishing Company, 2001, pp. 344Google Scholar
Schmidt, KG, Ulmer, HE, Silverman, NH, Kleinman, CS, Copel, JA. Perinatal outcome of fetal complete atrioventricular block: a multicenter experience. J Am Coll Cardiol. 1991; 17: 1360–6.Google Scholar
Simpson, JM, Sharland, GK. Fetal tachycardias: management and outcome of 127 consecutive cases. Heart. 1998; 79: 576–81.Google Scholar
Jaeggi, E, Fouron, JC, Drblik, SP. Fetal atrial flutter: diagnosis, clinical features, treatment, and outcome. J Pediatr. 1998; 132: 335–9.Google Scholar
Jaeggi, ET, Carvalho, JS, De Groot, E, Api, O, Clur, SA, Rammeloo, L, McCrindle BW, , Ryan, G, Manlhiot, C, Blom, NA. Comparison of transplacental treatment of fetal supraventricular tachyarrhythmias with digoxin, flecainide, and sotalol: results of a nonrandomized multicenter study. Circulation. 2011; 124: 1747–54.Google Scholar
Wacker-Gussmann, A, Strasburger, JF, Srinivasan, S, Cuneo, BF, Lutter, W, Wakai, RT. Fetal atrial flutter: electrophysiology and associations with rhythms involving an accessory pathway. J Am Heart Assoc. 2016; 5: e003673.Google Scholar
Kleinman, CS, Donnerstein, RL, Jaffe, CC, DeVore, GR, Weinstein, EM, Lynch, DC, Talner, NS, Berkowitz, RL, Hobbins, JC. Fetal echocardiography. A tool for evaluation of in utero cardiac arrhythmias and monitoring of in utero therapy: analysis of 71 patients. Am J Cardiol. 1983; 51: 237–43.Google Scholar
Jaeggi, E, Fouron, JC, Fournier, A, van Doesburg, N, Drblik, SP, Proulx, F. Ventriculo-atrial time interval measured on M mode echocardiography: a determining element in diagnosis, treatment, and prognosis of fetal supraventricular tachycardia. Heart. 1998; 79: 582–7.Google Scholar
Fouron, JC, Fournier, A, Proulx, F, Lamarche, J, Bigras, JL, Boutin, C, Brassard, M, Gamache, S. Management of fetal tachyarrhythmia based on superior vena cava/aorta Doppler flow recordings. Heart. 2003; 89: 1211–16.Google Scholar
Donofrio, MT, Moon-Grady, AJ, Hornberger, LK, Copel, JA, Sklansky, MS, Abuhamad, A, et al. Diagnosis and treatment of fetal cardiac disease: a scientific statement from the American Heart Association. Circulation. 2014; 129: 2183–242.Google Scholar
Kleinman, CS, Copel, JA, Weinstein, EM, Santulli, TV Jr., Hobbins, JC. Treatment of fetal supraventricular tachyarrhythmias. J Clin Ultrasound. 1985; 13: 265–73.Google Scholar
van Engelen, AD, Weijtens, O, Brenner, JI, Kleinman, CS, Copel, JA, Stoutenbeek, P, Meijboom, EJ. Management outcome and follow-up of fetal tachycardia. J Am Coll Cardiol. 1994; 24: 1371–5.Google Scholar
Frohn-Mulder, IM, Stewart, PA, Witsenburg, M, Den Hollander, NS, Wladimiroff, JW, Hess, J. The efficacy of flecainide versus digoxin in the management of fetal supraventricular tachycardia. Prenat Diagn. 1995; 15: 1297–302.Google Scholar
Ebenroth, ES, Cordes, TM, Darragh, RK. Second-line treatment of fetal supraventricular tachycardia using flecainide acetate. Pediatr Cardiol. 2001; 22: 483–7.Google Scholar
Jouannic, JM, Delahaye, S, Fermont, L, Le Bidois, J, Villain, E, Dumez, Y, Dommerques, M. Fetal supraventricular tachycardia: a role for amiodarone as second-line therapy? Prenat Diagn. 2003; 23: 152–6.Google Scholar
Sridharan, S, Sullivan, I, Tomek, V, Wolfenden, J, Škovranek, J, Yates, R, Janoušek, J, Dominguez, TE, Marek, J. Flecainide versus digoxin for fetal supraventricular tachycardia: comparison of two drug treatment protocols. Heart Rhythm. 2016; 13: 1913–19.Google Scholar
Ekman-Joelsson, BM, Mellander, M, Lagnefeldt, L, Sonesson, SE. Foetal tachyarrhythmia treatment remains challenging even if the vast majority of cases have a favourable outcome. Acta Paediatr. 2015; 104: 1090–7.Google Scholar
Hill, GD, Kovach, JR, Saudek, DE, Singh, AK, Wehrheim, K, Frommelt, MA. Transplacental treatment of fetal tachycardia: a systematic review and meta-analysis. Prenat Diagn. 2017; 37: 1076–83.Google Scholar
Allan, LD, Chita, SK, Sharland, GK, Maxwell, D, Priestley, K. Flecainide in the treatment of fetal tachycardias. Br Heart J. 1991; 65: 46–8.Google Scholar
Barjot, P, Hamel, P, Calmelet, P, Maragnes, P, Herlicoviez, M. Flecainide against fetal supraventricular tachycardia complicated by hydrops fetalis. Acta Obstet Gynecol Scand. 1998; 77: 353–8.Google Scholar
Strizek, B, Berg, C, Gottschalk, I, Herberg, U, Geipel, A, Gembruch, U. High-dose flecainide is the most effective treatment of fetal supraventricular tachycardia. Heart Rhythm. 2016; 13: 1283–8.Google Scholar
Ekiz, A, Kaya, B, Bornaun, H, Acar, DK, Avci, ME, Bestel, A, Yildirim, G. Flecainide as first-line treatment for fetal supraventricular tachycardia. J Matern Fetal Neonatal Med. 2018: 31: 407–12.Google Scholar
Sonesson, SE, Fouron, JC, Wesslen-Eriksson, E, Jaeggi, E, Winberg, P. Foetal supraventricular tachycardia treated with sotalol. Acta Paediatr. 1998; 87: 584–7.Google Scholar
Oudijk, MA, Michon, MM, Kleinman, CS, Kapusta, L, Stoutenbeek, P, Visser, GH, Meijboom, EJ. Sotalol in the treatment of fetal dysrhythmias. Circulation. 2000; 101: 2721–6.Google Scholar
Oudijk, MA, Ruskamp, JM, Ververs, FF, Ambachtsheer, EB, Stoutenbeek, P, Visser, GH, Meijboom, EJ. Treatment of fetal tachycardia with sotalol: transplacental pharmacokinetics and pharmacodynamics. J Am Coll Cardiol. 2003; 42: 765–70.Google Scholar
Shah, A, Moon-Grady, A, Bhogal, N, Collins, KK, Tacy, T, Brook, M, Hornberger, LK. Effectiveness of sotalol as first-line therapy for fetal supraventricular tachyarrhythmias. Am J Cardiol. 2012; 109: 1614–18.Google Scholar
van der Heijden, LB, Oudijk, MA, Manten, GT, ter Heide, H, Pistorius, L, Freund, MW. Sotalol as first-line treatment for fetal tachycardia and neonatal follow-up. Ultrasound Obstet Gynecol. 2013; 42: 285–93.Google Scholar
Uzun, O, Babaoglu, K, Sinha, A, Massias, S, Beattie, B. Rapid control of foetal supraventricular tachycardia with digoxin and flecainide combination treatment. Cardiol Young. 2012; 22: 372–80.Google Scholar
Strasburger, JF, Cuneo, BF, Michon, MM, Gotteiner, NL, Deal, BJ, McGregor, SN, Oudijk, MA, Meijboom, EJ, Feinkind, L, Hussey, M, Parilla, BV. Amiodarone therapy for drug-refractory fetal tachycardia. Circulation. 2004; 109: 375–9.Google Scholar
Krapp, M, Baschat, AA, Gembruch, U, Geipel, A, Germer, U. Flecainide in the intrauterine treatment of fetal supraventricular tachycardia. Ultrasound Obstet Gynecol. 2002; 19: 158–64.Google Scholar
Hansmann, M, Gembruch, U, Bald, R, Manz, M, Redel, DA. Fetal tachyarrhythmias: transplacental and direct treatment of the fetus – a report of 60 cases. Ultrasound Obstet Gynecol. 1991; 1: 162–8.Google Scholar
Parilla, BV, Strasburger, JF, Socol, ML. Fetal supraventricular tachycardia complicated by hydrops fetalis: a role for direct fetal intramuscular therapy. Am J Perinatol. 1996; 13: 483–6.Google Scholar
Moatassim, S, Touleimat, S, Hazelzet, T, Brasseur, MD, Diguet, A, Durand, I, Verspyck, E. Maternal complications induced by digoxin treatment of fetal tachycardia: a retrospective series of 18 cases. J Gynecol Obstet Hum Reprod. 2018; 47: 35–8.Google Scholar
Bourget, P, Pons, JC, Delouis, C, Fermont, L, Frydman, R. Flecainide distribution, transplacental passage, and accumulation in the amniotic fluid during the third trimester of pregnancy. Ann Pharmacother. 1994; 28: 1031–4.Google Scholar
Hopson, JR, Buxton, AE, Rinkenberger, RL, Nademanee, K, Heilman, JM, Kienzle, MG. Safety and utility of flecainide acetate in the routine care of patients with supraventricular tachyarrhythmias: results of a multicenter trial. The Flecainide Supraventricular Tachycardia Study Group. Am J Cardiol. 1996; 77: 72A82A.Google Scholar
Hohnloser, SH, Woosley, RL. Sotalol. N Engl J Med. 1994; 331: 31–8.Google Scholar
Peralta, AO, John, RM, Gaasch, WH, Taggart, PI, Martin, DT, Venditti, FJ. The class III antiarrhythmic effect of sotalol exerts a reverse use-dependent positive inotropic effect in the intact canine heart. J Am Coll Cardiol. 2000; 36: 1404–10.Google Scholar
Arnoux, P, Seyral, P, Llurens, M, Djiane, P, Potier, A, Unal, D, Cano, JP, Serradimigni, A, Rouault, F. Amiodarone and digoxin for refractory fetal tachycardia. Am J Cardiol. 1987; 59: 166–7.Google Scholar
Gembruch, U, Manz, M, Bald, R, Rüddel, H, Redel, DA, Schlebusch, H, Nitsch, J, Hansmann, M. Repeated intravascular treatment with amiodarone in a fetus with refractory supraventricular tachycardia and hydrops fetalis. Am Heart J. 1989; 118: 1335–8.Google Scholar
Bartalena, L, Bogazzi, F, Braverman, LE, Martino, E. Effects of amiodarone administration during pregnancy on neonatal thyroid function and subsequent neurodevelopment. J Endocrinol Invest. 2001; 24: 116–30.Google Scholar
Magee, LA, Nulman, I, Rovet, JF, Koren, G. Neurodevelopment after in utero amiodarone exposure. Neurotoxicol Teratol. 1999; 21: 261–5.Google Scholar
Vanbesien, J, Casteels, A, Bougatef, A, De Catte, L, Foulon, W, De Bock, S, Smitz, J, De Schepper, J. Transient fetal hypothyroidism due to direct fetal administration of amiodarone for drug resistant fetal tachycardia. Am J Perinatol. 2001; 18: 113–16.Google Scholar
Gembruch, U, Hansmann, M, Redel, DA, Bald, R. Intrauterine therapy of fetal tachyarrhythmias: intraperitoneal administration of antiarrhythmic drugs to the fetus in fetal tachyarrhythmias with severe hydrops fetalis. J Perinat Med. 1988; 16: 3944.Google Scholar
Gembruch, U, Manz, M, Bald, R, Ruddel, H, Redel, DA, Schlebusch, H, Nitsch, J, Hansmann, M. Repeated intravascular treatment with amiodarone in a fetus with refractory supraventricular tachycardia and hydrops fetalis. Am Heart J. 1989; 118: 1335–8.Google Scholar

References

Glickstein, JS, Buyon, J, Friedman, D. Pulsed Doppler echocardiographic assessment of the fetal PR interval. Am J Cardiol. 2000; 86: 236–9.Google Scholar
Fouron, JC, Fournier, A, Proulx, F, et al. Management of fetal tachyarrhythmia based on superior vena cava/aorta Doppler flow recordings. Heart. 2003; 89: 1211–16.Google Scholar
Carvalho, JS, Prefumo, F, Ciardelli, V, et al. Evaluation of fetal arrhythmias from simultaneous pulsed wave Doppler in pulmonary artery and vein. Heart. 2007; 93: 1448–53.Google Scholar
Wacker-Gussmann, A, Plankl, C, Sewald, M, et al. Fetal cardiac time intervals in healthy pregnancies – an observational study by fetal ECG (Monica Healthcare System). J Perin Medicine. 2017; 46: 587–92.Google Scholar
Wacker-Gussmann, A, Strasburger, JF, Srinivasan, S, et al. Fetal atrial flutter: electrophysiology and associations with rhythms involving an accessory pathway. J Am Heart Assoc. 2016; 5: e003673.Google Scholar
Eswaran, H, Escalona-Vargas, D, Bolin, EH, et al. Fetal magnetocardiography using optically pumped magnetometers: a more adaptable and less expensive alternative? Prenat Diagn. 2017; 37: 193–6.Google Scholar
Rasiah, SV, Ewer, AK, Miller, P, et al. Prenatal diagnosis, management and outcome of fetal dysrhythmia: a tertiary fetal medicine centre experience over an eight-year period. Fetal DiagnTher. 2011; 30: 122–7.Google Scholar
Fouron, JC. Fetal arrhythmias: the Saint-Justine hospital experience. Prenat Diagn. 2004; 24: 1068–80.Google Scholar
Cuneo, BF, Strasburger, JF, Wakai, RT, et al. Conduction system disease in fetuses evaluated for irregular cardiac rhythm. Fetal Diagn Ther. 2006; 21: 307–13.Google Scholar
Srinivasan, S, Strasburger, J. Overview of fetal arrhythmias. Curr Opin Pediatr. 2008; 20: 522–31.Google Scholar
Eliasson, H, Wahren-Herlenius, M, Sonesson, SE. Mechanisms in fetal bradyarrhythmia: 65 cases in a single center analyzed by Doppler flow echocardiographic techniques. Ultrasound Obstet Gynecol. 2011; 37:172–8.Google Scholar
Wiggins, DL, Strasburger, JF, Gotteiner, NL, et al. Magnetophysiologic and echocardiographic comparison of blocked atrial bigeminy and 2:1 atrioventricular block in the fetus. Heart Rhythm. 2013; 10: 1192–8.Google Scholar
Carvalho, JS. Primary bradycardia: keys and pitfalls in diagnosis. Ultrasound Obstet Gynecol. 2014; 44: 125–30.Google Scholar
Strasburger, JF, Wakai, RT. Fetal cardiac arrhythmia detection and in utero therapy. Nat Rev Cardiol. 2010; 7: 277–90.Google Scholar
Jaeggi, ET, Carvalho, JS, De Groot, E, et al. Comparison of transplacental treatment of fetal supraventricular tachyarrhythmias with digoxin, flecainide, and sotalol: results of a nonrandomized multicenter study. Circulation. 2011; 124: 1747–54.Google Scholar
Uzun, O, Babaoglu, K, Sinha, A, et al. Rapid control of foetal supraventricular tachycardia with digoxin and flecainide combination treatment. Cardiol Young. 2012; 22: 372–80.Google Scholar
Gembruch, U. Fetal Tachyarrhythmia. In Yagel, S, Silverman, NH, Gembruch, U, eds., Fetal Cardiology: Maternal-Fetal Medicine. New York: Informa Healthcare, 2009, pp. 461–81.Google Scholar
Oudijk, MA, Stoutenbeek, P, Sreeram, N, et al. Persistent junctional reciprocating tachycardia in the fetus. J Matern Fetal Neonatal Med. 2003; 13: 191–6.Google Scholar
Zaidi, SJ, Siddiqui, S, Cuneo, BF, et al. Prenatal diagnosis and management of junctional ectopic tachycardia. Heart Rhythm Case Rep. 2017; 3: 503–8.Google Scholar
Dubin, AM, Cuneo, BF, Strasburger, JF, et al. Congenital junctional ectopic tachycardia and congenital complete atrioventricular block: a shared etiology? Heart Rhythm. 2005; 2: 313–15.Google Scholar
Kang, SL, Howe, D, Coleman, M, et al. Foetal supraventricular tachycardia with hydrops fetalis: a role for direct intraperitoneal amiodarone. Cardiol Young. 2015; 25: 447–53.Google Scholar
Shah, A, Moon-Grady, A, Bhogal, N, et al. Effectiveness of sotalol as first-line therapy for fetal supraventricular tachyarrhythmias. Am J Cardiol. 2012; 109: 1614–18.Google Scholar
Sridharan, S, Sullivan, I, Tomek, V, et al. Flecainide versus digoxin for fetal supraventricular tachycardia: comparison of two drug treatment protocols. Heart Rhythm. 2016; 13: 1913–19.Google Scholar
Ekman-Joelsson, BM, Mellander, M, Lagnefeldt, L, et al. Foetal tachyarrhythmia treatment remains challenging even if the vast majority of cases have a favourable outcome. Acta Paediatr. 2015; 104: 1090–7.Google Scholar
Strizek, B, Berg, C, Gottschalk, I, et al. High-dose flecainide is the most effective treatment of fetal supraventricular tachycardia. Heart Rhythm. 2016; 13: 1283–8.Google Scholar
Ekiz, A, Kaya, B, Bornaun, H, et al. Flecainide as first-line treatment for fetal supraventricular tachycardia. J Matern Fetal Neonatal Med. 2018; 31: 407–12.Google Scholar
Hill, GD, Kovach, JR, Saudek, DE, et al. Transplacental treatment of fetal tachycardia: a systematic review and meta-analysis. Prenat Diagn. 2017; 37: 1076–83.Google Scholar
Strasburger, JF, Cuneo, BF, Michon, MM, et al. Amiodarone therapy for drug-refractory fetal tachycardia. Circulation. 2004; 109: 375–9.Google Scholar
Jouannic, JM, Delahaye, S, Fermont, L, et al. Fetal supraventricular tachycardia: a role for amiodarone as second-line therapy? Prenat Diagn. 2003; 23: 152–6.Google Scholar
Parilla, BV, Strasburger, JF, Socol, ML. Fetal supraventricular tachycardia complicated by hydrops fetalis: a role for direct fetal intramuscular therapy. Am J Perinatol. 1996; 13: 483–6.Google Scholar
Vigneswaran, TV, Callaghan, N, Andrews, RE, et al. Correlation of maternal flecainide concentrations and therapeutic effect in fetal supraventricular tachycardia. Heart Rhythm. 2014; 11: 2047–53.Google Scholar
Cuneo, BF, Strasburger, JF. We only find what we look for: fetal heart rate and the diagnosis of long-QT syndrome. Circ Arrhythm Electrophysiol. 2015; 8: 760–2.Google Scholar
Lopes, LM, Tavares, GM, Damiano, AP, et al. Perinatal outcome of fetal atrioventricular block: one-hundred-sixteen cases from a single institution. Circulation. 2008; 118: 1268–75.Google Scholar
Glatz, AC, Gaynor, JW, Rhodes, LA, et al. Outcome of high-risk neonates with congenital complete heart block paced in the first 24 hours after birth. J Thorac Cardiovasc Surg. 2008; 136: 767–73.Google Scholar
Baruteau, AE, Fouchard, S, Behaghel, A, et al. Characteristics and long-term outcome of non-immune isolated atrioventricular block diagnosed in utero or early childhood: a multicentre study. Eur Heart J. 2012; 33: 622–9.Google Scholar
Eliasson, H, Sonesson, SE, Sharland, G, et al. Isolated atrioventricular block in the fetus: a retrospective multinational, multicentre study of 175 patients. Circulation. 2011; 124: 1919–26.Google Scholar
Van Hare, GF. Magnetocardiography in the diagnosis of fetal arrhythmias. Heart Rhythm. 2013; 10: 1199–200.Google Scholar
Buyon, JP, Hiebert, R, Copel, J, et al. Autoimmune-associated congenital heart block: demographics, mortality, morbidity and recurrence rates obtained from a national neonatal lupus registry. J Am Coll Cardiol. 1998; 31: 1658–66.Google Scholar
Levesque, K, Morel, N, Maltret, A, et al. Description of 214 cases of autoimmune congenital heart block: results of the French neonatal lupus syndrome. Autoimmun Rev. 2015; 14: 1154–60.Google Scholar
Friedman, DM, Kim, MY, Copel, JA, et al. Utility of Cardiac Monitoring in Fetuses at Risk for Congenital Heart Block: The PR Interval and Dexamethasone Evaluation (PRIDE) Prospective Study. Circulation. 2008; 117: 485–93.Google Scholar
Jaeggi, E, Laskin, C, Hamilton, R, et al. The importance of the level of maternal anti-Ro/SSA antibodies as a prognostic marker of the development of cardiac neonatal lupus erythematosus a prospective study of 186 antibody-exposed fetuses and infants. J Am Coll Cardiol. 2010; 55: 2778–84.Google Scholar
Hutter, D, Silverman, ED, Jaeggi, ET. The benefits of transplacental treatment of isolated congenital complete heart block associated with maternal anti-Ro/SSA antibodies: a review. Scand J Immunol. 2010; 72: 235–41.Google Scholar
Saxena, A, Izmirly, PM, Mendez, B, et al. Prevention and treatment in utero of autoimmune-associated congenital heart block. Cardiol Rev. 2014; 22: 263–7.Google Scholar
Vest, AN, Zhou, L, Huang, X, et al. Design and testing of a transcutaneous RF recharging system for a fetal micropacemaker. IEEE Trans Biomed Circuits Syst. 2017; 11: 336–46.Google Scholar
Cuneo, BF, Mitchell, MB, Marwan, AI, et al. Ex utero intrapartum treatment to ventricular pacing: a novel delivery strategy for complete atrioventricular block with severe bradycardia. Fetal Diagn Ther. 2017; 42: 311–14.Google Scholar
Izmirly, PM, Costedoat-Chalumeau, N, Pisoni, C, et al. Maternal use of hydroxychloroquine is associated with a reduced risk of recurrent anti-SSA/RO associated cardiac manifestations of neonatal lupus. Circulation. 2012; 126: 7682.Google Scholar
Ciardulli, A, D’Antonio, F, Magro-Malosso, ER, et al. Maternal steroid therapy for fetuses with second-degree immune-mediated congenital atrioventricular block: a systematic review and meta-analysis. Acta Obstet Gynecol Scand. 2018; 97: 787–94.Google Scholar
Van den Berg, NW, Slieker, MG, van Beynum, IM, et al. Fluorinated steroids do not improve outcome of isolated atrioventricular block. Int J Cardiol. 2016; 225: 167–71.Google Scholar
Izmirly, PM, Saxena, A, Sahl, SK, et al. Assessment of fluorinated steroids to avert progression and mortality in anti-SSA/Ro-associated cardiac injury limited to the fetal conduction system. Ann Rheum Dis. 2016; 75: 1161–5.Google Scholar
Cuneo, BF, Ambrose, SE, Tworetzky, W. Detection and successful treatment of emergent anti-SSA-mediated fetal atrioventricular block. Am J Obstet Gynecol. 2016; 215: 27–8.Google Scholar
Cuneo, BF, Zhao, H, Strasburger, JF, et al. Atrial and ventricular rate response and patterns of heart rate acceleration during maternal-fetal terbutaline treatment of fetal complete heart block. Am J Cardiol. 2007; 100: 661–5.Google Scholar
Friedman, DM, Llanos, C, Izmirly, PM, et al. Evaluation of fetuses in a study of intravenous immunoglobulin as preventive therapy for congenital heart block: results of a multicenter, prospective, open-label clinical trial. Arthritis Rheum. 2010; 62: 1138–46.Google Scholar
Pisoni, CN, Brucato, A, Ruffatti, A, et al. Failure of intravenous immunoglobulin to prevent congenital heart block: findings of a multicenter, prospective, observational study. Arthritis Rheum. 2010; 62: 1147–52.Google Scholar
Trucco, SM, Jaeggi, E, Cuneo, B, et al. Use of intravenous gamma globulin and corticosteroids in the treatment of maternal autoantibody-mediated cardiomyopathy. J Am Coll Cardiol. 2011; 57: 715–23.Google Scholar
Ruffatti, A, Cerutti, A, Favaro, M, et al. Plasmapheresis, intravenous immunoglobulins and bethametasone – a combined protocol to treat autoimmune congenital heart block: a prospective cohort study. Clin Exp Rheumatol. 2016; 34: 706–13.Google Scholar
Rein, AJ, Mevorach, D, Perles, Z, et al. Early diagnosis and treatment of atrioventricular block in the fetus exposed to maternal anti-SSA/ RO-SSB/LA antibodies. a prospective, observational, fetal kinetocardiogram-based study. Circulation. 2009; 119: 1867–72.Google Scholar
Jaeggi, ET, Silverman, ED, Laskin, C, et al. Prolongation of the atrioventricular conduction in fetuses exposed to maternal anti-Ro/SSA and anti-La/SSB antibodies did not predict progressive heart block. A prospective observational study on the effects of maternal antibodies on 165 fetuses. J Am Coll Cardiol. 2011; 57: 1487–92.Google Scholar
Bergman, G, Eliasson, H, Bremme, K, et al. Anti-Ro52/SSA antibody-exposed fetuses with prolonged atrioventricular time intervals show signs of decreased cardiac performance. Ultrasound Obstet Gynecol. 2009; 34: 543–9.Google Scholar
Kan, N, Silverman, ED, Kingdom, J, et al. Serial echocardiography for immune-mediated heart disease in the fetus: results of a risk-based prospective surveillance strategy. Prenat Diagn. 2017; 37: 375–82.Google Scholar
Cuneo, BF, Moon-Grady, AJ, Sonesson, SE, et al. Heart sounds at home: feasibility of an ambulatory fetal heart rhythm surveillance program for anti-SSA-positive pregnancies. J Perinatol. 2017; 37: 226–30.Google Scholar

References

Greizerstein, HB. Placental and fetal composition during the last trimester of gestation in the rat. Biol Reprod. 1982; 26: 847–53.Google Scholar
Engle, WA, Lemons, JA. Composition of the fetal and maternal guinea pig throughout gestation. Pediatr Res. 1986; 20: 1156–60.Google Scholar
Hartnoll, G, Betremieux, P, Modi, N. Randomised controlled trial of postnatal sodium supplementation on body composition in 25 to 30 week gestational age infants. Arch Dis Child Fetal Neonatal Ed. 2000; 82: F24–8.Google Scholar
Barker, G, Boyd, RD, D’Souza, SW, et al. Placental water content and distribution. Placenta. 1994; 15: 4756.Google Scholar
Goodwin, JW, Godden, JO, Chance, GW. Perinatal Medicine: The Basic Science Underlying Clinical Practice. Baltimore: The Williams and Wilkins Co, 1976.Google Scholar
Campbell, J, Wathen, N, Macintosh, M, et al. Biochemical composition of amniotic fluid and extraembryonic coelomic fluid in the first trimester of pregnancy. Br J Obstet Gynaecol. 1992; 99: 563–5.Google Scholar
Faber, J, Gault, TJ, Long, LR, Thornburg, KL. Chloride and the generation of amniotic fluid in the early embryo. J Exp Zool. 1973; 183: 343–52.Google Scholar
Gillibrand, PN. Changes in the electrolytes, urea and osmolality of the amniotic fluid with advancing pregnancy. J Obstet Gynaecol Br Commonw. 1969; 76: 898905.Google Scholar
Desai, M, Ladella, S, Ross, MG. Reversal of pregnancy-mediated plasma hypotonicity in the near-term rat. J Matern Fetal Neonatal Med. 2003; 13: 197202.Google Scholar
Cheung, CY, Brace, RA. Amniotic fluid volume and composition in mouse pregnancy. J Soc Gynecol Investig. 2005; 12: 558–62.Google Scholar
Brace, RA, Wolf, EJ. Normal amniotic fluid volume changes throughout pregnancy. Am J Obstet Gynecol. 1989; 161: 382–8.Google Scholar
Gadd, RL. The volume of the liquor amnii in normal and abnormal pregnancies. J Obstet Gynaecol Br Commonw. 1966; 73: 1122.Google Scholar
Beischer, NA, Brown, JB, Townsend, L. Studies in prolonged pregnancy. 3. Amniocentesis in prolonged pregnancy. Am J Obstet Gynecol. 1969; 103: 496503.Google Scholar
Queenan, JT, Von Gal, HV, Kubarych, SF. Amniography for clinical evaluation of erythroblastosis fetalis. Am J Obstet Gynecol. 1968; 102: 264–74.Google Scholar
Sibley, CP, Boyd, DH. Mechanisms of transfer across the human placenta. In Polin, RA, Fox, WW, Abman, S, eds., Fetal and Neonatal Physiology. Philadelphia: WB Saunders, 2006, pp. 111–22.Google Scholar
Stulc, J, Stulcova, B, Sibley, CP. Evidence for active maternal-fetal transport of Na+ across the placenta of the anaesthetized rat. J Physiol. 1993; 470: 637–49.Google Scholar
Faber, JJ, Anderson, DF. Current topic: water volume of the ovine conceptus; point of view. Placenta. 1992; 13: 199212.Google Scholar
Lumbers, ER, Smith, FG, Stevens, AD. Measurement of net transplacental transfer of fluid to the fetal sheep. J Physiol. 1985; 364: 289–99.Google Scholar
Faichney, GJ, Fawcett, AA, Boston, RC. Water exchange between the pregnant ewe, the foetus and its amniotic and allantoic fluids. J Comp Physiol B. 2004; 174: 503–10.Google Scholar
Brace, RA. Progress toward understanding the regulation of amniotic fluid volume: water and solute fluxes in and through the fetal membranes. Placenta. 1995; 16: 118.Google Scholar
Schroder, HJ. Basics of placental structures and transfer functions. In Brace, RA, Ross, MG, Robillard, JE, eds., Fetal & Neonatal Body Fluids. Ithaca: Perinatology Press, 1989, pp. 187226.Google Scholar
Hempstock, J, Bao, YP, Bar-Issac, M, et al. Intralobular differences in antioxidant enzyme expression and activity reflect the pattern of maternal arterial bloodflow within the human placenta. Placenta. 2003; 24: 517–23.Google Scholar
Stulc, J, Stulcova, B. Asymmetrical transfer of inert hydrophilic solutes across rat placenta. Am J Physiol. 1993; 265: R670–5.Google Scholar
Schroder, H, Nelson, P, Power, G. Fluid shift across the placenta: I. The effect of dextran T 40 in the isolated guinea-pig placenta. Placenta. 1982; 3: 327–38.Google Scholar
Hanson, RS, Powrie, RO, Larson, L. Diabetes insipidus in pregnancy: a treatable cause of oligohydramnios. Obstet Gynecol. 1997; 89: 816–17.Google Scholar
Ross, MG, Cedars, L, Nijland, MJ, Ogundipe, A. Treatment of oligohydramnios with maternal 1-deamino-[8-D-arginine] vasopressin-induced plasma hypoosmolality. Am J Obstet Gynecol. 1996; 174: 1608–13.Google Scholar
Ross, MG, Nijland, MJ, Kullama, LK. 1-Deamino-[8-D-arginine] vasopressin-induced maternal plasma hypoosmolality increases ovine amniotic fluid volume. Am J Obstet Gynecol. 1996; 174: 1125–7.Google Scholar
Gizzo, S, Noventa, M, Vitagliano, A, et al. An update on maternal hydration strategies for amniotic fluid improvement in isolated oligohydramnios and normohydramnios: evidence from a systematic review of literature and meta-analysis. PLoS ONE. 2015. 10: e0144334.Google Scholar
Leichtweiss, HP, Schroder, H. The effect of elevated outflow pressure on flow resistance and the transfer of THO, albumin and glucose in the isolated guinea pig placenta. Pflugers Arch. 1977; 371: 251–6.Google Scholar
Brace, RA, Moore, TR. Transplacental, amniotic, urinary, and fetal fluid dynamics during very-large-volume fetal intravenous infusions. Am J Obstet Gynecol. 1991; 164: 907–16.Google Scholar
Brownbill, P, Sibley, CP. Regulation of transplacental water transfer: the role of fetoplacental venous tone. Placenta. 2006; 27: 560–7.Google Scholar
Reynolds, LP, Redmer, DA. Utero-placental vascular development and placental function. J Anim Sci. 1995; 73: 1839–51.Google Scholar
Coan, PM, Ferguson-Smith, AC, Burton, GJ. Developmental dynamics of the definitive mouse placenta assessed by stereology. Biol Reprod. 2004; 70: 1806–13.Google Scholar
Jansson, T, Powell, TL, Illsley, NP. Gestational development of water and non-electrolyte permeability of human syncytiotrophoblast plasma membranes. Placenta. 1999; 20: 155–60.Google Scholar
Faber, JJ, Thornburg, KL. Fetal homeostasis in relation to placental water exchange. Ann Rech Vet. 1977; 8: 353–61.Google Scholar
Jansson, T, Illsley, NP. Osmotic water permeabilities of human placental microvillous and basal membranes. J Membr Biol. 1993; 132: 147–55.Google Scholar
Liu, H, Koukoulas, I, Ross, MC, Wang, S, Wintour, EM. Quantitative comparison of placental expression of three aquaporin genes. Placenta. 2004; 25: 475–8.Google Scholar
Beall, MH, Chaudhri, N, Amidi, F, et al. Increased expression of aquaporins in placenta of the late gestation mouse fetus. J Soc Gynecol Investig. 2005; 12 (Suppl.): 780.Google Scholar
Zhu, X, Jiang, S, Zou, S, Hu, Y, Wang, Y. Expression of aquaporin 3 and aquaporin 9 in placenta and fetal membrane with idiopathic polyhydramnios. Zhonghua Fu Chan Ke Za Zhi. 2009; 144: 920–3.Google Scholar
Zhu, XQ, Jiang, SS, Zhu, XJ, et al. Expression of aquaporin 1 and aquaporin 3 in fetal membranes and placenta in human term pregnancies with oligohydramnios. Placenta. 2009; 30: 670–6.Google Scholar
Rabinowitz, R, Peters, MT, Vyas, S, Campbell, S, Nicolaides, KH. Measurement of fetal urine production in normal pregnancy by real-time ultrasonography. Am J Obstet Gynecol. 1989; 161: 1264–6.Google Scholar
Fagerquist, M, Fagerquist, U, Oden, A, Blomberg, SG. Fetal urine production and accuracy when estimating fetal urinary bladder volume. Ultrasound Obstet Gynecol. 2001; 17: 132–9.Google Scholar
Ross, MG, Ervin, MG, Rappaport, VJ, et al. Ovine fetal urine contribution to amniotic and allantoic compartments. Biol Neonate. 1988; 53: 98104.Google Scholar
Wlodek, ME, Challis, JR, Patrick, J. Urethral and urachal urine output to the amniotic and allantoic sacs in fetal sheep. J Dev Physiol. 1988; 10: 309–19.Google Scholar
Gresham, EL, Rankin, JH, Makowski, EL, Meschia, G, Battaglia, FC. An evaluation of fetal renal function in a chronic sheep preparation. J Clin Invest. 1972; 51: 149–56.Google Scholar
Hargrave, BY, Castle, MC. Effects of phenylephrine induced increase in arterial pressure and closure of the ductus arteriosus on the secretion of atrial natriuretic peptide (ANP) and renin in the ovine fetus. Life Sci. 1995; 57: 3143.Google Scholar
Silberbach, M, Woods, LL, Hohimer, AR, et al. Role of endogenous atrial natriuretic peptide in chronic anemia in the ovine fetus: effects of a non-peptide antagonist for atrial natriuretic peptide receptor. Pediatr Res. 1995; 38: 722–8.Google Scholar
Lee, SM, Jun, JK, Lee, EJ, et al. Measurement of fetal urine production to differentiate causes of increased amniotic fluid volume. Ultrasound Obstet Gynecol. 2010; 36: 191–5.Google Scholar
Xu, Z, Glenda, C, Day, L, Yao, J, Ross, MG. Osmotic threshold and sensitivity for vasopressin release and fos expression by hypertonic NaCl in ovine fetus. Am J Physiol Endocrinol Metab. 2000; 279: E1207–15.Google Scholar
Horne, RS, MacIsaac, RJ, Moritz, KM, Tangalakis, K, Wintour, EM. Effect of arginine vasopressin and parathyroid hormone-related protein on renal function in the ovine foetus. Clin Exp Pharmacol Physiol. 1993; 20: 569–77.Google Scholar
Cabrol, D, Landesman, R, Muller, J, Sureau, C, Saxena, BB. Treatment of polyhydramnios with prostaglandin synthetase inhibitor (indomethacin). Am J Obstet Gynecol. 1987; 157: 422–6.Google Scholar
Kullama, LK, Nijland, MJ, Ervin, MG, Ross, MG. Intraamniotic deamino(D-Arg8)-vasopressin: prolonged effects on ovine fetal urine flow and swallowing. Am J Obstet Gynecol. 1996; 174: 7884.Google Scholar
Brace, RA, Wlodek, ME, Cock, ML, Harding, R. Swallowing of lung liquid and amniotic fluid by the ovine fetus under normoxic and hypoxic conditions. Am J Obstet Gynecol. 1994; 171: 764–70.Google Scholar
Evrard, VA, Flageole, H, Deprest, JA, et al. Intrauterine tracheal obstruction, a new treatment for congenital diaphragmatic hernia, decreases amniotic fluid sodium and chloride concentrations in the fetal lamb. Ann Surg. 1997; 226: 753–8.Google Scholar
Ross, MG, Ervin, G, Leake, RD, Fu, P, Fisher, DA. Fetal lung liquid regulation by neuropeptides. Am J Obstet Gynecol. 1984; 150: 421–5.Google Scholar
Lawson, EE, Brown, ER, Torday, JS, Madansky, DL, Taeusch, HWJ. The effect of epinephrine on tracheal fluid flow and surfactant efflux in fetal sheep. Am Rev Respir Dis. 1978; 118: 1023–6.Google Scholar
Dodic, M, Wintour, EM. Effects of prolonged (48 h) infusion of cortisol on blood pressure, renal function and fetal fluids in the immature ovine foetus. Clin Exp Pharmacol Physiol. 1994; 21: 971–80.Google Scholar
Jain, L, Eaton, DC. Physiology of fetal lung fluid clearance and the effect of labor. Semin Perinatol. 2006; 30: 3443.Google Scholar
Norlin, A, Folkesson, HG. Ca(2+)-dependent stimulation of alveolar fluid clearance in near-term fetal guinea pigs. Am J Physiol Lung Cell Mol Physiol. 2002; 282: L642–9.Google Scholar
Pritchard, JA. Fetal swallowing and amniotic fluid volume. Obstet Gynecol. 1966; 28: 606–10.Google Scholar
Bradley, RM, Mistretta, CM. Swallowing in fetal sheep. Science. 1973; 179: 1016–17.Google Scholar
Nijland, MJ, Day, L, Ross, MG. Ovine fetal swallowing: expression of preterm neurobehavioral rhythms. J Matern Fetal Med. 2001; 10: 251–7.Google Scholar
Matsumoto, LC, Cheung, CY, Brace, RA. Effect of esophageal ligation on amniotic fluid volume and urinary flow rate in fetal sheep. Am J Obstet Gynecol. 2000; 182: 699705.Google Scholar
Xu, Z, Nijland, MJ, Ross, MG. Plasma osmolality dipsogenic thresholds and c-fos expression in the near-term ovine fetus. Pediatr Res. 2001; 49: 678–85.Google Scholar
El-Haddad, MA, Ismail, Y, Gayle, D, Ross, MG. Central angiotensin II AT1 receptors mediate fetal swallowing and pressor responses in the near term ovine fetus. Am J Physiol Regul Integr Comp Physiol. 2005; 288: R1014–20.Google Scholar
El-Haddad, MA, Ismail, Y, Guerra, C, Day, L, Ross, MG. Neuropeptide Y administered into cerebral ventricles stimulates sucrose ingestion in the near-term ovine fetus. Am J Obstet Gynecol. 2003; 189: 949–52.Google Scholar
El-Haddad, MA, Ismail, Y, Guerra, C, Day, L, Ross, MG. Effect of oral sucrose on ingestive behavior in the near-term ovine fetus. Am J Obstet Gynecol. 2002; 187: 898901.Google Scholar
Sherman, DJ, Ross, MG, Day, L, Humme, J, Ervin, MG. Fetal swallowing: response to graded maternal hypoxemia. J Appl Physiol. 1991; 71: 1856–61.Google Scholar
Queenan, JT, Allen, FH Jr., Fuchs, F, et al. Studies on the method of intrauterine transfusion. I. Question of erythrocyte absorption from amniotic fluid. Am J Obstet Gynecol. 1965; 92: 1009–13.Google Scholar
Gilbert, WM, Brace, RA. The missing link in amniotic fluid volume regulation: intramembranous absorption. Obstet Gynecol. 1989; 74: 748–54.Google Scholar
Gilbert, WM, Cheung, CY, Brace, RA. Rapid intramembranous absorption into the fetal circulation of arginine vasopressin injected intraamniotically. Am J Obstet Gynecol. 1991; 164: 1013–18.Google Scholar
Jang, PR, Brace, RA. Amniotic fluid composition changes during urine drainage and tracheoesophageal occlusion in fetal sheep. Am J Obstet Gynecol. 1992; 167: 1732–41.Google Scholar
Brace, RA. Physiology of amniotic fluid volume regulation. Clin Obstet Gynecol. 1997; 40: 280–9.Google Scholar
Hedriana, HL, Gilbert, WM, Brace, RA. Arginine vasopressin-induced changes in blood flow to the ovine chorion, amnion, and placenta across gestation. J Soc Gynecol Invest. 1997; 4: 203–8.Google Scholar
Verkman, AS, Dix, JA. Effect of unstirred layers on binding and reaction kinetics at a membrane surface. Anal Biochem. 1984; 142: 109–16.CrossRefGoogle Scholar
Daneshmand, SS, Cheung, CY, Brace, RA. Regulation of amniotic fluid volume by intramembranous absorption in sheep: role of passive permeability and vascular endothelial growth factor. Am J Obstet Gynecol. 2003; 188: 786–93.Google Scholar
Wynn, RM, French, GL. Comparative ultrastructure of the mammalian amnion. Obstet Gynecol. 1968; 31: 759–74.Google Scholar
Curran, MA, Nijland, MJ, Mann, SE, Ross, MG. Human amniotic fluid mathematical model: determination and effect of intramembranous sodium flux. Am J Obstet Gynecol. 1998; 178: 484–90.Google Scholar
Faber, JJ, Anderson, DF. Absorption of amniotic fluid by amniochorion in sheep. Am J Physiol Heart Circ Physiol. 2002; 282: H850–4.Google Scholar
Matsumoto, LC, Cheung, CY, Brace, RA. Increased urinary flow without development of polyhydramnios in response to prolonged hypoxia in the ovine fetus. Am J Obstet Gynecol. 2001; 184: 1008–14.Google Scholar
Faber, JJ, Anderson, DF. Regulatory response of intramembranous absorption of amniotic fluid to infusion of exogenous fluid in sheep. Am J Physiol. 1999; 277: R236–42.Google Scholar
Matsumoto, LC, Bogic, L, Brace, RA, Cheung, CY. Prolonged hypoxia upregulates vascular endothelial growth factor messenger RNA expression in ovine fetal membranes and placenta. Am J Obstet Gynecol. 2002; 186: 303–10.Google Scholar
Zhu, X, Jiang, S, Hu, Y, et al. The expression of aquaporin 8 and aquaporin 9 in fetal membranes and placenta in term pregnancies complicated by idiopathic polyhydramnios. Early Hum Dev. 2010; 86: 657–63.Google Scholar
Huang, J, Qi, HB. Expression of aquaporin 8 in human fetal membrane and placenta of idiopathic polyhydramnios. Zhonghua Fu Chan Ke Za Zhi. 2009; 44: 1922.Google Scholar
Qi, H, Li, L, Zong, W, Hyer, BJ, Huang, J. Expression of aquaporin 8 is diversely regulated by osmotic stress in amnion epithelial cells. J Obstet Gynaecol Res. 2009; 35: 1019–25.Google Scholar
Wang, S, Amidi, F, Yin, S, Beall, M, Ross, MG. Cyclic adenosine monophosphate regulation of aquaporin gene expression in human amnion epithelia. Reprod Sci. 2007; 14: 234–40.Google Scholar
Ross, MG, Ervin, MG, Leake, RD, et al. Bulk flow of amniotic fluid water in response to maternal osmotic challenge. Am J Obstet Gynecol. 1983; 147: 697701.Google Scholar
Leontic, EA, Tyson, JE. Prolactin and fetal osmoregulation: water transport across isolated human amnion. Am J Physiol. 1977; 232: R124–7.Google Scholar
Holt, WF, Perks, AM. The effect of prolactin on water movement through the isolated amniotic membrane of the guinea pig. Gen Comp Endocrinol. 1975; 26: 153–64.Google Scholar

References

Magann, EF, Sanderson, M, Martin, JN, et al. The amniotic fluid index, single deepest pocket, and two-diameter pocket in normal human pregnancy. Am J Obstet Gynecol. 2001; 182: 1581–8.Google Scholar
Sandlin, AT, Chauhan, SP, Magann, EF. Clinical relevance of sonographically estimated amniotic fluid volume. J Ultrasound Med. 2013; 32: 851–63.Google Scholar
Karkhanis, P, Patni, S. Polyhydramnios in singleton pregnancies: perinatal outcomes and management. Obstet Gynaecol. 2014; 16: 207–13.Google Scholar
Dashe, JS, McIntire, DD, Ramus, RM, et al. Hydramnios: anomaly prevalence and sonographic detection. Obstet Gynecol. 2002; 100: 134–9.Google Scholar
Martinez-Frias, MJ, Bermejo, E, Rodriguez-Pinilla, E, et al. Maternal and fetal factors related to abnormal amniotic fluid. J Perinatol. 1999; 19: 514–20.Google Scholar
Golan, A, Wolman, I, Langer, R, et al. Fetal malformations associated with chronic polyhydramnios in singleton pregnancies. Eur J Obstet Gynecol. 1992; 47: 185–8.Google Scholar
Brady, K, Polzin, WJ, Kopelman, JN, et al. Risk of chromosomal abnormalities in patients with idiopathic polyhydramnios. Obstet Gynecol. 1992; 79: 234–8.Google Scholar
Lee, JF, Wang, KK, Lan, CC. Risk of fetal chromosomal abnormalities in idiopathic polyhydramnios. Zhonghua Yi Xue Za Zhi (Taipei). 1996; 57: 42–6.Google Scholar
Lazebnik, N, Many, A. The severity of polyhydramnios, estimated fetal weight and preterm delivery are independent risk factors for the presence of congenital malformations. Gynecol Obstet Invest. 1999; 48: 2832.Google Scholar
Sickler, GK, Nyberg, DA, Sohaey, R, et al. Polyhydramnios and fetal intrauterine growth restriction: ominous combination. J Ultrasound Med. 1997; 16: 609–14.Google Scholar
Sagi-Dain, L, Sagi, S. Chromosomal aberrations in idiopathic polyhydramnios: a systematic review and meta-analysis. Eur J Med Genet. 2015; 58: 409–15.Google Scholar
Magann, EF, Chauhan, SP, Boherty, DA, et al. A review of idiopathic hydramnios and pregnancy outcomes. Obstet Gynecol Survey. 2007; 62: 795801.Google Scholar
Morris, RK, Meller, CH, Tamblyn, J, et al. Association and prediction of amniotic fluid measurements for adverse pregnancy outcome: systematic review and meta-analysis. BJOG. 2014; 121: 686–99.Google Scholar
Many, A, Hill, LM, Lazebnik, N, Martin, JG. The association between polyhydramnios and preterm delivery. Obstet Gynecol. 1995; 86: 389–91.Google Scholar
Odibo, IN, Newville, TM, Ounpraseuth, ST, et al. Idiopathic polyhydramnios: persistence across gestation and impact on pregnancy outcomes. Eur J Obstet Gynecol Reprod Biol. 2016; 199: 175–8.Google Scholar
Fisk, NM, Vaughn, J, Talbert, D. Impaired fetal blood gas status in polyhydramnios and its relation to raised amniotic fluid pressure. Fetal Diag Ther. 1994; 9: 713.Google Scholar
Hershkovitz, R, Furman, B, Bashiri, A, et al. Evidence for abnormal middle cerebral artery values in patients with idiopathic hydramnios. J Matern Fetal Med. 2001; 10: 404–8.Google Scholar
Dickinson, JE, Tjioe, YY, Jude, E, et al. Amnioreduction in the management of polyhydramnios complicating singleton pregnancies. Am J Obstet Gynecol. 2014; 211: e1–7.Google Scholar
Dickinson, JE, Evans, SF. Obstetric and perinatal outcomes from the Australian and New Zealand Twin-Twin Transfusion Syndrome Registry. Am J Obstet Gynecol. 2000; 182: 706–12.Google Scholar
Taylor, MJO, Fisk, NM. Hydramnios and oligohydramnios. In James, DK, Steer, PJ, Weiner, CP, Gonik, B, eds., High Risk Pregnancy: Management Options, 3rd edn. Philadelphia: Elsevier Publishers, 2006, pp. 272–90.Google Scholar
Denbow, ML, Fisk, NM. The consequences of monochorionic placentation. Baillieres Clin Obstet Gynecol. 1998; 12: 3751.Google Scholar
Moise, KJ Jr. Polyhydramnios. Clin Obstet Gynecol. 1997; 40: 266–79.Google Scholar
Van den Veyver, I, Moise, KJ, Ou, C-N, et al. The effect of gestational age and fatal indomethacin levels on the incidence of constriction of the ductus arteriosus. Obstet Gynecol. 1993; 182: 500–3.Google Scholar
Kramer, WB, Saade, GS, Ou, C-N, et al. Placental transfer of sulindac and its active sulfide metabolite in humans. Am J Obstet Gynecol. 1995; 172: 886–90.Google Scholar
Rasanen, J, Jouppila, P. Fetal cardiac function and ductus arteriosus during indomethacin and sulindac therapy for threatened preterm labor: a randomized study. Am J Obstet Gynecol. 1995; 173: 20–5.Google Scholar
Sawdy, RJ, Lye, S, Fisk, NM, et al. A double-blind randomized study of fetal side effects during and after the short-term maternal administration of indomethacin, sulindac, and nimesulide for the treatment of preterm labor. Am J Obstet Gynecol. 2003; 188: 1046–51.Google Scholar
Society for Maternal-Fetal Medicine, Dashe, JS, Pressman, EK, Hibbard, JU. SMFM Consult Series #46: Evaluation and Management of PolyhydramniosAm J Obstet Gynecol. 2018; 219: B2–8.Google Scholar
Moore, TR, Longo, J, Leopold, GR, Casola, G, Gosink, BB. The reliability and predictive value of an amniotic fluid scoring system in severe second trimester oligohydramnios. Obstet Gynecol. 1989; 73: 739–42.Google Scholar
Fisk, NF. Oligohydramnios-related pulmonary hypoplasia. Contemp Rev Obstet and Gynecol. 1992; 4: 191210.Google Scholar
Yoshimura, S, Masuzaki, H, Miura, K, Hayashi, H, Gotoh, H, Ishimaru, T. The effects of oligohydramnios and cervical cord transection on lung growth in experimental pulmonary hypoplasia in rabbits. Am J Obstet Gynecol. 1997; 177: 72–7.Google Scholar
Nelson, SM, Hajivassiliou, CA, Haddock, G, Cameron, AD, Robertson, L, Olver, RE, Hume, R. Rescue of the hypoplastic lung by prenatal cyclical strain. Am J Resp Critical Care Med. 2005; 171: 1395–402.Google Scholar
van Teeffelen, ASP, van der Heijden, J, Oei, SG, Porath, MM, Willekes, C, Opmeer, B, Mol, BWJ. Accuracy of imagining parameters in the prediction of lethal pulmonary hypoplasia secondary to mid-trimester prelabour rupture of membranes: a systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2012; 39: 495–9.Google Scholar
Golbus, MS, Harrison, MR, Filly, RA, et al. In utero treatment of urinary tract obstruction. Am J Obstet Gynecol. 1982; 142: 383–8.Google Scholar
Morris, RK, Malin, GL, Quinlan‐Jones, E, Middleton, LJ, Hemming, K, Burke, D, et al. Percutaneous vesicoamniotic shunting versus conservative management for fetal lower urinary tract obstruction (PLUTO): a randomised trial. Lancet. 2013; 382: 1496–506.Google Scholar
Caughey, AB, Robinson, JN, Norwitz, ER. Contemporary diagnosis and management of preterm premature rupture of membranes. Rev Obstet Gynecol. 2008; 1: 1122.Google Scholar
Garite, TJ, Freeman, RK, Linzey, EM, Braly, P. The use of amniocentesis in patients with premature rupture of membranes. Obstet Gynecol. 1979; 54: 226–30.Google Scholar
Lee, J, Romero, R, Kim, SM, Chaemsaithong, P, Yoon, BH. A new antibiotic regimen treats and prevents intra-amniotic inflammation/infection in patients with preterm PROM. J Matern Fetal Neonatal Med. 2016; 29: 2727–37.Google Scholar
Devlieger, R, Millar, LK, Bryant-Greenwood, G, Lewi, L, Deprest, JA. Fetal membrane healing after spontaneous and iatrogenic membrane rupture: a review of current evidence. Am J Obstet Gynecol. 2006; 195: 1512–20.Google Scholar
Quintero, RA, Mendoza, GA, Allen, M, Arroyo, J, Bornick, PW, Morales, WJ, et al. In vivo laser welding of collagen-based graft material to the amnion in a rabbit model of ruptured membranes. Prenat Neonat Med. 1999; 4: 453–6.Google Scholar
Borgida, A, Mills, A, Feldman, D, et al. Outcome of pregnancies complicated by ruptured membranes after genetic amniocentesis. Am J Obstet Gynecol. 2000; 183: 937–9.Google Scholar
Harrison, MR. Surgically correctable fetal disease. Am J Surg. 2000; 180: 335–42.Google Scholar
Beck, V, Lewi, P, Gucciardo, L, Devlieger, R. Preterm prelabour rupture of membranes and fetal survival after minimally invasive fetal surgery: a systematic review of the literature. Fetal Diagn Ther. 2012; 31: 19.Google Scholar
Akkermans, J, Peeters, SHP, Klumper, FJ, Lopriore, E, Middeldorp, JM, Oepkes, D. Twenty-five years of fetoscopic laser coagulation of twin-twin transfusion syndrome: a systematic review. Fetal Diagn Ther. 2015; 38: 241–53.Google Scholar
Senat, MV, Deprest, J, Boulvain, M, Paupe, A, Winer, N, Ville, Y. Endoscopic laser surgery versus serial amnioreduction for severe twin-to-twin transfusion syndrome. N Engl J Med. 2004; 351: 136–44.Google Scholar
Stirnemann, J, Djaafri, F, Kim, A, Mediouni, I, Bussieres, L, Spaggiari, E, et al. Preterm premature rupture of membranes is a collateral effect of improvement in perinatal outcomes following fetoscopic coagulation of chorionic vessels for twin-twin transfusion syndrome: a retrospective observational study of 1092 casesBJOG. 2018; 125: 1154–62.Google Scholar
Gratacὀs, E, Sanin-Blair, J, Lewi, L, Toran, N, Verbist, L, Cabero, L, et al. A histological study of fetoscopic membrane defects to document membrane healing. Placenta. 2006; 27: 452–6.Google Scholar
Dewan, H, Morris, J. A systematic review of pregnancy outcome following preterm premature rupture of membranes at a previable gestational age. Aust N Z J Obstet Gynecol. 2001; 41: 389–94.Google Scholar
Chauleur, C, Rochigneux, S, Seffert, P, et al. Neonatal outcomes and four-year follow-up after spontaneous or iatrogenic preterm prelabour rupture of membranes before 24 weeks. Acta Obstet Gynecol Scand. 2009; 88: 801–6.Google Scholar
Linehan, LA, Walsh, J, Morris, A, Kenny, L, O’Donoghue, K, Dempsey, E, Russell, N. Neonatal and maternal outcomes following midtrimester preterm premature rupture of the membranes: a retrospective cohort study. BMC Pregnancy Childbirth. 2016; 16: 25.Google Scholar
van der Heyden, JL, van der Ham, DP, van Kuijk, S, Notten, KJ, Janssen, T, Nijhuis, JG, et al. Outcome of pregnancies with preterm prelabor rupture of membranes before 27 weeks’ gestation: a retrospective cohort study. Eur J Obstet Gynecol Reprod Biol. 2013; 170: 125130.Google Scholar
Vergani, P, Ghidini, A, Locatelli, A, et al. Risk factors for pulmonary hypoplasia in second-trimester premature rupture of membranes. Am J Obstet Gynecol. 1994; 170: 1359–64.Google Scholar
Kilbride, HW, Yeast, J, Thibeault, DW. Defining the limits of survival: lethal pulmonary hypoplasia after midtrimester premature rupture of membranes. Am J Obstet Gynecol. 1996; 175: 675–81.Google Scholar
Snowise, S, Mann, LK, Moise, JR, Johnson, M, Bebbington, MW, Papanna, R. Preterm prelabour rupture of membranes after fetoscopic laser surgery for twin-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2017; 49: 607–11.Google Scholar
Genz, HJ, Gerlach, H, Metzger, H. Behandlung des vorzeitigen Blasensprungs durch fibrinklebung. Med Welt. 1979; 42: 1557.Google Scholar
Quintero, RA, Morales, WJ, Allen, M, Bornick, PW, Arroyo, J, LeParc, G. Treatment of iatrogenic previable premature rupture of membranes with intra-amniotic injection of platelets and cryo-precipitate (amniopatch): preliminary experience. Am J Obstet Gynecol. 1999; 181: 744–9.Google Scholar
Richter, J, Henry, A, Ryan, G, Lewi, L, Deprest, J. Amniopatch procedure after previable iatrogenic rupture of the membranes: a two-centre review. Prenat Diagn. 2013; 33: 391–6.Google Scholar
Chmait, RH, Kontopoulos, EV, Chon, AH, Korst, LM, Llanes, A, Quintero, RA. Amniopatch treatment of iatrogenic preterm premature rupture of membranes (iPPROM) after fetoscopic laser surgery for twin–twin transfusion syndrome. J Matern Fetal Neonatal Med. 2017; 30: 1349–54.Google Scholar
Locatelli, A, Vergani, P, Di Pirro, G, Doria, V, Biffi, A, Ghidini, A. Role of amnioinfusion in the management of premature rupture of the membranes at <26 weeks’ gestation. Am J Obstet Gynecol. 2000; 183: 878–82.Google Scholar
Vergani, P, Locatelli, A, Verderio, M, Assi, F. Premature rupture of the membranes at <26 weeks’ gestation: role of amnioinfusion in the management of oligohydramnios. Acta Biomed. 2004; 75 (Suppl.): 62–6.Google Scholar
Hofmeyr, GJ, Essilfie-Appiah, G, Lawrie, TA. Amnioinfusion for preterm premature rupture of membranes. Cochrane Database Syst Rev. 2011; 12: CD000942.Google Scholar
Porat, S, Amsalem, H, Shah, PS, Murphy, KE. Transabdominal amnioinfusion for preterm premature rupture of membranes: a systematic review and meta analysis of randomized and observational studies. Am J Obstet Gynecol. 2012; 207: 393. e1–11.Google Scholar
Vergani, P, Locatelli, A, Strobelt, N, et al. Amnioinfusion for prevention of pulmonary hypoplasia in second-trimester rupture of membranes. Am J Perinatol. 1997; 14: 325–9.Google Scholar
Ogunyemi, D, Thompson, W. A case controlled study of serial transabdominal amnioinfusions in the management of second trimester oligohydramnios due to premature rupture of membranes. Eur J Obstet Gynecol Reprod Biol. 2002; 102: 167–72.Google Scholar
De Santis, M, Scavo, M, Noia, G, et al. Transabdominal amnioinfusion treatment of severe oligohydramnios in preterm premature rupture of membranes at less than 26 gestational weeks. Fetal Diagn Ther. 2003; 18: 412–17.Google Scholar
Roberts, D, Vause, S, Martin, W, Green, P, Walkinshaw, S, Bricker, L, et al. Amnioinfusion in very early preterm premature rupture of membranes – pregnancy, neonatal and maternal outcomes in the AMIPROM randomised controlled pilot study. Ultrasound Obstet Gynecol. 2013; 43: 490–9.Google Scholar
van Kempen, LEM, van Teeffelen, AS, de Ruigh, AA, Oepkes, D, Haak, MC, van Leeuwen, E, et al. Amnioinfusion compared with no intervention in women with second-trimester rupture of membranes: a randomized controlled trial. Obstet Gynecol. 2019; 133: 129136.Google Scholar
Hofmeyr, GJ, Eke, AC, Lawrie, TA. Amnioinfusion for third trimester preterm premature rupture of membranes. Cochrane Database Syst Rev. 2014; 3: CD000942.Google Scholar
Nageotte, MP, Freeman, RK, Garite, TJ, Dorchester, W. Prophylactic intrapartum amnioinfusion in patients with preterm premature rupture of membranes. Am J Obstet Gynecol. 1985; 153: 557–62.Google Scholar
Puertas, A, Tirado, P, Perez, I, Lopez, MS, Montoya, F, Canizares, JM, et al. Transcervical intrapartum amnioinfusion for preterm premature rupture of the membranes. Eur J Obstet Gynecol Reprod Biol. 2007; 131: 40–4.Google Scholar
Singla, A, Yadav, P, Vaid, NB, Suneja, A, Faridi, MMA. Transabdominal amnioinfusion in preterm premature rupture of membranes. Int J Gynecol Obstet. 2010; 108: 199202.Google Scholar
Tranquilli, AL, Giannubilo, SR, Bezzeccheri, V, Scagnoli, C. Transabdominal amnioinfusion in preterm premature rupture of membranes: a randomised controlled trial. BJOG. 2005; 112: 759–63.Google Scholar
Hofmeyr, GJ, Xu, H, Eke, AC. Amnioinfusion for meconium-stained liquor in labour. Cochrane Database Syst Rev. 2014; 1: CD000014.Google Scholar
Fraser, WD, Hofmeyr, J, Lede, R, Faron, G, Alexander, S, Goffinet, F, et al. Amnioinfusion for the prevention of the meconium aspiration syndrome. N Engl J Med. 2005; 353: 909–17.Google Scholar
Hofmeyr, GJ. Amnioinfusion for meconium-stained liquor in labour. Cochrane Database Syst Rev. 2002; 1: CD000014.Google Scholar
Hemming, K, Hutton, JL. Intrapartum amnioinfusion for meconium-stained amniotic fluid: evidence for small study effect bias? BJOG. 2009; 116: 128–9.Google Scholar

References

O’Shea, JJ, Paul, WE. Mechanisms Underlying lineage commitment and plasticity of helper CD4+ T cells. Science. 2010; 327: 1098–102.Google Scholar
Billingham, RE, Brent, L, Medawar, PB. Actively acquired tolerance of foreign cells. Nature. 1953; 172: 603–6.Google Scholar
Adkins, B, Leclerc, C, Marshall-Clarke, S. Neonatal adaptive immunity comes of age. Nat Rev Immunol. 2004; 4: 553–64.Google Scholar
Sarzotti, M, Robbins, DS, Hoffman, PM. Induction of protective CTL responses in newborn mice by a murine retrovirus. Science. 1996; 271: 1726–8.Google Scholar
Ridge, JP, Fuchs, EJ, Matzinger, P. Neonatal tolerance revisited: turning on newborn T cells with dendritic cells. Science. 1996; 271: 1723–6.Google Scholar
Forsthuber, T, Yip, HC, Lehmann, PV. Induction of TH1 and TH2 immunity in neonatal mice. Science. 1996; 271: 1728–30.Google Scholar
Marchant, A, Goldman, M. T cell-mediated immune responses in human newborns: ready to learn? Clin Exp Immunol. 2005; 141: 1018.Google Scholar
Tavian, M, Péault, B. Embryonic development of the human hematopoietic system. Int J Dev Biol. 2005; 49: 243–50.Google Scholar
Hong, DK, Lewis, DB. Developmental Immunology and Role of Host Defenses in Fetal and Neonatal Susceptibility to Infection. In Remington and Klein’s Infectious Diseases of the Fetus and Newborn Infant, 8th edn. Philadelphia: Elsevier Saunders, 2016, pp. 90197.Google Scholar
Willems, F, Vollstedt, S, Suter, M. Phenotype and function of neonatal DC. Eur J Immunol. 2009; 39: 2635.Google Scholar
Lemoine, S, Jaron, B, Tabka, S, Ettreiki, C, Deriaud, E, Zhivaki, D, et al. Dectin-1 activation unlocks IL12A expression and reveals the TH1 potency of neonatal dendritic cells. J Allergy Clin Immunol. 2015; 136: 13551368. e15.Google Scholar
Salio, M, Dulphy, N, Renneson, J, Herbert, M, McMichael, A, Marchant, A, et al. Efficient priming of antigen-specific cytotoxic T lymphocytes by human cord blood dendritic cells. Int Immunol. 2003; 15: 1265–73.Google Scholar
Rechavi, E, Somech, R. Survival of the fetus: fetal B and T cell receptor repertoire development. Semin Immunopathol. 2017; 39: 577–83.Google Scholar
White, GP, Watt, PM, Holt, BJ, Holt, PG. Differential patterns of methylation of the IFN-promoter at CpG and non-CpG sites underlie differences in IFN-gene expression between human neonatal and adult CD45RO-T cells. J Immunol. 2002; 168: 2820–7.Google Scholar
Zhang, X, Mozeleski, B, Lemoine, S, Deriaud, E, Lim, A, Zhivaki, D, et al. CD4 T Cells with effector memory phenotype and function develop in the sterile environment of the fetus. Sci Transl Med. 2014; 6: 238ra72.Google Scholar
Rechavi, E, Lev, A, Lee, YN, Simon, AJ, Yinon, Y, Lipitz, S, et al. Timely and spatially regulated maturation of B and T cell repertoire during human fetal development. Sci Transl Med. 2015; 7: 276ra25.Google Scholar
Suryani, S, Fulcher, DA, Santner-Nanan, B, Nanan, R, Wong, M, Shaw, PJ, et al. Differential expression of CD21 identifies developmentally and functionally distinct subsets of human transitional B cells. Blood. 2010; 115: 519–29.Google Scholar
Griffin, DO, Holodick, NE, Rothstein, TL. Human B1 cells in umbilical cord and adult peripheral blood express the novel phenotype CD20+ CD27+ CD43+ CD70-. J Exp Med. 2011; 208: 6780.Google Scholar
Vermijlen, D, Prinz, I. Ontogeny of Innate T lymphocytes – some innate lymphocytes are more innate than others. Front Immunol. 2014; 5: 486.Google Scholar
Dimova, T, Brouwer, M, Gosselin, F, Tassignon, J, Leo, O, Donner, C, et al. Effector Vγ9Vδ2 T cells dominate the human fetal γδ T-cell repertoire. Proc Natl Acad Sci. 2015; 112: E556–65.Google Scholar
Michaëlsson, J, Mold, JE, McCune, JM, Nixon, DF. Regulation of T cell responses in the developing human fetus. J Immunol. 2006; 176: 5741–8.Google Scholar
Mold, JE, Michaëlsson, J, Burt, TD, Muench, MO, Beckerman, KP, Busch, MP, et al. Maternal alloantigens promote the development of tolerogenic fetal regulatory T cells in utero. Science. 2008; 322: 1562–5.Google Scholar
Morand, A, Zandotti, C, Charrel, R, Minodier, P, Fabre, A, Chabrol, B, et al. De TORCH à TORCHZ: fœtopathies infectieuses à virus Zika et autres. Arch Pédiatrie. 2017; 24: 911–13.Google Scholar
Maldonado, YA, Nizet, V, Klein, JO, Remington, JS, Wilson, CB. Current concepts of infections of the fetus and newborn infant. In Remington and Klein’s Infectious Diseases of the Fetus and Newborn Infant, 8th edn. Philadelphia: Elsevier Saunders, 2016, pp. 323.Google Scholar
Renneson, J, Dutta, B, Goriely, S, Danis, B, Lecomte, S, Laes, J-F, et al. IL-12 and type I IFN response of neonatal myeloid DC to human CMV infection. Eur J Immunol. 2009; 39: 2789–99.Google Scholar
Marchant, A, Appay, V, van der Sande, M, Dulphy, N, Liesnard, C, Kidd, M, et al. Mature CD8+ T lymphocyte response to viral infection during fetal life. J Clin Invest. 2003; 111: 1747–55.Google Scholar
Pédron, B, Guérin, V, Jacquemard, F, Munier, A, Daffos, F, Thulliez, P, et al. Comparison of CD8+ T cell responses to cytomegalovirus between human fetuses and their transmitter mothers. J Infect Dis. 2007; 196: 1033–43.Google Scholar
Gibson, L, Piccinini, G, Lilleri, D, Revello, MG, Wang, Z, Markel, S, et al. Human cytomegalovirus proteins pp65 and immediate early protein 1 are common targets for CD8+ T cell responses in children with congenital or postnatal human cytomegalovirus infection. J Immunol. 2004; 172: 2256–64.Google Scholar
Miles, DJC, van der Sande, M, Jeffries, D, Kaye, S, Ismaili, J, Ojuola, O, et al. Cytomegalovirus infection in Gambian infants leads to profound CD8 T-cell differentiation. J Virol. 2007; 81: 5766–76.Google Scholar
Gibson, L, Dooley, S, Trzmielina, S, Somasundaran, M, Fisher, D, Revello, MG, et al. Cytomegalovirus (CMV) IE1‐ and pp65‐specific CD8+ T cell responses broaden over time after primary CMV infection in infants. J Infect Dis. 2007; 195: 1789–98.Google Scholar
Pass, RF, Stagno, S, Britt, WJ, Alford, CA. Specific cell-mediated immunity and the natural history of congenital infection with cytomegalovirus. J Infect Dis. 1983; 148: 953–61.Google Scholar
Starr, SE, Tolpin, MD, Friedman, HM, Paucker, K, Plotkin, SA. Impaired Cellular immunity to cytomegalovirus in congenitally infected children and their mothers. J Infect Dis. 1979; 140: 500–5.Google Scholar
Huygens, A, Lecomte, S, Tackoen, M, Olislagers, V, Delmarcelle, Y, Burny, W, et al. Functional exhaustion limits CD4+ and CD8+ T-cell responses to congenital cytomegalovirus infection. J Infect Dis. 2015; 212: 484–94.Google Scholar
Tu, W, Chen, S, Sharp, M, Dekker, C, Manganello, AM, Tongson, EC, et al. Persistent and selective deficiency of CD4+ T cell immunity to cytomegalovirus in immunocompetent young children. J Immunol. 2004; 172: 3260–7.Google Scholar
Vermijlen, D, Brouwer, M, Donner, C, Liesnard, C, Tackoen, M, Van Rysselberge, M, et al. Human cytomegalovirus elicits fetal γδ T cell responses in utero. J Exp Med. 2010; 207: 807–21.Google Scholar
Brizić, I, Hiršl, L, Britt, WJ, Krmpotić, A, Jonjić, S. Immune responses to congenital cytomegalovirus infection. Microbes Infect. 2017; 20: 543–51.Google Scholar
Rovito, R, Korndewal, MJ, van Zelm, MC, Ziagkos, D, Wessels, E, van der Burg, M, et al. T and B cell markers in dried blood spots of neonates with congenital cytomegalovirus infection: B cell numbers at birth are associated with long-term outcomes. J Immunol. 2017; 198: 102–9.Google Scholar
Huygens, A, Dauby, N, Vermijlen, D, Marchant, A. Immunity to cytomegalovirus in early life. Front Immunol. 2014; 5: 552.Google Scholar
Noyola, DE, Fortuny, C, Muntasell, A, Noguera-Julian, A, Muñoz-Almagro, C, Alarcón, A, et al. Influence of congenital human cytomegalovirus infection and the NKG2C genotype on NK-cell subset distribution in children: immunity to infection. Eur J Immunol. 2012; 42: 3256–66.Google Scholar
Shetty, A, Maldonado, YA. Human Immunodeficiency Virus/Acquired Immunodeficiency Syndrome in the Infant. In Remington and Klein’s Infectious Diseases of the Fetus and Newborn Infant, 8th edn. Philadelphia: Elsevier Saunders, 2016, pp. 623–78.Google Scholar
Luzuriaga, K, Holmes, D, Hereema, A, Wong, J, Panicali, DL, Sullivan, JL. HIV-1-specific cytotoxic T lymphocyte responses in the first year of life. J Immunol. 1995; 154: 433–43.CrossRefGoogle ScholarPubMed
Thobakgale, CF, Ramduth, D, Reddy, S, Mkhwanazi, N, de Pierres, C, Moodley, E, et al. Human immunodeficiency virus-specific CD8+ T-cell activity is detectable from birth in the majority of in utero-infected infants. J Virol. 2007; 81: 12775–84.Google Scholar
Lohman, BL, Slyker, JA, Richardson, BA, Farquhar, C, Mabuka, JM, Crudder, C, et al. Longitudinal assessment of human immunodeficiency virus type 1 (HIV-1)-specific gamma interferon responses during the first year of life in HIV-1-infected infants. J Virol. 2005; 79: 8121–30.CrossRefGoogle ScholarPubMed
Streeck, H, Nixon, DF. T cell immunity in acute HIV‐1 infection. J Infect Dis. 2010; 202: S302–8.Google Scholar
Voelkerding, KV, Sandhaus, LM, Belov, L, Frenkel, L, Ettinger, LJ, Raska, K. Clonal B-cell proliferation in an infant with congenital HIV infection and immune thrombocytopenia. Am J Clin Pathol. 1988; 90: 470–4.Google Scholar
Pugatch, D, Sullivan, JL, Pikora, CA, Luzuriaga, K. Delayed generation of antibodies mediating human immunodeficiency virus type 1-specific antibody-dependent cellular cytotoxicity in vertically infected infants. WITS Study Group. Women and Infants Transmission Study. J Infect Dis. 1997; 176: 643–8.Google Scholar
Munoz, M, Liesenfeld, O, Heimesaat, MM. Immunology of Toxoplasma gondii. Immunol Rev. 2011; 240: 269–85.CrossRefGoogle ScholarPubMed
Fatoohi, AF, Cozon, GJN, Wallon, M, Kahi, S, Gay-Andrieu, F, Greenland, T, et al. Cellular immunity to Toxoplasma gondii in congenitally infected newborns and immunocompetent infected hosts. Eur J Clin Microbiol Infect Dis. 2003; 22: 181–4.Google Scholar
Ciardelli, L, Meroni, V, Avanzini, MA, Bollani, L, Tinelli, C, Garofoli, F, et al. Early and accurate diagnosis of congenital toxoplasmosis. Pediatr Infect Dis J. 2008; 27: 125–9.Google Scholar
Chapey, E, Wallon, M, Debize, G, Rabilloud, M, Peyron, F. Diagnosis of congenital toxoplasmosis by using a whole-blood gamma interferon release assay. J Clin Microbiol. 2010; 48: 41–5.Google Scholar
Guglietta, S, Beghetto, E, Spadoni, A, Buffolano, W, Del Porto, P, Gargano, N. Age-dependent impairment of functional helper T cell responses to immunodominant epitopes of Toxoplasma gondii antigens in congenitally infected individuals. Microbes Infect. 2007; 9: 127–33.Google Scholar
McLeod, R, Mack, DG, Boyer, K, Mets, M, Roizen, N, Swisher, C, et al. Phenotypes and functions of lymphocytes in congenital toxoplasmosis. J Lab Clin Med. 1990; 116: 623–35.Google Scholar
Hara, T, Ohashi, S, Yamashita, Y, Abe, T, Hisaeda, H, Himeno, K, et al. Human V delta 2+ gamma delta T-cell tolerance to foreign antigens of Toxoplasma gondii. Proc Natl Acad Sci U S A. 1996; 93: 5136–40.Google Scholar
Carlier, Y, Truyens, C. Maternal–fetal transmission of Trypanosoma cruzi. In American Trypanosomiasis: Chagas Disease, 2nd edn. Amsterdam: Elsevier, 2017, pp. 517–59.Google Scholar
Hermann, E, Truyens, C, Alonso-Vega, C, Even, J, Rodriguez, P, Berthe, A, et al. Human fetuses are able to mount an adultlike CD8 T-cell response. Blood. 2002; 100: 2153–8.Google Scholar
Hermann, E, Alonso-Vega, C, Berthe, A, Truyens, C, Flores, A, Cordova, M, et al. Human congenital infection with Trypanosoma cruzi induces phenotypic and functional modifications of cord blood NK cells. Pediatr Res. 2006; 60: 3843.Google Scholar
Rodriguez, P, Truyens, C, Alonso-Vega, C, Flores, A, Cordova, M, Suarez, E, et al. Serum levels for IgM and IgA antibodies to anti-trypanosoma cruzi in samples of blood from newborns from mothers with positive serology for Chagas disease. Rev Soc Bras Med Trop. 2005; 38 (Suppl. 2): 62–4.Google Scholar
Baud, D, Gubler, DJ, Schaub, B, Lanteri, MC, Musso, D. An update on Zika virus infection. Lancet. 2017; 390: 2099–109.Google Scholar
Weisblum, Y, Oiknine-Djian, E, Vorontsov, OM, Haimov-Kochman, R, Zakay-Rones, Z, Meir, K, et al. Zika Virus infects early- and midgestation human maternal decidual tissues, inducing distinct innate tissue responses in the maternal-fetal interface. J Virol. 2017; 91: e01905–16.Google Scholar
Yockey, LJ, Jurado, KA, Arora, N, Millet, A, Rakib, T, Milano, KM, et al. Type I interferons instigate fetal demise after Zika virus infection. Sci Immunol. 2018; 3: eaao1680.Google Scholar
Nem de Oliveira Souza, I, Frost, PS, França, JV, Nascimento-Viana, JB, Neris, RLS, Freitas, L, et al. Acute and chronic neurological consequences of early-life Zika virus infection in mice. Sci Transl Med. 2018; 10: eaar2749.Google Scholar
Dauby, N, Goetghebuer, T, Kollmann, TR, Levy, J, Marchant, A. Uninfected but not unaffected: chronic maternal infections during pregnancy, fetal immunity, and susceptibility to postnatal infections. Lancet Infect Dis. 2012; 12: 330–40.Google Scholar
Abu Raya, B, Smolen, K, Willems, F, Kollmann, T, Marchant, A. Transfer of maternal anti-microbial immunity to HIV-exposed uninfected newborns. Front Immunol. 2016; 7: 338.CrossRefGoogle Scholar
Slogrove, AL, Goetghebuer, T, Cotton, MF, Singer, J, Bettinger, JA. Pattern of infectious morbidity in HIV-exposed uninfected infants and children. Front Immunol. 2016; 7: 164.Google Scholar

References

Valeur-Jensen, AK, Pedersen, CB, Westergaard, T, Jensen, IP, Lebech, M, Andersen, PK, et al. Risk factors for parvovirus B19 infection in pregnancy. JAMA. 1999; 281: 1099–105.Google Scholar
Harger, JH, Adler, SP, Koch, WC, Harger, GF. Prospective evaluation of 618 pregnant women exposed to parvovirus B19: risks and symptoms. Obstet Gynecol. 1998; 91: 413–20.Google Scholar
Gratacós, E, Torres, PJ, Vidal, J, Antolín, E, Costa, J, Jiménez de Anta, MT, et al. The incidence of human parvovirus B19 infection during pregnancy and its impact on perinatal outcome. J Infect Dis. 1995; 171: 1360–3.Google Scholar
Enders, M, Weidner, A, Zoellner, I, Searle, K, Enders, G. Fetal morbidity and mortality after acute human parvovirus B19 infection in pregnancy: prospective evaluation of 1018 cases. Prenat Diagn. 2004; 24: 513–18.Google Scholar
Enders, M, Klingel, K, Weidner, A, Baisch, C, Kandolf, R, Schalasta, G, et al. Risk of fetal hydrops and non-hydropic late intrauterine fetal death after gestational parvovirus B19 infection. J Clin Virol. 2010; 49: 163–8.Google Scholar
Nyman, M, Tolfvenstam, T, Petersson, K, Krassny, C, Skjöldebrand-Sparre, L, Broliden, K. Detection of human parvovirus B19 infection in first-trimester fetal loss. Obstet Gynecol. 2002; 99: 795–8.Google ScholarPubMed
Lassen, J, Jensen, AKV, Bager, P, Pedersen, CB, Panum, I, Nørgaard-Pedersen, B, et al. Parvovirus B19 infection in the first trimester of pregnancy and risk of fetal loss: a population-based case-control study. Am J Epidemiol. 2012; 176: 803–7.Google Scholar
Tolfvenstam, T, Papadogiannakis, N, Norbeck, O, Petersson, K, Broliden, K. Frequency of human parvovirus B19 infection in intrauterine fetal death. Lancet. 2001; 357: 1494–7.Google Scholar
de Haan, TR, van den Akker, ESA, Porcelijn, L, Oepkes, D, Kroes, ACM, Walther, FJ. Thrombocytopenia in hydropic fetuses with parvovirus B19 infection: incidence, treatment and correlation with fetal B19 viral load. BJOG. 2008; 115: 7681.Google Scholar
Melamed, N, Whittle, W, Kelly, EN, Windrim, R, Seaward, PGR, Keunen, J, et al. Fetal thrombocytopenia in pregnancies with fetal human parvovirus-B19 infection. Am J Obstet Gynecol. 2015; 212: 793. e18.Google Scholar
Rogers, BB, Over, CE. Parvovirus B19 in fetal hydrops. Hum Pathol. 1999; 30: 247.Google Scholar
von Kaisenberg, CS, Bender, G, Scheewe, J, Hirt, SW, Lange, M, Stieh, J, et al. A case of fetal parvovirus B19 myocarditis, terminal cardiac heart failure, and perinatal heart transplantation. Fetal Diagn Ther. 2001; 16: 427–32.Google Scholar
Brochot, C, Collinet, P, Provost, N, Subtil, D. Mirror syndrome due to parvovirus B19 hydrops complicated by severe maternal pulmonary effusion. Prenat Diagn. 2006; 26: 179–80.Google Scholar
Carbillon, L, Oury, JF, Guerin, JM, Azancot, A, Blot, P. Clinical biological features of Ballantyne syndrome and the role of placental hydrops. Obstet Gynecol Surv. 1997; 52: 310–14.Google Scholar
Fairley, CK, Smoleniec, JS, Caul, OE, Miller, E. Observational study of effect of intrauterine transfusions on outcome of fetal hydrops after parvovirus B19 infection. Lancet. 1995; 346: 1335–7.Google Scholar
Maisonneuve, E, Garel, C, Friszer, S, Pénager, C, Carbonne, B, Pernot, F, et al. Fetal brain injury associated with parvovirus B19 congenital infection requiring intrauterine transfusion. Fetal Diagn Ther. 2018; 20; 111.Google Scholar
Lindenburg, ITM, van Klink, JM, Smits-Wintjens, VEHJ, van Kamp, IL, Oepkes, D, Lopriore, E. Long-term neurodevelopmental and cardiovascular outcome after intrauterine transfusions for fetal anaemia: a review. Prenat Diagn. 2013; 33: 815–22.Google Scholar
Mari, G, Deter, RL, Carpenter, RL, Rahman, F, Zimmerman, R, Moise, KJ, et al. Noninvasive diagnosis by Doppler ultrasonography of fetal anemia due to maternal red-cell alloimmunization. Collaborative Group for Doppler Assessment of the Blood Velocity in Anemic Fetuses. N Engl J Med. 2000; 342: 914.CrossRefGoogle Scholar
Mari, G, Detti, L, Oz, U, Zimmerman, R, Duerig, P, Stefos, T. Accurate prediction of fetal hemoglobin by Doppler ultrasonography. Obstet Gynecol. 2002; 99: 589–93.Google Scholar
Delle Chiaie, L, Buck, G, Grab, D, Terinde, R. Prediction of fetal anemia with Doppler measurement of the middle cerebral artery peak systolic velocity in pregnancies complicated by maternal blood group alloimmunization or parvovirus B19 infection. Ultrasound Obstet Gynecol. 2001; 18: 232–6.Google Scholar
Völker, F, Cooper, P, Bader, O, Uy, A, Zimmermann, O, Lugert, R, et al. Prevalence of pregnancy-relevant infections in a rural setting of Ghana. BMC Pregnancy Childbirth. 2017; 17: 172.Google Scholar
Pembrey, L, Raynor, P, Griffiths, P, Chaytor, S, Wright, J, Hall, AJ. Seroprevalence of cytomegalovirus, Epstein Barr virus and varicella zoster virus among pregnant women in Bradford: a cohort study. PLoS ONE. 2013; 8: e81881.Google Scholar
Zhang, HJ, Patenaude, V, Abenhaim, HA. Maternal outcomes in pregnancies affected by varicella zoster virus infections: population-based study on 7.7 million pregnancy admissions. J Obstet Gynaecol Res. 2015; 41: 62–8.Google Scholar
Weber, DM, Pellecchia, JA. Varicella pneumonia: study of prevalence in adult men. JAMA. 1965; 192: 572–3.Google Scholar
Mirouse, A, Vignon, P, Piron, P, Robert, R, Papazian, L, Géri, G, et al. Severe varicella-zoster virus pneumonia: a multicenter cohort study. Crit Care. 2017; 21:137.Google Scholar
Ellis, ME, Neal, KR, Webb, AK. Is smoking a risk factor for pneumonia in adults with chickenpox? Br Med J Clin Res Ed. 1987; 294:1002.Google Scholar
Harris, RE, Rhoades, ER. Varicella pneumonia complicating pregnancy: report of a case and review of the literature. Obstet Gynecol. 1965; 25: 734–40.Google Scholar
Harger, JH, Ernest, JM, Thurnau, GR, Moawad, A, Momirova, V, Landon, MB, et al. Risk factors and outcome of varicella-zoster virus pneumonia in pregnant women. J Infect Dis. 2002; 185: 422–7.Google Scholar
Trotta, M, Borchi, B, Niccolai, A, Venturini, E, Giaché, S, Sterrantino, G, et al. Epidemiology, management and outcome of varicella in pregnancy: a 20-year experience at the Tuscany Reference Centre for Infectious Diseases in Pregnancy. Infection. 2018; 46: 693–9.Google Scholar
Zambrano, MA, Martínez, A, Mínguez, JA, Vázquez, F, Palencia, R. Varicella pneumonia complicating pregnancy. Acta Obstet Gynecol Scand. 1995; 74: 318–20.Google Scholar
Field, N, Amirthalingam, G, Waight, P, Andrews, N, Ladhani, SN, van Hoek, AJ, et al. Validity of a reported history of chickenpox in targeting varicella vaccination at susceptible adolescents in England. Vaccine. 2014; 32: 1213–17.Google Scholar
Chris Maple, PA, Gunn, A, Sellwood, J, Brown, DWG, Gray, JJ. Comparison of fifteen commercial assays for detecting Varicella Zoster virus IgG with reference to a time resolved fluorescence immunoassay (TRFIA) and the performance of two commercial assays for screening sera from immunocompromised individuals. J Virol Methods. 2009; 155: 143–9.Google Scholar
Chris Maple, PA, Gray, J, Brown, K, Brown, D. Performance characteristics of a quantitative, standardised varicella zoster IgG time resolved fluorescence immunoassay (VZV TRFIA) for measuring antibody following natural infection. J Virol Methods. 2009; 157: 90–2.Google Scholar
Maple, PA, Rathod, P, Smit, E, Gray, J, Brown, D, Boxall, EH. Comparison of the performance of the LIAISON VZV-IgG and VIDAS automated enzyme linked fluorescent immunoassays with reference to a VZV-IgG time-resolved fluorescence immunoassay and implications of choice of cut-off for LIAISON assay. J Clin Virol. 2009; 44: 914.CrossRefGoogle ScholarPubMed
Boxall, EH, Maple, PA, Rathod, P, Smit, E. Follow-up of pregnant women exposed to chicken pox: an audit of relationship between level of antibody and development of chicken pox. Eur J Clin Microbiol Infect Dis. 2011; 30: 1193–200.CrossRefGoogle ScholarPubMed
Enders, G, Miller, E, Cradock-Watson, J, Bolley, I, Ridehalgh, M. Consequences of varicella and herpes zoster in pregnancy: prospective study of 1739 cases. Lancet. 1994; 343: 1548–51.Google Scholar
Pastuszak, AL, Levy, M, Schick, B, Zuber, C, Feldkamp, M, Gladstone, J, et al. Outcome after maternal varicella infection in the first 20 weeks of pregnancy. N Engl J Med. 1994; 330: 901–5.Google Scholar
Mouly, F, Mirlesse, V, Méritet, JF, Rozenberg, F, Poissonier, MH, Lebon, P, et al. Prenatal diagnosis of fetal varicella-zoster virus infection with polymerase chain reaction of amniotic fluid in 107 cases. Am J Obstet Gynecol. 1997; 177: 894–8.Google Scholar
Preblud, SR. Age-specific risks of varicella complications. Pediatrics. 1981; 68: 1417.Google Scholar
Meyers, JD. Congenital varicella in term infants: risk reconsidered. J Infect Dis. 1974; 129: 215–17.Google Scholar
Miller, E, Cradock-Watson, JE, Ridehalgh, MK. Outcome in newborn babies given anti-varicella-zoster immunoglobulin after perinatal maternal infection with varicella-zoster virus. Lancet. 1989; 2: 371–3.Google Scholar
Pretorius, DH, Hayward, I, Jones, KL, Stamm, E. Sonographic evaluation of pregnancies with maternal varicella infection. J Ultrasound Med. 1992; 11: 459–63.Google Scholar
Pons, JC, Rozenberg, F, Imbert, MC, Lebon, P, Olivennes, F, Lelaidier, C, et al. Prenatal diagnosis of second-trimester congenital varicella syndrome. Prenat Diagn. 1992; 12: 975–6.Google Scholar
Lamont, RF, Sobel, JD, Carrington, D, Mazaki-Tovi, S, Kusanovic, JP, Vaisbuch, E, et al. Varicella-zoster virus (chickenpox) infection in pregnancy. BJOG Int J Obstet Gynaecol. 2011; 118: 1155–62.Google Scholar
Koren, G, Money, D, Boucher, M, Aoki, F, Petric, M, Innocencion, G, et al. Serum concentrations, efficacy, and safety of a new, intravenously administered varicella zoster immune globulin in pregnant women. J Clin Pharmacol. 2002; 42: 267–74.Google Scholar
Winsnes, R. Efficacy of zoster immunoglobulin in prophylaxis of varicella in high-risk patients. Acta Paediatr Scand. 1978; 67: 7782.Google Scholar
Pasternak, B, Hviid, A. Use of acyclovir, valacyclovir, and famciclovir in the first trimester of pregnancy and the risk of birth defects. JAMA. 2010; 304: 859–66.Google Scholar
Marin, M, Willis, ED, Marko, A, Rasmussen, SA, Bialek, SR, Dana, A, et al. Closure of varicella-zoster virus-containing vaccines pregnancy registry - United States, 2013. MMWR Morb Mortal Wkly Rep. 2014; 63: 732–3.Google Scholar
Bohlke, K, Galil, K, Jackson, LA, Schmid, DS, Starkovich, P, Loparev, VN, et al. Postpartum varicella vaccination: is the vaccine virus excreted in breast milk? Obstet Gynecol. 2003; 102: 970–7.Google Scholar
Boppana, SB, Ross, SA, Fowler, KB. Congenital cytomegalovirus infection: clinical outcome. Clin Infect Dis. 2013; 57 (Suppl. 4): S178–181.Google Scholar
Goderis, J, De Leenheer, E, Smets, K, Van Hoecke, H, Keymeulen, A, Dhooge, I. Hearing loss and congenital CMV infection: a systematic review. Pediatrics. 2014; 134: 972–82.Google Scholar
Smithers-Sheedy, H, Raynes-Greenow, C, Badawi, N, Fernandez, MA, Kesson, A, McIntyre, S, et al. Congenital Cytomegalovirus among children with cerebral palsy. J Pediatr. 2017; 181: 267–71. e1.Google Scholar
Kenneson, A, Cannon, MJ. Review and meta-analysis of the epidemiology of congenital cytomegalovirus (CMV) infection. Rev Med Virol. 2007; 17: 253–76.Google Scholar
Dollard, SC, Grosse, SD, Ross, DS. New estimates of the prevalence of neurological and sensory sequelae and mortality associated with congenital cytomegalovirus infection. Rev Med Virol. 2007; 17: 355–63.Google Scholar
Townsend, CL, Forsgren, M, Ahlfors, K, Ivarsson, S-A, Tookey, PA, Peckham, CS. Long-term outcomes of congenital cytomegalovirus infection in Sweden and the United Kingdom. Clin Infect Dis. 2013; 56: 1232–9.Google Scholar
Gratacap-Cavallier, B, Bosson, JL, Morand, P, Dutertre, N, Chanzy, B, Jouk, PS, et al. Cytomegalovirus seroprevalence in French pregnant women: parity and place of birth as major predictive factors. Eur J Epidemiol. 1998; 14: 147–52.Google Scholar
Colugnati, FAB, Staras, SAS, Dollard, SC, Cannon, MJ. Incidence of cytomegalovirus infection among the general population and pregnant women in the United States. BMC Infect Dis. 2007; 7: 71.Google Scholar
Ross, SA, Arora, N, Novak, Z, Fowler, KB, Britt, WJ, Boppana, SB. Cytomegalovirus reinfections in healthy seroimmune women. J Infect Dis. 2010; 201: 386–9.Google Scholar
Leruez-Ville, M, Magny, J-F, Couderc, S, Pichon, C, Parodi, M, Bussières, L, et al. Risk factors for congenital cytomegalovirus infection following primary and nonprimary maternal infection: a prospective neonatal screening study using polymerase chain reaction in saliva. Clin Infect Dis. 2017; 65: 398404.Google Scholar
Mussi-Pinhata, MM, Yamamoto, AY, Moura Brito, RM, de Lima, IM, de Carvalho e Oliveira, PF, Boppana, S, et al. Birth prevalence and natural history of congenital cytomegalovirus infection in a highly seroimmune population. Clin Infect Dis. 2009; 49: 522–8.Google Scholar
Ross, SA, Fowler, KB, Ashrith, G, Stagno, S, Britt, WJ, Pass, RF, et al. Hearing loss in children with congenital cytomegalovirus infection born to mothers with preexisting immunity. J Pediatr. 2006; 148: 332–6.Google Scholar
Picone, O, Vauloup-Fellous, C, Cordier, A-G, Parent Du Châtelet, I, Senat, M-V, Frydman, R, et al. A 2-year study on cytomegalovirus infection during pregnancy in a French hospital. BJOG. 2009; 116: 818–23.Google Scholar
Leruez-Ville, M, Sellier, Y, Salomon, LJ, Stirnemann, JJ, Jacquemard, F, Ville, Y. Prediction of fetal infection in cases with cytomegalovirus immunoglobulin M in the first trimester of pregnancy: a retrospective cohort. Clin Infect Dis. 2013; 56: 1428–35.Google Scholar
Delforge, ML, Desomberg, L, Montesinos, I. Evaluation of the new LIAISON(®) CMV IgG, IgM and IgG Avidity II assays. J Clin Virol. 2015; 72: 42–5.Google Scholar
Sellier, Y, Guilleminot, T, Ville, Y, Leruez-Ville, M. Comparison of the LIAISON(®) CMV IgG Avidity II and the VIDAS(®) CMV IgG Avidity II assays for the diagnosis of primary infection in pregnant women. J Clin Virol. 2015; 72: 46–8.Google Scholar
Chiereghin, A, Pavia, C, Gabrielli, L, Piccirilli, G, Squarzoni, D, Turello, G, et al. Clinical evaluation of the new Roche platform of serological and molecular cytomegalovirus-specific assays in the diagnosis and prognosis of congenital cytomegalovirus infection. J Virol Methods. 2017; 248: 250–4.Google Scholar
Bodéus, M, Hubinont, C, Bernard, P, Bouckaert, A, Thomas, K, Goubau, P. Prenatal diagnosis of human cytomegalovirus by culture and polymerase chain reaction: 98 pregnancies leading to congenital infection. Prenat Diagn. 1999; 19: 314–17.Google Scholar
Enders, M, Daiminger, A, Exler, S, Ertan, K, Enders, G, Bald, R. Prenatal diagnosis of congenital cytomegalovirus infection in 115 cases: a 5 years’ single center experience. Prenat Diagn. 2017; 37: 389–98.Google Scholar
Revello, MG, Furione, M, Rognoni, V, Arossa, A, Gerna, G. Cytomegalovirus DNAemia in pregnant women. J Clin Virol. 2014; 61: 590–2.Google Scholar
Bilavsky, E, Pardo, J, Attias, J, Levy, I, Magny, J-F, Ville, Y, et al. Clinical Implications for children born with congenital cytomegalovirus infection following a negative amniocentesis. Clin Infect Dis. 2016; 63: 33–8.Google Scholar
Revello, MG, Furione, M, Zavattoni, M, Tassis, B, Nicolini, U, Fabbri, E, et al. Human cytomegalovirus (HCMV) DNAemia in the mother at amniocentesis as a risk factor for iatrogenic HCMV infection of the fetus. J Infect Dis. 2008; 197: 593–6.Google Scholar
Boppana, SB, Pass, RF, Britt, WJ, Stagno, S, Alford, CA. Symptomatic congenital cytomegalovirus infection: neonatal morbidity and mortality. Pediatr Infect Dis J. 1992; 11: 93–9.Google Scholar
Anderson, KS, Amos, CS, Boppana, S, Pass, R. Ocular abnormalities in congenital cytomegalovirus infection. J Am Optom Assoc. 1996; 67: 273–8.Google Scholar
Fowler, KB, Boppana, SB. Congenital cytomegalovirus (CMV) infection and hearing deficit. J Clin Virol. 2006; 35: 226–31.Google Scholar
Barbi, M, Binda, S, Caroppo, S, Ambrosetti, U, Corbetta, C, Sergi, P. A wider role for congenital cytomegalovirus infection in sensorineural hearing loss. Pediatr Infect Dis J. 2003; 22: 3942.Google Scholar
Avettand-Fenoël, V, Marlin, S, Vauloup-Fellous, C, Loundon, N, François, M, Couloigner, V, et al. Congenital cytomegalovirus is the second most frequent cause of bilateral hearing loss in young French children. J Pediatr. 2013; 162: 593–9.Google Scholar
Binda, S, Caroppo, S, Didò, P, Primache, V, Veronesi, L, Calvario, A, et al. Modification of CMV DNA detection from dried blood spots for diagnosing congenital CMV infection. J Clin Virol. 2004; 30: 276–9.Google Scholar
Leruez-Ville, M, Vauloup-Fellous, C, Couderc, S, Parat, S, Castel, C, Avettand-Fenoel, V, et al. Prospective identification of congenital cytomegalovirus infection in newborns using real-time polymerase chain reaction assays in dried blood spots. Clin Infect Dis. 2011; 52: 575–81.Google Scholar
Pass, RF, Fowler, KB, Boppana, SB, Britt, WJ, Stagno, S. Congenital cytomegalovirus infection following first trimester maternal infection: symptoms at birth and outcome. J Clin Virol. 2006; 35: 216–20.Google Scholar
Foulon, I, Naessens, A, Foulon, W, Casteels, A, Gordts, F. A 10-year prospective study of sensorineural hearing loss in children with congenital cytomegalovirus infection. J Pediatr. 2008; 153: 84–8.Google Scholar
Lipitz, S, Yinon, Y, Malinger, G, Yagel, S, Levit, L, Hoffman, C, et al. Risk of cytomegalovirus-associated sequelae in relation to time of infection and findings on prenatal imaging. Ultrasound Obstet Gynecol. 2013; 41: 508–14.Google Scholar
Enders, G, Daiminger, A, Bäder, U, Exler, S, Enders, M. Intrauterine transmission and clinical outcome of 248 pregnancies with primary cytomegalovirus infection in relation to gestational age. J Clin Virol. 2011; 52: 244–6.Google Scholar
Picone, O, Vauloup-Fellous, C, Cordier, AG, Guitton, S, Senat, MV, Fuchs, F, et al. A series of 238 cytomegalovirus primary infections during pregnancy: description and outcome. Prenat Diagn. 2013; 33: 751–8.Google Scholar
Bodéus, M, Hubinont, C, Goubau, P. Increased risk of cytomegalovirus transmission in utero during late gestation. Obstet Gynecol. 1999; 93: 658–60.Google Scholar
Feldman, B, Yinon, Y, Tepperberg Oikawa, M, Yoeli, R, Schiff, E, Lipitz, S. Pregestational, periconceptional, and gestational primary maternal cytomegalovirus infection: prenatal diagnosis in 508 pregnancies. Am J Obstet Gynecol. 2011; 205: 342. e1–6.Google Scholar
Revello, MG, Zavattoni, M, Furione, M, Lilleri, D, Gorini, G, Gerna, G. Diagnosis and outcome of preconceptional and periconceptional primary human cytomegalovirus infections. J Infect Dis. 2002; 186: 553–7.Google Scholar
Guerra, B, Simonazzi, G, Puccetti, C, Lanari, M, Farina, A, Lazzarotto, T, et al. Ultrasound prediction of symptomatic congenital cytomegalovirus infection. Am J Obstet Gynecol. 2008; 198: 380. e1–7.Google Scholar
Malinger, G, Lev, D, Lerman-Sagie, T. Imaging of fetal cytomegalovirus infection. Fetal Diagn Ther. 2011; 29: 117–26.Google Scholar
Nigro, G, La Torre, R, Sali, E, Auteri, M, Mazzocco, M, Maranghi, L, et al. Intraventricular haemorrhage in a fetus with cerebral cytomegalovirus infection. Prenat Diagn. 2002; 22: 558–61.CrossRefGoogle Scholar
Enders, G, Bäder, U, Lindemann, L, Schalasta, G, Daiminger, A. Prenatal diagnosis of congenital cytomegalovirus infection in 189 pregnancies with known outcome. Prenat Diagn. 2001; 21: 362–77.Google Scholar
Picone, O, Vauloup-Fellous, C, Cordier, AG, Grangeot-Keros, L, Frydman, R, Senat, MV. Late onset of ultrasound abnormalities in a case of periconceptional congenital cytomegalovirus infection. Ultrasound Obstet Gynecol. 2008; 31: 481–3.Google Scholar
Garel, C, Chantrel, E, Brisse, H, Elmaleh, M, Luton, D, Oury, JF, et al. Fetal cerebral cortex: normal gestational landmarks identified using prenatal MR imaging. AJNR Am J Neuroradiol. 2001; 22: 184–9.Google Scholar
Barkovich, AJ, Lindan, CE. Congenital cytomegalovirus infection of the brain: imaging analysis and embryologic considerations. AJNR Am J Neuroradiol. 1994; 15: 703–15.Google Scholar
Benoist, G, Salomon, LJ, Mohlo, M, Suarez, B, Jacquemard, F, Ville, Y. Cytomegalovirus-related fetal brain lesions: comparison between targeted ultrasound examination and magnetic resonance imaging. Ultrasound Obstet Gynecol. 2008; 32: 900–5.CrossRefGoogle ScholarPubMed
Picone, O, Simon, I, Benachi, A, Brunelle, F, Sonigo, P. Comparison between ultrasound and magnetic resonance imaging in assessment of fetal cytomegalovirus infection. Prenat Diagn. 2008; 28: 753–8.Google Scholar
Benoist, G, Salomon, LJ, Jacquemard, F, Daffos, F, Ville, Y. The prognostic value of ultrasound abnormalities and biological parameters in blood of fetuses infected with cytomegalovirus. BJOG. 2008; 115: 823–9.CrossRefGoogle ScholarPubMed
Farkas, N, Hoffmann, C, Ben-Sira, L, Lev, D, Schweiger, A, Kidron, D, et al. Does normal fetal brain ultrasound predict normal neurodevelopmental outcome in congenital cytomegalovirus infection? Prenat Diagn. 2011; 31: 360–6.Google Scholar
Leruez-Ville, M, Stirnemann, J, Sellier, Y, Guilleminot, T, Dejean, A, Magny, J-F, et al. Feasibility of predicting the outcome of fetal infection with cytomegalovirus at the time of prenatal diagnosis. Am J Obstet Gynecol. 2016; 215: 342. e1–9.Google Scholar
Lipitz, S, Hoffmann, C, Feldman, B, Tepperberg-Dikawa, M, Schiff, E, Weisz, B. Value of prenatal ultrasound and magnetic resonance imaging in assessment of congenital primary cytomegalovirus infection. Ultrasound Obstet Gynecol. 2010; 36: 709–17.Google Scholar
Gouarin, S, Gault, E, Vabret, A, Cointe, D, Rozenberg, F, Grangeot-Keros, L, et al. Real-time PCR quantification of human cytomegalovirus DNA in amniotic fluid samples from mothers with primary infection. J Clin Microbiol. 2002; 40: 1767–72.Google Scholar
Fabbri, E, Revello, MG, Furione, M, Zavattoni, M, Lilleri, D, Tassis, B, et al. Prognostic markers of symptomatic congenital human cytomegalovirus infection in fetal blood. BJOG. 2011; 118: 448–56.Google Scholar
Nigro, G, Adler, SP, La Torre, R, Best, AM, Congenital Cytomegalovirus Collaborating Group. Passive immunization during pregnancy for congenital cytomegalovirus infection. N Engl J Med. 2005; 353: 1350–62.Google Scholar
Revello, MG, Lazzarotto, T, Guerra, B, Spinillo, A, Ferrazzi, E, Kustermann, A, et al. A randomized trial of hyperimmune globulin to prevent congenital cytomegalovirus. N Engl J Med. 2014; 370: 1316–26.Google Scholar
Lowance, D, Neumayer, HH, Legendre, CM, Squifflet, JP, Kovarik, J, Brennan, PJ, et al. Valacyclovir for the prevention of cytomegalovirus disease after renal transplantation. International Valacyclovir Cytomegalovirus Prophylaxis Transplantation Study Group. N Engl J Med. 1999; 340: 1462–70.Google Scholar
Jacquemard, F, Yamamoto, M, Costa, J-M, Romand, S, Jaqz-Aigrain, E, Dejean, A, et al. Maternal administration of valaciclovir in symptomatic intrauterine cytomegalovirus infection. BJOG. 2007; 114: 1113–21.Google Scholar
Leruez-Ville, M, Ghout, I, Bussières, L, Stirnemann, J, Magny, J-F, Couderc, S, et al. In utero treatment of congenital cytomegalovirus infection with valacyclovir in a multicenter, open-label, phase II study. Am J Obstet Gynecol. 2016; 215: 462. e1–462. e10.Google Scholar
Pappas, G, Roussos, N, Falagas, ME. Toxoplasmosis snapshots: global status of Toxoplasma gondii seroprevalence and implications for pregnancy and congenital toxoplasmosis. Int J Parasitol. 2009; 39: 1385–94.Google Scholar
Rudin, C, Hirsch, HH, Spaelti, R, Schaedelin, S, Klimkait, T. Decline of Seroprevalence and incidence of congenital toxoplasmosis despite changing prevention policy – three decades of cord-blood screening in North-Western Switzerland. Pediatr Infect Dis J. 2018; 37: 1087–92.Google Scholar
Robert-Gangneux, F, Dardé, M-L. Epidemiology of and diagnostic strategies for toxoplasmosis. Clin Microbiol Rev. 2012; 25: 264–96.Google Scholar
Villard, O, Breit, L, Cimon, B, Franck, J, Fricker-Hidalgo, H, Godineau, N, et al. Comparison of four commercially available avidity tests for Toxoplasma gondii-specific IgG antibodies. Clin Vaccine Immunol. 2013; 20: 197204.Google Scholar
Desmonts, G, Couvreur, J. Congenital toxoplasmosis. A prospective study of 378 pregnancies. N Engl J Med. 1974; 290: 1110–16.Google Scholar
Daffos, F, Forestier, F, Capella-Pavlovsky, M, Thulliez, P, Aufrant, C, Valenti, D, et al. Prenatal management of 746 pregnancies at risk for congenital toxoplasmosis. N Engl J Med. 1988; 318: 271–5.CrossRefGoogle ScholarPubMed
Elbez-Rubinstein, A, Ajzenberg, D, Dardé, M-L, Cohen, R, Dumètre, A, Yera, H, et al. Congenital toxoplasmosis and reinfection during pregnancy: case report, strain characterization, experimental model of reinfection, and review. J Infect Dis. 2009; 199: 280–5.CrossRefGoogle ScholarPubMed
SYROCOT (Systematic Review on Congenital Toxoplasmosis) study group, Thiébaut, R, Leproust, S, Chêne, G, Gilbert, R. Effectiveness of prenatal treatment for congenital toxoplasmosis: a meta-analysis of individual patients’ data. Lancet. 2007; 369: 115–22.Google Scholar
de Oliveira Azevedo, CT, do Brasil, PEAA, Guida, L, Lopes Moreira, ME. Performance of Polymerase Chain Reaction Analysis of the Amniotic Fluid of Pregnant Women for Diagnosis of Congenital Toxoplasmosis: A Systematic Review and Meta-Analysis. PLoS ONE. 2016; 11: e0149938.Google Scholar
Filisetti, D, Sterkers, Y, Brenier-Pinchart, M-P, Cassaing, S, Dalle, F, Delhaes, L, et al. Multicentric comparative assessment of the bio-evolution Toxoplasma gondii detection kit with eight laboratory-developed PCR assays for molecular diagnosis of congenital toxoplasmosis. J Clin Microbiol. 2015; 53: 2934.Google Scholar
Costa, JM, Ernault, P, Gautier, E, Bretagne, S. Prenatal diagnosis of congenital toxoplasmosis by duplex real-time PCR using fluorescence resonance energy transfer hybridization probes. Prenat Diagn. 2001; 21: 85–8.Google Scholar
Yamamoto, L, Targa, LS, Sumita, LM, Shimokawa, PT, Rodrigues, JC, Kanunfre, KA, et al. Association of parasite load levels in amniotic fluid with clinical outcome in congenital toxoplasmosis. Obstet Gynecol. 2017; 130: 335–45.Google Scholar
Pratlong, F, Boulot, P, Issert, E, Msika, M, Dupont, F, Bachelard, B, et al. Fetal diagnosis of toxoplasmosis in 190 women infected during pregnancy. Prenat Diagn. 1994; 14: 191–8.CrossRefGoogle ScholarPubMed
Berrebi, A, Bardou, M, Bessieres, M-H, Nowakowska, D, Castagno, R, Rolland, M, et al. Outcome for children infected with congenital toxoplasmosis in the first trimester and with normal ultrasound findings: a study of 36 cases. Eur J Obstet Gynecol Reprod Biol. 2007; 135: 53–7.Google Scholar
Hohlfeld, P, Daffos, F, Thulliez, P, Aufrant, C, Couvreur, J, MacAleese, J, et al. Fetal toxoplasmosis: outcome of pregnancy and infant follow-up after in utero treatment. J Pediatr. 1989; 115: 765–9.Google Scholar
Mombrò, M, Perathoner, C, Leone, A, Nicocia, M, Moiraghi Ruggenini, A, Zotti, C, et al. Congenital toxoplasmosis: 10-year follow up. Eur J Pediatr. 1995; 154: 635–9.Google Scholar
Wilson, CB, Remington, JS, Stagno, S, Reynolds, DW. Development of adverse sequelae in children born with subclinical congenital Toxoplasma infection. Pediatrics. 1980; 66: 767–74.Google Scholar
McAuley, J, Boyer, KM, Patel, D, Mets, M, Swisher, C, Roizen, N, et al. Early and longitudinal evaluations of treated infants and children and untreated historical patients with congenital toxoplasmosis: the Chicago Collaborative Treatment Trial. Clin Infect Dis. 1994; 18: 3872.Google Scholar
Roizen, N, Swisher, CN, Stein, MA, Hopkins, J, Boyer, KM, Holfels, E, et al. Neurologic and developmental outcome in treated congenital toxoplasmosis. Pediatrics. 1995; 95: 1120.Google Scholar
Patel, DV, Holfels, EM, Vogel, NP, Boyer, KM, Mets, MB, Swisher, CN, et al. Resolution of intracranial calcifications in infants with treated congenital toxoplasmosis. Radiology. 1996; 199: 433–40.Google Scholar
Peyron, F, Wallon, M, Bernardoux, C. Long-term follow-up of patients with congenital ocular toxoplasmosis. N Engl J Med. 1996; 334: 993–4.Google Scholar
Guerina, NG, Hsu, HW, Meissner, HC, Maguire, JH, Lynfield, R, Stechenberg, B, et al. Neonatal serologic screening and early treatment for congenital Toxoplasma gondii infection. The New England Regional Toxoplasma Working Group. N Engl J Med. 1994; 330: 1858–63.Google Scholar
Koppe, JG, Loewer-Sieger, DH, de Roever-Bonnet, H. Results of 20-year follow-up of congenital toxoplasmosis. Lancet. 1986; 1: 254–6.Google Scholar
Montazeri, M, Sharif, M, Sarvi, S, Mehrzadi, S, Ahmadpour, E, Daryani, A. A systematic review of in vitro and in vivo activities of anti-toxoplasma drugs and compounds (2006-2016). Front Microbiol. 2017; 8: 25.Google Scholar
van der Ven, AJ, Schoondermark-van de Ven, EM, Camps, W, Melchers, WJ, Koopmans, PP, van der Meer, JW, et al. Anti-toxoplasma effect of pyrimethamine, trimethoprim and sulphonamides alone and in combination: implications for therapy. J Antimicrob Chemother. 1996; 38: 7580.Google Scholar
Hotop, A, Hlobil, H, Gross, U. Efficacy of rapid treatment initiation following primary Toxoplasma gondii infection during pregnancy. Clin Infect Dis. 2012; 54: 1545–52.Google Scholar
Prusa, A-R, Kasper, DC, Sawers, L, Walter, E, Hayde, M, Stillwaggon, E. Congenital toxoplasmosis in Austria: Prenatal screening for prevention is cost-saving. PLoS Negl Trop Dis. 2017; 11: e0005648.Google Scholar
Mandelbrot, L, Kieffer, F, Sitta, R, Laurichesse-Delmas, H, Winer, N, Mesnard, L, et al. Prenatal therapy with pyrimethamine + sulfadiazine vs spiramycin to reduce placental transmission of toxoplasmosis: a multicenter, randomized trial. Am J Obstet Gynecol. 2018; 219: 386. e1386.Google Scholar
Rothova, A, Meenken, C, Buitenhuis, HJ, Brinkman, CJ, Baarsma, GS, Boen-Tan, TN, et al. Therapy for ocular toxoplasmosis. Am J Ophthalmol. 1993; 115: 517–23.Google Scholar
McCabe, R, Remington, JS. Toxoplasmosis: the time has come. N Engl J Med. 1988; 318: 313–15.Google Scholar
Foulon, W, Naessens, A, Lauwers, S, De Meuter, F, Amy, JJ. Impact of primary prevention on the incidence of toxoplasmosis during pregnancy. Obstet Gynecol. 1988; 72: 363–6.Google Scholar
Bouthry, E, Picone, O, Hamdi, G, Grangeot-Keros, L, Ayoubi, J-M, Vauloup-Fellous, C. Rubella and pregnancy: diagnosis, management and outcomes. Prenat Diagn. 2014; 34: 1246–53.Google Scholar
Dimech, W, Grangeot-Keros, L, Vauloup-Fellous, C. Standardization of assays that detect anti-rubella virus IgG antibodies. Clin Microbiol Rev. 2016; 29: 163–74.Google Scholar
Miller, E, Cradock-Watson, JE, Pollock, TM. Consequences of confirmed maternal rubella at successive stages of pregnancy. Lancet. 1982; 2: 781–4.Google Scholar
Daffos, F, Forestier, F, Grangeot-Keros, L, Capella Pavlovsky, M, Lebon, P, Chartier, M, et al. Prenatal diagnosis of congenital rubella. Lancet. 1984; 2: 13.Google Scholar
Enders, G, Nickerl-Pacher, U, Miller, E, Cradock-Watson, JE. Outcome of confirmed periconceptional maternal rubella. Lancet. 1988; 1: 1445–7.Google Scholar
Givens, KT, Lee, DA, Jones, T, Ilstrup, DM. Congenital rubella syndrome: ophthalmic manifestations and associated systemic disorders. Br J Ophthalmol. 1993; 77: 358–63.Google Scholar
Forrest, JM, Turnbull, FM, Sholler, GF, Hawker, RE, Martin, FJ, Doran, TT, et al. Gregg’s congenital rubella patients 60 years later. Med J Aust. 2002; 177: 664–7.Google Scholar
O’Neill, JF. The ocular manifestations of congenital infection: a study of the early effect and long-term outcome of maternally transmitted rubella and toxoplasmosis. Trans Am Ophthalmol Soc. 1998; 96: 813–79.Google Scholar
Macé, M, Cointe, D, Six, C, Levy-Bruhl, D, Parent du Chatelet, I, Ingrand, D, et al. Diagnostic value of reverse transcription PCR of amniotic fluid for prenatal diagnosis of congenital rubella infection in pregnant women with confirmed primary rubella infection. J Clin Microbiol; 2004; 42: 4818–20.Google Scholar

References

Boers, KE, Vijgen, SM, Bijlenga, D, van der Post, JA, Bekedam, DJ, Kwee, A, et al. Induction versus expectant monitoring for intrauterine growth restriction at term: randomised equivalence trial (DIGITAT). BMJ. 2010; 341: c7087.Google Scholar
Audette, MC, Kingdom, JC. Screening for fetal growth restriction and placental insufficiency. Semin Fetal Neonatal Med. 2018; 23: 119–25.Google Scholar
Gaccioli, F, Aye, I, Sovio, U, Charnock-Jones, DS, Smith, GCS. Screening for fetal growth restriction using fetal biometry combined with maternal biomarkers. Am J Obstet Gynecol. 2018; 218: S725–37.Google Scholar
Kingdom, JC, Audette, MC, Hobson, SR, Windrim, RC, Morgen, E. A placenta clinic approach to the diagnosis and management of fetal growth restriction. Am J Obstet Gynecol. 2018; 218: S803–17.Google Scholar
Jackson, MR, Walsh, AJ, Morrow, RJ, Mullen, JB, Lye, SJ, Ritchie, JW. Reduced placental villous tree elaboration in small-for-gestational-age pregnancies: relationship with umbilical artery Doppler waveforms. Am J Obstet Gynecol. 1995; 172: 518–25.Google Scholar
Robson, SC, Simpson, H, Ball, E, Lyall, F, Bulmer, JN. Punch biopsy of the human placental bed. Am J Obstet Gynecol. 2002; 187: 1349–55.Google Scholar
Burton, GJ. Oxygen, the Janus gas; its effects on human placental development and function. J Anat. 2009; 215: 2735.Google Scholar
Burton, GJ, Watson, AL, Hempstock, J, Skepper, JN, Jauniaux, E. Uterine glands provide histiotrophic nutrition for the human fetus during the first trimester of pregnancy. J Clin Endocrinol Metab. 2002; 87: 2954–9.Google Scholar
Burton, GJ, Jauniaux, E, Watson, AL. Maternal arterial connections to the placental intervillous space during the first trimester of human pregnancy: the Boyd collection revisited. Am J Obstet Gynecol. 1999; 181: 718–24.Google Scholar
Burton, GJ, Hempstock, J, Jauniaux, E. Oxygen, early embryonic metabolism and free radical-mediated embryopathies. Reprod Biomed Online. 2003; 6: 8496.Google Scholar
Jauniaux, E, Hempstock, J, Greenwold, N, Burton, GJ. Trophoblastic oxidative stress in relation to temporal and regional differences in maternal placental blood flow in normal and abnormal early pregnancies. Am J Pathol. 2003; 162: 115–25.Google Scholar
Kaufmann, P, Black, S, Huppertz, B. Endovascular trophoblast invasion: implications for the pathogenesis of intrauterine growth retardation and preeclampsia. Biol Reprod. 2003; 69: 17.Google Scholar
Burton, GJ, Jauniaux, E, Charnock-Jones, DS. The influence of the intrauterine environment on human placental development. Int J Dev Biol. 2010; 54: 303–12.Google Scholar
Nordenvall, M, Ullberg, U, Laurin, J, Lingman, G, Sandstedt, B, Ulmsten, U. Placental morphology in relation to umbilical artery blood velocity waveforms. Eur J Obstet Gynecol Reprod Biol. 1991; 40: 179–90.CrossRefGoogle ScholarPubMed
Proctor, LK, Toal, M, Keating, S, Chitayat, D, Okun, N, Windrim, RC, et al. Placental size and the prediction of severe early-onset intrauterine growth restriction in women with low pregnancy-associated plasma protein-A. Ultrasound Obstet Gynecol. 2009; 34: 274–82.Google Scholar
Wright, E, Audette, MC, Ye, XY, Keating, S, Hoffman, B, Lye, SJ, et al. Maternal vascular malperfusion and adverse perinatal outcomes in low-risk nulliparous women. Obstet Gynecol. 2017; 130: 1112–20.Google Scholar
Dunk, C, Smith, S, Hazan, A, Whittle, W, Jones, RL. Promotion of angiogenesis by human endometrial lymphocytes. Immunol Invest. 2008; 37: 583610.Google Scholar
Kadyrov, M, Kingdom, JC, Huppertz, B. Divergent trophoblast invasion and apoptosis in placental bed spiral arteries from pregnancies complicated by maternal anemia and early-onset preeclampsia/intrauterine growth restriction. Am J Obstet Gynecol. 2006; 194: 557–63.Google Scholar
Burton, GJ, Woods, AW, Jauniaux, E, Kingdom, JC. Rheological and physiological consequences of conversion of the maternal spiral arteries for uteroplacental blood flow during human pregnancy. Placenta. 2009; 30: 473–82.Google Scholar
Nanaev, A, Chwalisz, K, Frank, HG, Kohnen, G, Hegele-Hartung, C, Kaufmann, P. Physiological dilation of uteroplacental arteries in the guinea pig depends on nitric oxide synthase activity of extravillous trophoblast. Cell Tissue Res. 1995; 282: 407–21.Google Scholar
Lyall, F, Barber, A, Myatt, L, Bulmer, JN, Robson, SC. Hemeoxygenase expression in human placenta and placental bed implies a role in regulation of trophoblast invasion and placental function. Faseb J. 2000; 14: 208–19.Google Scholar
Kaufmann, P, Mayhew, TM, Charnock-Jones, DS. Aspects of human fetoplacental vasculogenesis and angiogenesis. II. Changes during normal pregnancy. Placenta. 2004; 25: 114–26.Google Scholar
Simmons, DG, Natale, DR, Begay, V, Hughes, M, Leutz, A, Cross, JC. Early patterning of the chorion leads to the trilaminar trophoblast cell structure in the placental labyrinth. Development. 2008; 135: 2083–91.Google Scholar
Simpson, RA, Mayhew, TM, Barnes, PR. From 13 weeks to term, the trophoblast of human placenta grows by the continuous recruitment of new proliferative units: a study of nuclear number using the disector. Placenta. 1992; 13: 501–12.Google Scholar
Baczyk, D, Drewlo, S, Proctor, L, Dunk, C, Lye, S, Kingdom, J. Glial cell missing-1 transcription factor is required for the differentiation of the human trophoblast. Cell Death Differ. 2009; 16: 719–27.Google Scholar
Liang, CY, Wang, LJ, Chen, CP, Chen, LF, Chen, YH, Chen, H. GCM1 regulation of the expression of syncytin 2 and its cognate receptor MFSD2A in human placenta. Biol Reprod. 2010; 83: 387–95.Google Scholar
Baczyk, D, Kibschull, M, Mellstrom, B, Levytska, K, Rivas, M, Drewlo, S, et al. DREAM mediated regulation of GCM1 in the human placental trophoblast. PLoS ONE. 2013; 8: e51837.Google Scholar
Kingdom, JC, Drewlo, S. Is heparin a placental anticoagulant in high-risk pregnancies? Blood. 2011; 118: 4780–8.Google Scholar
Tanaka, S, Kunath, T, Hadjantonakis, AK, Nagy, A, Rossant, J. Promotion of trophoblast stem cell proliferation by FGF4. Science. 1998; 282: 2072–5.Google Scholar
Baczyk, D, Dunk, C, Huppertz, B, Maxwell, C, Reister, F, Giannoulias, D, et al. Bi-potential behaviour of cytotrophoblasts in first trimester chorionic villi. Placenta. 2006; 27: 367–74.Google Scholar
Nosi, U, Lanner, F, Huang, T, Cox, B. Overexpression of trophoblast stem cell-enriched microRNAs promotes trophoblast fate in embryonic stem cells. Cell Rep. 2017; 19: 1101–9.Google Scholar
Macara, L, Kingdom, JC, Kaufmann, P, Kohnen, G, Hair, J, More, IA, et al. Structural analysis of placental terminal villi from growth-restricted pregnancies with abnormal umbilical artery Doppler waveforms. Placenta. 1996; 17: 3748.Google Scholar
Huppertz, B, Frank, HG, Kingdom, JC, Reister, F, Kaufmann, P. Villous cytotrophoblast regulation of the syncytial apoptotic cascade in the human placenta. Histochem Cell Biol. 1998; 110: 495508.Google Scholar
Ellery, PM, Cindrova-Davies, T, Jauniaux, E, Ferguson-Smith, AC, Burton, GJ. Evidence for transcriptional activity in the syncytiotrophoblast of the human placenta. Placenta. 2009; 30: 329–34.Google Scholar
Fogarty, NM, Mayhew, TM, Ferguson-Smith, AC, Burton, GJ. A quantitative analysis of transcriptionally active syncytiotrophoblast nuclei across human gestation. J Anat. 2011; 219: 601–10.Google Scholar
Rajakumar, A, Cerdeira, AS, Rana, S, Zsengeller, Z, Edmunds, L, Jeyabalan, A, et al. Transcriptionally active syncytial aggregates in the maternal circulation may contribute to circulating soluble fms-like tyrosine kinase 1 in preeclampsia. Hypertension. 2012; 59: 256–64.CrossRefGoogle ScholarPubMed
Burton, GJ, Jones, CJ. Syncytial knots, sprouts, apoptosis, and trophoblast deportation from the human placenta. Taiwan J Obstet Gynecol. 2009; 48: 2837.Google Scholar
Parham, P, Guethlein, LA. Pregnancy immunogenetics: NK cell education in the womb? J Clin Invest. 2010; 120: 3801–4.Google Scholar
Munn, DH, Zhou, M, Attwood, JT, Bondarev, I, Conway, SJ, Marshall, B, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998; 281: 1191–3.Google Scholar
Walker, MG, Fitzgerald, B, Keating, S, Ray, JG, Windrim, R, Kingdom, JCP. Sex-specific basis of severe placental dysfunction leading to extreme preterm delivery. Placenta. 2011; 33: 568–71.Google Scholar
Saraswat, L, Bhattacharya, S, Maheshwari, A. Maternal and perinatal outcome in women with threatened miscarriage in the first trimester: a systematic review. BJOG. 2010; 117: 245–57.Google Scholar
Porat, S, Fitzgerald, B, Wright, E, Keating, S, Kingdom, JC. Placental hyperinflation and the risk of adverse perinatal outcome. Ultrasound Obstet Gynecol. 2013; 42: 315–21.Google Scholar
Fitzgerald, B, Shannon, P, Kingdom, J, Keating, S. Rounded intraplacental haematomas due to decidual vasculopathy have a distinctive morphology. J Clin Pathol. 2011; 64: 729–32.Google Scholar
Korzeniewski, SJ, Romero, R, Chaiworapongsa, T, Chaemsaithong, P, Kim, CJ, Kim, YM, et al. Maternal plasma angiogenic index-1 (placental growth factor/soluble vascular endothelial growth factor receptor-1) is a biomarker for the burden of placental lesions consistent with uteroplacental underperfusion: a longitudinal case-cohort study. Am J Obstet Gynecol. 2016; 214: 629. e1–e17.Google Scholar
Walker, MG, Fitzgerald, B, Keating, S, Ray, JG, Windrim, R, Kingdom, JC. Sex-specific basis of severe placental dysfunction leading to extreme preterm delivery. Placenta. 2012; 33: 568–71.Google Scholar
Krebs, C, Macara, LM, Leiser, R, Bowman, AW, Greer, IA, Kingdom, JC. Intrauterine growth restriction with absent end-diastolic flow velocity in the umbilical artery is associated with maldevelopment of the placental terminal villous tree. Am J Obstet Gynecol. 1996; 175: 1534–42.Google Scholar
Fitzgerald, B, Levytska, K, Kingdom, J, Walker, M, Baczyk, D, Keating, S. Villous trophoblast abnormalities in extremely preterm deliveries with elevated second trimester maternal serum hCG or inhibin-A. Placenta. 2011; 32: 339–45.Google Scholar
Yung, HW, Calabrese, S, Hynx, D, Hemmings, BA, Cetin, I, Charnock-Jones, DS, et al. Evidence of placental translation inhibition and endoplasmic reticulum stress in the etiology of human intrauterine growth restriction. Am J Pathol. 2008; 173: 451–62.Google Scholar
Sharp, AN, Heazell, AE, Baczyk, D, Dunk, CE, Lacey, HA, Jones, CJ, et al. Preeclampsia is associated with alterations in the p53-pathway in villous trophoblast. PLoS ONE. 2014; 9: e87621.Google Scholar
Ray, JE, Garcia, J, Jurisicova, A, Caniggia, I. Mtd/Bok takes a swing: proapoptotic Mtd/Bok regulates trophoblast cell proliferation during human placental development and in preeclampsia. Cell Death Differ. 2010; 17: 846–59.CrossRefGoogle ScholarPubMed
Baczyk, D, Audette, MC, Coyaud, E, Raught, B, Kingdom, JC. Spatiotemporal distribution of small ubiquitin-like modifiers during human placental development and in response to oxidative and inflammatory stress. J Physiol. 2018; 596: 1587–600.Google Scholar
Drewlo, S, Levytska, K, Sobel, M, Baczyk, D, Lye, SJ, Kingdom, JC. Heparin promotes soluble VEGF receptor expression in human placental villi to impair endothelial VEGF signaling. J Thromb Haemost. 2011; 9: 2486–97.Google Scholar
Nevo, O, Soleymanlou, N, Wu, Y, Xu, J, Kingdom, J, Many, A, et al. Increased expression of sFlt-1 in in vivo and in vitro models of human placental hypoxia is mediated by HIF-1. Am J Physiol Regul Integr Comp Physiol. 2006; 291: R1085–93.Google Scholar
Tache, V, LaCoursiere, DY, Saleemuddin, A, Parast, MM. Placental expression of vascular endothelial growth factor receptor-1/soluble vascular endothelial growth factor receptor-1 correlates with severity of clinical preeclampsia and villous hypermaturity. Hum Pathol. 2011; 42: 1283–8.Google Scholar
Buhimschi, IA, Nayeri, UA, Zhao, G, Shook, LL, Pensalfini, A, Funai, EF, et al. Protein misfolding, congophilia, oligomerization, and defective amyloid processing in preeclampsia. Sci Transl Med. 2014; 6: 245ra92.Google Scholar
Zeisler, H, Llurba, E, Chantraine, F, Vatish, M, Staff, AC, Sennstrom, M, et al. Predictive value of the sFlt-1:PlGF ratio in women with suspected preeclampsia. N Engl J Med. 2016; 374: 1322.Google Scholar
Griffin, M, Seed, PT, Duckworth, S, North, R, Myers, J, Mackillop, L, et al. Predicting delivery of a small-for-gestational-age infant and adverse perinatal outcome in women with suspected pre-eclampsia. Ultrasound Obstet Gynecol. 2018; 51: 387–95.Google Scholar
Franco, C, Walker, M, Robertson, J, Fitzgerald, B, Keating, S, McLeod, A, et al. Placental infarction and thrombophilia. Obstet Gynecol. 2011; 117: 929–34.Google Scholar
Viero, S, Chaddha, V, Alkazaleh, F, Simchen, MJ, Malik, A, Kelly, E, et al. Prognostic value of placental ultrasound in pregnancies complicated by absent end-diastolic flow velocity in the umbilical arteries. Placenta. 2004; 25: 735–41.Google Scholar
Proctor, LK, Whittle, WL, Keating, S, Viero, S, Kingdom, JC. Pathologic basis of echogenic cystic lesions in the human placenta: role of ultrasound-guided wire localization. Placenta. 2010; 31: 1111–15.Google Scholar
Walker, M, Whittle, W, Keating, S, Kingdom, J. Sonographic diagnosis of chronic abruption. J Obstet Gynaecol Can. 2010; 32: 1056–8.Google Scholar
D’Souza, R, Keating, S, Walker, M, Drewlo, S, Kingdom, J. Unfractionated heparin and placental pathology in high-risk pregnancies: secondary analysis of a pilot randomized controlled trial. Placenta. 2014; 35: 816–23.Google Scholar
Rolnik, DL, Wright, D, Poon, LC, O’Gorman, N, Syngelaki, A, de Paco Matallana, C, et al. Aspirin versus placebo in pregnancies at high risk for preterm preeclampsia. N Engl J Med. 2017; 377: 613–22.Google Scholar
Figueras, F, Caradeux, J, Crispi, F, Eixarch, E, Peguero, A, Gratacos, E. Diagnosis and surveillance of late-onset fetal growth restriction. Am J Obstet Gynecol. 2018; 218: S790–802. e1.Google Scholar
Ting, JY, Kingdom, JC, Shah, PS. Antenatal glucocorticoids, magnesium sulfate, and mode of birth in preterm fetal small for gestational age. Am J Obstet Gynecol. 2018; 218: S818–28.Google Scholar
Nicolaides, KH. Screening for fetal aneuploidies at 11 to 13 weeks. Prenat Diagn. 2011; 31: 715.Google Scholar
Dugoff, L. First- and second-trimester maternal serum markers for aneuploidy and adverse obstetric outcomes. Obstet Gynecol. 2010; 115: 1052–61.Google Scholar
Smith, GC, Crossley, JA, Aitken, DA, Pell, JP, Cameron, AD, Connor, JM, et al. First-trimester placentation and the risk of antepartum stillbirth. JAMA. 2004; 292: 2249–54.Google Scholar
Smith, GC, Shah, I, Crossley, JA, Aitken, DA, Pell, JP, Nelson, SM, et al. Pregnancy-associated plasma protein A and alpha-fetoprotein and prediction of adverse perinatal outcome. Obstet Gynecol. 2006; 107: 161–6.Google Scholar
Crovetto, F, Triunfo, S, Crispi, F, Rodriguez-Sureda, V, Roma, E, Dominguez, C, et al. First-trimester screening with specific algorithms for early- and late-onset fetal growth restriction. Ultrasound Obstet Gynecol. 2016; 48: 340–8.Google Scholar
Rizzo, G, Capponi, A, Pietrolucci, ME, Capece, A, Arduini, D. First-trimester placental volume and vascularization measured by 3-dimensional power Doppler sonography in pregnancies with low serum pregnancy-associated plasma protein a levels. J Ultrasound Med. 2009; 28: 1615–22.Google Scholar
Yigiter, AB, Kavak, ZN, Durukan, B, Isci, H, Uzuner, A, Uyar, E, et al. Placental volume and vascularization flow indices by 3D power Doppler US using VOCAL technique and correlation with IGF-1, free beta-hCG, PAPP-A, and uterine artery Doppler at 11-14 weeks of pregnancy. J Perinat Med. 2011; 39: 137–41.Google Scholar
Alanjari, A, Wright, E, Keating, S, Ryan, G, Kingdom, J. Prenatal diagnosis, clinical outcomes, and associated pathology in pregnancies complicated by massive subchorionic thrombohematoma (Breus’ mole). Prenat Diagn. 2013; 33: 973–8.Google Scholar
Saleemuddin, A, Tantbirojn, P, Sirois, K, Crum, CP, Boyd, TK, Tworoger, S, et al. Obstetric and perinatal complications in placentas with fetal thrombotic vasculopathy. Pediatr Dev Pathol. 2010; 13: 459–64.Google Scholar
Salafia, CM, Pezzullo, JC, Minior, VK, Divon, MY. Placental pathology of absent and reversed end-diastolic flow in growth-restricted fetuses. Obstet Gynecol. 1997; 90: 830–6.Google Scholar
Redline, RW. Placental pathology: a systematic approach with clinical correlations. Placenta. 2008; 29 (Suppl. A): S86–91.Google Scholar
Cox, P, Marton, T. Pathological assessment of intrauterine growth restriction. Best Pract Res Clin Obstet Gynaecol. 2009; 23: 751–64.Google Scholar
Klaritsch, P, Haeusler, M, Karpf, E, Schlembach, D, Lang, U. Spontaneous intrauterine umbilical artery thrombosis leading to severe fetal growth restriction. Placenta. 2008; 29: 374–7.Google Scholar
Reister, F, Frank, HG, Kingdom, JC, Heyl, W, Kaufmann, P, Rath, W, et al. Macrophage-induced apoptosis limits endovascular trophoblast invasion in the uterine wall of preeclamptic women. Lab Invest. 2001; 81: 1143–52.Google Scholar
Contro, E, deSouza, R, Bhide, A. Chronic intervillositis of the placenta: a systematic review. Placenta. 2010; 31: 1106–10.CrossRefGoogle ScholarPubMed
Uxa, R, Baczyk, D, Kingdom, JC, Viero, S, Casper, R, Keating, S. Genetic polymorphisms in the fibrinolytic system of placentas with massive perivillous fibrin deposition. Placenta. 2010; 31: 499505.Google Scholar
Fitzgerald, B, Baczyk, D, J. K, Keating, S. Villous Cytotrophoblast Phenotype Switching in Massive Perivillous Fibrinoid Deposition. Submitted for publication. 2011.Google Scholar
Boog, G. Chronic villitis of unknown etiology. Eur J Obstet Gynecol Reprod Biol. 2008; 136: 915.Google Scholar
Katzman, PJ, Murphy, SP, Oble, DA. Immunohistochemical analysis reveals an influx of regulatory T cells and focal trophoblastic STAT-1 phosphorylation in chronic villitis of unknown etiology. Pediatr Dev Pathol. 2011; 14: 284–93.Google Scholar
Tang, Z, Abrahams, VM, Mor, G, Guller, S. Placental Hofbauer cells and complications of pregnancy. Ann N Y Acad Sci. 2011; 1221: 103–8.Google Scholar
Derricott, H, Jones, RL, Greenwood, SL, Batra, G, Evans, MJ, Heazell, AE. Characterizing villitis of unknown etiology and inflammation in stillbirth. Am J Pathol. 2016; 186: 952–61.Google Scholar
Levytska, K, Higgins, M, Keating, S, Melamed, N, Walker, M, Sebire, NJ, et al. Placental pathology in relation to uterine artery doppler findings in pregnancies with severe intrauterine growth restriction and abnormal umbilical artery doppler changes. Am J Perinatol. 2017; 34: 451–7.Google Scholar
Walker, MG, Hindmarsh, PC, Geary, M, Kingdom, JC. Sonographic maturation of the placenta at 30 to 34 weeks is not associated with second trimester markers of placental insufficiency in low-risk pregnancies. J Obstet Gynaecol Can. 2010; 32: 1134–9.Google Scholar
Cooley, SM, Donnelly, JC, Walsh, T, McMahon, C, Gillan, J, Geary, MP. The impact of ultrasonographic placental architecture on antenatal course, labor and delivery in a low-risk primigravid population. J Matern Fetal Neonatal Med. 2011; 24: 493–7.Google Scholar
Laskin, CA, Bombardier, C, Hannah, ME, Mandel, FP, Ritchie, JW, Farewell, V, et al. Prednisone and aspirin in women with autoantibodies and unexplained recurrent fetal loss. N Engl J Med. 1997; 337: 148–53.Google Scholar
Rodger, MA, Gris, JC, de Vries, JIP, Martinelli, I, Rey, E, Schleussner, E, et al. Low-molecular-weight heparin and recurrent placenta-mediated pregnancy complications: a meta-analysis of individual patient data from randomised controlled trials. Lancet. 2016; 388: 2629–41.Google Scholar
Wat, JM, Audette, M, Kingdom, JC. Molecular actions of heparin and their implications in preventing preeclampsia. J Thromb Haemost. 2018; 16 [Epub ahead of print]Google Scholar
Bewley, S, Cooper, D, Campbell, S. Doppler investigation of uteroplacental blood flow resistance in the second trimester: a screening study for pre-eclampsia and intrauterine growth retardation. BJOG. 1991; 98: 871–9.Google Scholar
Yu, CK, Smith, GC, Papageorghiou, AT, Cacho, AM, Nicolaides, KH. An integrated model for the prediction of preeclampsia using maternal factors and uterine artery Doppler velocimetry in unselected low-risk women. Am J Obstet Gynecol. 2005; 193: 429–36.Google Scholar
Pardi, G, Cetin, I, Marconi, AM, Bozzetti, P, Buscaglia, M, Makowski, EL, et al. Venous drainage of the human uterus: respiratory gas studies in normal and fetal growth-retarded pregnancies. Am J Obstet Gynecol. 1992; 166: 699706.Google Scholar
Alkazaleh, F, Chaddha, V, Viero, S, Malik, A, Anastasiades, C, Sroka, H, et al. Second-trimester prediction of severe placental complications in women with combined elevations in alpha-fetoprotein and human chorionic gonadotrophin. Am J Obstet Gynecol. 2006; 194: 821–7.Google Scholar
Huang, T, Hoffman, B, Meschino, W, Kingdom, J, Okun, N. Prediction of adverse pregnancy outcomes by combinations of first and second trimester biochemistry markers used in the routine prenatal screening of Down syndrome. Prenat Diagn. 2010; 30: 471–7.Google Scholar
Schwartz, N, Coletta, J, Pessel, C, Feng, R, Timor-Tritsch, IE, Parry, S, et al. Novel 3-dimensional placental measurements in early pregnancy as predictors of adverse pregnancy outcomes. J Ultrasound Med. 2010; 29: 1203–12.CrossRefGoogle ScholarPubMed
Kingdom, JC, Walker, M, Proctor, LK, Keating, S, Shah, PS, McLeod, A, et al. Unfractionated heparin for second trimester placental insufficiency: a pilot randomized trial. J Thromb Haemost. 2011; 9: 1483–92.Google Scholar
McLaughlin, K, Baczyk, D, Potts, A, Hladunewich, M, Parker, JD, Kingdom, JC. Low molecular weight heparin improves endothelial function in pregnant women at high risk of preeclampsia. Hypertension. 2017; 69: 180–8.Google Scholar
Myers, JE, Kenny, LC, McCowan, LM, Chan, EH, Dekker, GA, Poston, L, et al. Angiogenic factors combined with clinical risk factors to predict preterm pre-eclampsia in nulliparous women: a predictive test accuracy study. BJOG. 2013; 120: 1215–23.Google Scholar
McLaughlin, K, Scholten, RR, Parker, JD, Ferrazzi, E, Kingdom, JCP. Low molecular weight heparin for the prevention of severe preeclampsia: where next? Br J Clin Pharmacol. 2018; 84: 673–8.Google Scholar
Preston, FE, Rosendaal, FR, Walker, ID, Briet, E, Berntorp, E, Conard, J, et al. Increased fetal loss in women with heritable thrombophilia. Lancet. 1996; 348: 913–16.Google Scholar
Mousa, HA, Alfirevic, Z. Do placental lesions reflect thrombophilia state in women with adverse pregnancy outcome? Hum Reprod. 2000; 15: 1830–3.Google Scholar
Farine, D, Ryan, G, Kelly, EN, Morrow, RJ, Laskin, C, Ritchie, JW. Absent end-diastolic flow velocity waveforms in the umbilical artery—the subsequent pregnancy. Am J Obstet Gynecol. 1993; 168: 637–40.Google Scholar
Toal, M, Chan, C, Fallah, S, Alkazaleh, F, Chaddha, V, Windrim, RC, et al. Usefulness of a placental profile in high-risk pregnancies. Am J Obstet Gynecol. 2007; 196: 363. e1–7.Google Scholar
Staff, AC, Dechend, R, Pijnenborg, R. Learning from the placenta: acute atherosis and vascular remodeling in preeclampsia – novel aspects for atherosclerosis and future cardiovascular health. Hypertension. 2010; 56: 1026–34.Google Scholar

References

Royal College of Obstetricians and Gynaecologists. (2014). The Investigation and Management of the Small-for-Gestational-Age Fetus. Green-top Guideline No. 31. https://www.rcog.org.uk/globalassets/documents/guidelines/gtg_31.pdfGoogle Scholar
Flenady, V, Middleton, P, Smith, GC, Duke, W, Erwich, JJ, Khong, TY, et al. Stillbirths: the way forward in high-income countries. Lancet. 2011; 377: 1703–17.Google Scholar
Gardosi, J, Madurasinghe, V, Williams, M, Malik, A, Francis, A. Maternal and fetal risk factors for stillbirth: population based study. BMJ. 2013; 346: f108.Google Scholar
Barker, DJ, Osmond, C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet. 1986; 1: 1077–81.Google Scholar
Mifsud, W, Sebire, NJ. Placental pathology in early-onset and late-onset fetal growth restriction. Fetal Diagn Ther. 2014; 36: 117–28.Google Scholar
Gordijn, SJ, Beune, IM, Thilaganathan, B, Papageorghiou, A, Baschat, AA, Baker, PN, et al. Consensus definition of fetal growth restriction: a Delphi procedure. Ultrasound Obstet Gynecol. 2016; 48: 333–9.Google Scholar
Lean, SC, Derricott, H, Jones, RL, Heazell, AEP. Advanced maternal age and adverse pregnancy outcomes: a systematic review and meta-analysis. PLoS ONE. 2017; 12: e0186287.Google Scholar
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: 1331–40.Google Scholar
Schieve, LA, Meikle, SF, Ferre, C, Peterson, HB, Jeng, G, Wilcox, LS. Low and very low birth weight in infants conceived with use of assisted reproductive technology. N Engl J Med. 2002; 346: 731–7.Google Scholar
Lawn, JE, Blencowe, H, Waiswa, P, Amouzou, A, Mathers, C, Hogan, D, et al. Stillbirths: rates, risk factors, and acceleration towards 2030. Lancet. 2016; 387: 587603.Google Scholar
McDonald, SD, Han, Z, Mulla, S, Murphy, KE, Beyene, J, Ohlsson, A. Preterm birth and low birth weight among in vitro fertilization singletons: a systematic review and meta-analyses. Eur J Obstet Gynecol Reprod Biol. 2009; 146: 138–48.Google Scholar
Helmerhorst, FM, Perquin, DA, Donker, D, Keirse, MJ. Perinatal outcome of singletons and twins after assisted conception: a systematic review of controlled studies. BMJ. 2004; 328: 261.Google Scholar
Steegers, EA, von Dadelszen, P, Duvekot, JJ, Pijnenborg, R. Pre-eclampsia. Lancet. 2010; 376: 631–44.Google Scholar
Schreiber, K, Radin, M, Sciascia, S. Current insights in obstetric antiphospholipid syndrome. Curr Opin Obstet Gynecol. 2017; 29: 397403.Google Scholar
Sammaritano, LR. Management of systemic lupus erythematosus during pregnancy. Annu Rev Med. 2017; 68: 271–85.Google Scholar
Foo, FL, Mahendru, AA, Masini, G, Fraser, A, Cacciatore, S, MacIntyre, DA, et al. Association between prepregnancy cardiovascular function and subsequent preeclampsia or fetal growth restriction. Hypertension. 2018; 72: 442–50.Google Scholar
Tay, J, Foo, L, Masini, G, Bennett, PR, McEniery, CM, Wilkinson, IB, et al. Early and late preeclampsia are characterized by high cardiac output, but in the presence of fetal growth restriction, cardiac output is low: insights from a prospective study. Am J Obstet Gynecol. 2018; 218: 517. e1–e12.Google Scholar
Simcox, LE, Ormesher, L, Tower, C, Greer, IA. Thrombophilia and pregnancy complications. Int J Mol Sci. 2015; 16: 28418–28.Google Scholar
Rodger, MA, Gris, JC, de Vries, JIP, Martinelli, I, Rey, E, Schleussner, E, et al. Low-molecular-weight heparin and recurrent placenta-mediated pregnancy complications: a meta-analysis of individual patient data from randomised controlled trials. Lancet. 2016; 388: 2629–41.Google Scholar
Kingdom, JC, Drewlo, S. Is heparin a placental anticoagulant in high-risk pregnancies? Blood. 2011; 118: 4780–8.Google Scholar
Bundhun, PK, Soogund, MZ, Huang, F. Impact of systemic lupus erythematosus on maternal and fetal outcomes following pregnancy: a meta-analysis of studies published between years 2001-2016. J Autoimmun. 2017; 79: 1727.Google Scholar
Chakravarty, EF, Khanna, D, Chung, L. Pregnancy outcomes in systemic sclerosis, primary pulmonary hypertension, and sickle cell disease. Obstet Gynecol. 2008; 111: 927–34.Google Scholar
Greutmann, M, Pieper, PG. Pregnancy in women with congenital heart disease. Eur Heart J. 2015; 36: 2491–9.Google Scholar
Luewan, S, Srisupundit, K, Tongsong, T. Outcomes of pregnancies complicated by beta-thalassemia/hemoglobin E disease. Int J Gynaecol Obstet. 2009; 104: 203–5.Google Scholar
Mortola, JP, Frappell, PB, Aguero, L, Armstrong, K. Birth weight and altitude: a study in Peruvian communities. J Pediatr. 2000; 136: 324–9.Google Scholar
Stein, AD, Lumey, LH. The relationship between maternal and offspring birth weights after maternal prenatal famine exposure: the Dutch Famine Birth Cohort Study. Hum Biol. 2000; 72: 641–54.Google Scholar
Saccone, G, Berghella, V, Sarno, L, Maruotti, GM, Cetin, I, Greco, L, et al. Celiac disease and obstetric complications: a systematic review and metaanalysis. Am J Obstet Gynecol. 2016; 214: 225–34.Google Scholar
Khashan, AS, Henriksen, TB, Mortensen, PB, McNamee, R, McCarthy, FP, Pedersen, MG, et al. The impact of maternal celiac disease on birthweight and preterm birth: a Danish population-based cohort study. Hum Reprod. 2010; 25: 528–34.Google Scholar
Anjum, N, Baker, PN, Robinson, NJ, Aplin, JD. Maternal celiac disease autoantibodies bind directly to syncytiotrophoblast and inhibit placental tissue transglutaminase activity. Reprod Biol Endocrinol. 2009; 7: 16.Google Scholar
Kiefte-de Jong, JC, Jaddoe, VW, Uitterlinden, AG, Steegers, EA, Willemsen, SP, Hofman, A, et al. Levels of antibodies against tissue transglutaminase during pregnancy are associated with reduced fetal weight and birth weight. Gastroenterology. 2013; 144: 726–35. e2.Google Scholar
Suter, MA, Anders, AM, Aagaard, KM. Maternal smoking as a model for environmental epigenetic changes affecting birthweight and fetal programming. Mol Hum Reprod. 2013; 19: 16.Google Scholar
Blatt, K, Moore, E, Chen, A, Van Hook, J, DeFranco, EA. Association of reported trimester-specific smoking cessation with fetal growth restriction. Obstet Gynecol. 2015; 125: 1452–9.Google Scholar
Carter, RC, Jacobson, JL, Molteno, CD, Dodge, NC, Meintjes, EM, Jacobson, SW. Fetal alcohol growth restriction and cognitive impairment. Pediatrics. 2016; 138: e20160775.Google Scholar
Carter, RC, Wainwright, H, Molteno, CD, Georgieff, MK, Dodge, NC, Warton, F, et al. Alcohol, methamphetamine, and marijuana exposure have distinct effects on the human placenta. Alcohol Clin Exp Res. 2016; 40: 753–64.Google Scholar
Gundogan, F, Elwood, G, Longato, L, Tong, M, Feijoo, A, Carlson, RI, et al. Impaired placentation in fetal alcohol syndrome. Placenta. 2008; 29: 148–57.Google Scholar
Committee on Obstetric Practice. Committee Opinion No. 711: Opioid Use and Opioid Use Disorder in Pregnancy. Obstet Gynecol. 2017; 130: e81–94.Google Scholar
Zedler, BK, Mann, AL, Kim, MM, Amick, HR, Joyce, AR, Murrelle, EL, et al. Buprenorphine compared with methadone to treat pregnant women with opioid use disorder: a systematic review and meta-analysis of safety in the mother, fetus and child. Addiction. 2016; 111: 2115–28.Google Scholar
Soto, E, Bahado-Singh, R. Fetal abnormal growth associated with substance abuse. Clin Obstet Gynecol. 2013; 56: 142–53.Google Scholar
Bada, HS, Das, A, Bauer, CR, Shankaran, S, Lester, B, Wright, LL, et al. Gestational cocaine exposure and intrauterine growth: maternal lifestyle study. Obstet Gynecol. 2002; 100: 916–24.Google Scholar
Kalaitzopoulos, DR, Chatzistergiou, K, Amylidi, AL, Kokkinidis, DG, Goulis, DG. Effect of methamphetamine hydrochloride on pregnancy outcome: a systematic review and meta-analysis. J Addict Med. 2018; 12: 220–6.Google Scholar
Ray, S, Stowe, ZN. The use of antidepressant medication in pregnancy. Best Pract Res Clin Obstet Gynaecol. 2014; 28: 7183.Google Scholar
Bloise, E, Feuer, SK, Rinaudo, PF. Comparative intrauterine development and placental function of ART concepti: implications for human reproductive medicine and animal breeding. Hum Reprod Update. 2014; 20: 822–39.Google Scholar
Snijders, RJ, Sherrod, C, Gosden, CM, Nicolaides, KH. Fetal growth retardation: associated malformations and chromosomal abnormalities. Am J Obstet Gynecol. 1993; 168: 547–55.Google Scholar
Rochelson, B, Kaplan, C, Guzman, E, Arato, M, Hansen, K, Trunca, C. A quantitative analysis of placental vasculature in the third-trimester fetus with autosomal trisomy. Obstet Gynecol. 1990; 75: 5963.Google Scholar
Fujimoto, A, Wilson, MG. Growth retardation in Wolf-Hirschhorn syndrome. Hum Genet. 1990; 84: 296–7.Google Scholar
Sagi-Dain, L, Peleg, A, Sagi, S. Risk for chromosomal aberrations in apparently isolated intrauterine growth restriction: A systematic review. Prenat Diagn. 2017; 37: 1061–6.Google Scholar
Khoury, MJ, Erickson, JD, Cordero, JF, McCarthy, BJ. Congenital malformations and intrauterine growth retardation: a population study. Pediatrics. 1988; 82: 8390.Google Scholar
Sun, L, Macgowan, CK, Sled, JG, Yoo, SJ, Manlhiot, C, Porayette, P, et al. Reduced fetal cerebral oxygen consumption is associated with smaller brain size in fetuses with congenital heart disease. Circulation. 2015; 131: 1313–23.Google Scholar
Naeye, RL. Unsuspected organ abnormalities associated with congenital heart disease. Am J Pathol. 1965; 47: 905–15.Google Scholar
Hamilton, ST, Scott, G, Naing, Z, Iwasenko, J, Hall, B, Graf, N, et al. Human cytomegalovirus-induces cytokine changes in the placenta with implications for adverse pregnancy outcomes. PLoS ONE. 2012; 7: e52899.Google Scholar
Adams Waldorf, KM, McAdams, RM. Influence of infection during pregnancy on fetal development. Reproduction. 2013; 146: R151–62.Google Scholar
Umbers, AJ, Aitken, EH, Rogerson, SJ. Malaria in pregnancy: small babies, big problem. Trends Parasitol. 2011; 27: 168–75.Google Scholar
McCarthy, FP, Giles, ML, Rowlands, S, Purcell, KJ, Jones, CA. Antenatal interventions for preventing the transmission of cytomegalovirus (CMV) from the mother to fetus during pregnancy and adverse outcomes in the congenitally infected infant. Cochrane Database Syst Rev. 2011; 3: CD008371.Google Scholar
Sassoon, DA, Castro, LC, Davis, JL, Hobel, CJ. Perinatal outcome in triplet versus twin gestations. Obstet Gynecol. 1990; 75: 817–20.Google Scholar
Coutinho Nunes, F, Domingues, AP, Vide Tavares, M, Belo, A, Ferreira, C, Fonseca, E, et al. Monochorionic versus dichorionic twins: are obstetric outcomes always different? J Obstet Gynaecol. 2016; 36: 598601.Google Scholar
Stirrup, OT, Khalil, A, D’Antonio, F, Thilaganathan, B, Southwest Thames Obstetric Research Collaborative. Fetal growth reference ranges in twin pregnancy: analysis of the Southwest Thames Obstetric Research Collaborative (STORK) multiple pregnancy cohort. Ultrasound Obstet Gynecol. 2015; 45: 301–7.Google Scholar
Cheong-See, F, Schuit, E, Arroyo-Manzano, D, Khalil, A, Barrett, J, Joseph, KS, et al. Prospective risk of stillbirth and neonatal complications in twin pregnancies: systematic review and meta-analysis. BMJ. 2016; 354: i4353.Google Scholar
Hall, JG. Twinning. Lancet. 2003; 362: 735–43.Google Scholar
Sparks, TN, Nakagawa, S, Gonzalez, JM. Hypertension in dichorionic twin gestations: how is birthweight affected? J Matern Fetal Neonatal Med. 2017; 30: 380–5.Google Scholar
Lewi, L, Cannie, M, Blickstein, I, Jani, J, Huber, A, Hecher, K, et al. Placental sharing, birthweight discordance, and vascular anastomoses in monochorionic diamniotic twin placentas. Am J Obstet Gynecol. 2007; 197: 587. e1–8.Google Scholar
Audette, MC, Kingdom, JC. Screening for fetal growth restriction and placental insufficiency. Semin Fetal Neonatal Med. 2018; 23: 119–25.Google Scholar
Kingdom, JC, Audette, MC, Hobson, SR, Windrim, RC, Morgen, E. A placenta clinic approach to the diagnosis and management of fetal growth restriction. Am J Obstet Gynecol. 2018; 218: S803–17.Google Scholar
Pay, AS, Wiik, J, Backe, B, Jacobsson, B, Strandell, A, Klovning, A. Symphysis-fundus height measurement to predict small-for-gestational-age status at birth: a systematic review. BMC Pregnancy Childbirth. 2015; 15: 22.Google Scholar
Robert Peter, J, Ho, JJ, Valliapan, J, Sivasangari, S. Symphysial fundal height (SFH) measurement in pregnancy for detecting abnormal fetal growth. Cochrane Database Syst Rev. 2015; 9: CD008136.Google Scholar
Hadlock, FP, Harrist, RB, Sharman, RS, Deter, RL, Park, SK. Estimation of fetal weight with the use of head, body, and femur measurements—a prospective study. Am J Obstet Gynecol. 1985; 151: 333–7.Google Scholar
Papageorghiou, AT, Ohuma, EO, Altman, DG, Todros, T, Cheikh Ismail, L, Lambert, A, et al. International standards for fetal growth based on serial ultrasound measurements: the Fetal Growth Longitudinal Study of the INTERGROWTH-21st Project. Lancet. 2014; 384: 869–79.Google Scholar
Gardosi, J. Customized fetal growth standards: rationale and clinical application. Semin Perinatol. 2004; 28: 3340.Google Scholar
Ego, A, Subtil, D, Grange, G, Thiebaugeorges, O, Senat, MV, Vayssiere, C, et al. Customized versus population-based birth weight standards for identifying growth restricted infants: a French multicenter study. Am J Obstet Gynecol. 2006; 194: 1042–9.Google Scholar
Reddy, M, Wallace, EM, Mockler, JC, Stewart, L, Knight, M, Hodges, R, et al. Maternal Asian ethnicity and obstetric intrapartum intervention: a retrospective cohort study. BMC Pregnancy Childbirth. 2017; 17: 3.Google Scholar
Lockie, E, McCarthy, EA, Hui, L, Churilov, L, Walker, SP. Feasibility of using self-reported ethnicity in pregnancy according to the gestation-related optimal weight classification: a cross-sectional study. BJOG. 2018; 125: 704–9.Google Scholar
Kiserud, T, Piaggio, G, Carroli, G, Widmer, M, Carvalho, J, Neerup Jensen, L, et al. The World Health Organization Fetal Growth Charts: A Multinational Longitudinal Study of Ultrasound Biometric Measurements and Estimated Fetal Weight. PLoS Medicine. 2017; 14: e1002220.Google Scholar
Bricker, L, Medley, N, Pratt, JJ. Routine ultrasound in late pregnancy (after 24 weeks’ gestation). Cochrane Database Syst Rev. 2015; 6: CD001451.Google Scholar
Sovio, U, White, IR, Dacey, A, Pasupathy, D, Smith, GCS. Screening for fetal growth restriction with universal third trimester ultrasonography in nulliparous women in the Pregnancy Outcome Prediction (POP) study: a prospective cohort study. Lancet. 2015; 386: 2089–97.Google Scholar
Monier, I, Blondel, B, Ego, A, Kaminiski, M, Goffinet, F, Zeitlin, J. Poor effectiveness of antenatal detection of fetal growth restriction and consequences for obstetric management and neonatal outcomes: a French national study. BJOG. 2015; 122: 518–27.Google Scholar
Deter, RL. Individualized growth assessment: evaluation of growth using each fetus as its own control. Semin Perinatol. 2004; 28: 2332.Google Scholar
Kingdom, JC, Burrell, SJ, Kaufmann, P. Pathology and clinical implications of abnormal umbilical artery Doppler waveforms. Ultrasound Obstet Gynecol. 1997; 9: 271–86.Google Scholar
Alfirevic, Z, Stampalija, T, Dowswell, T. Fetal and umbilical Doppler ultrasound in high-risk pregnancies. Cochrane Database Syst Rev. 2017; 6: CD007529.Google Scholar
Alfirevic, Z, Stampalija, T, Medley, N. Fetal and umbilical Doppler ultrasound in normal pregnancy. Cochrane Database Syst Rev. 2015; 4: CD001450.Google Scholar
Rodriguez, A, Tuuli, MG, Odibo, AO. First-, second-, and third-trimester screening for preeclampsia and intrauterine growth restriction. Clin Lab Med. 2016; 36: 331–51.Google Scholar
Roberts, LA, Ling, HZ, Poon, L, Nicolaides, KH, Kametas, NA. Maternal hemodynamics, fetal biometry and Dopplers in pregnancies followed up for suspected fetal growth restriction. Ultrasound Obstet Gynecol. 2018; 52: 507–14.Google Scholar
Smith, GC. First-trimester determination of complications of late pregnancy. JAMA. 2010; 303: 561–2.Google Scholar
Gaccioli, F, Aye, I, Sovio, U, Charnock-Jones, DS, Smith, GCS. Screening for fetal growth restriction using fetal biometry combined with maternal biomarkers. Am J Obstet Gynecol. 2018; 218: S725–37.Google Scholar
Tan, MY, Poon, LC, Rolnik, DL, Syngelaki, A, de Paco Matallana, C, Akolekar, R, et al. Prediction and prevention of small-for-gestational-age neonates: evidence from SPREE and ASPRE. Ultrasound Obstet Gynecol. 2018; 52: 52–9.Google Scholar
Sovio, U, Smith, GCS. The effect of customization and use of a fetal growth standard on the association between birthweight percentile and adverse perinatal outcome. Am J Obstet Gynecol. 2018; 218: S738–44.Google Scholar
Griffin, M, Seed, PT, Duckworth, S, North, R, Myers, J, Mackillop, L, et al. Predicting delivery of a small-for-gestational-age infant and adverse perinatal outcome in women with suspected pre-eclampsia. Ultrasound Obstet Gynecol. 2018; 51: 387–95.Google Scholar
Sharp, A, Chappell, LC, Dekker, G, Pelletier, S, Garnier, Y, Zeren, O, et al. Placental Growth Factor informed management of suspected pre-eclampsia or fetal growth restriction: the MAPPLE cohort study. Pregnancy Hypertens. 2018; 14: 228–33.Google Scholar
Ormesher, L, Johnstone, ED, Shawkat, E, Dempsey, A, Chmiel, C, Ingram, E, et al. A clinical evaluation of placental growth factor in routine practice in high-risk women presenting with suspected pre-eclampsia and/or fetal growth restriction. Pregnancy Hypertens. 2018; 14: 243–9.Google Scholar
Griffin, M, Seed, PT, Webster, L, Myers, J, MacKillop, L, Simpson, N, et al. Diagnostic accuracy of placental growth factor and ultrasound parameters to predict the small-for-gestational-age infant in women presenting with reduced symphysis-fundus height. Ultrasound Obstet Gynecol. 2015; 46: 182–90.Google Scholar
Lees, C, Marlow, N, Arabin, B, Bilardo, CM, Brezinka, C, Derks, JB, et al. Perinatal morbidity and mortality in early-onset fetal growth restriction: cohort outcomes of the trial of randomized umbilical and fetal flow in Europe (TRUFFLE). Ultrasound Obstet Gynecol. 2013; 42: 400–8.Google Scholar
Figueras, F, Caradeux, J, Crispi, F, Eixarch, E, Peguero, A, Gratacós, E. Diagnosis and surveillance of late-onset fetal growth restriction. Am J Obstet Gynecol. 2018; 218: S790–802. e1.Google Scholar
Figueras, F, Gratacós, E. An integrated approach to fetal growth restriction. Best Pract Res Clin Obstet Gynaecol. 2017; 38: 4858.Google Scholar
Sebire, NJ. Umbilical artery Doppler revisited: pathophysiology of changes in intrauterine growth restriction revealed. Ultrasound Obstet Gynecol. 2003; 21: 419–22.Google Scholar
Caradeux, J, Martinez-Portilla, RJ, Basuki, TR, Kiserud, T, Figueras, F. Risk of fetal death in growth-restricted fetuses with umbilical and/or ductus venosus absent or reversed end-diastolic velocities before 34 weeks of gestation: a systematic review and meta-analysis. Am J Obstet Gynecol. 2018; 218: S774–82. e21.Google Scholar
Vollgraff Heidweiller-Schreurs, CA, De Boer, MA, Heymans, MW, Schoonmade, LJ, Bossuyt, PMM, Mol, BWJ, et al. Prognostic accuracy of cerebroplacental ratio and middle cerebral artery Doppler for adverse perinatal outcome: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2018; 51: 313–22.Google Scholar
Grivell, RM, Alfirevic, Z, Gyte, GM, Devane, D. Antenatal cardiotocography for fetal assessment. Cochrane Database Syst Rev. 2015; 9: CD007863.Google Scholar
Pattison, N, McCowan, L. Cardiotocography for antepartum fetal assessment. Cochrane Database Syst Rev. 2000; 2: CD001068.Google Scholar
Frusca, T, Todros, T, Lees, C, Bilardo, CM, TRUFFLE Investigators. Outcome in early-onset fetal growth restriction is best combining computerized fetal heart rate analysis with ductus venosus Doppler: insights from the Trial of Umbilical and Fetal Flow in Europe. Am J Obstet Gynecol. 2018; 218: S783–9.Google Scholar
Lalor, JG, Fawole, B, Alfirevic, Z, Devane, D. Biophysical profile for fetal assessment in high risk pregnancies. Cochrane Database Syst Rev. 2008; 1: CD000038.Google Scholar
Kaur, S, Picconi, JL, Chadha, R, Kruger, M, Mari, G. Biophysical profile in the treatment of intrauterine growth-restricted fetuses who weigh <1000 g. Am J Obstet Gynecol. 2008; 199: 264. e1–4.Google Scholar
Thornton, JG, Hornbuckle, J, Vail, A, Spiegelhalter, DJ, Levene, M, GRIT Study Group. Infant wellbeing at 2 years of age in the Growth Restriction Intervention Trial (GRIT): multicentred randomised controlled trial. Lancet. 2004; 364: 513–20.Google Scholar
Walker, DM, Marlow, N, Upstone, L, Gross, H, Hornbuckle, J, Vail, A, et al. The Growth Restriction Intervention Trial: long-term outcomes in a randomized trial of timing of delivery in fetal growth restriction. Am J Obstet Gynecol. 2011; 204: 34. e1–9.Google Scholar
Lees, CC, Marlow, N, van Wassenaer-Leemhuis, A, Arabin, B, Bilardo, CM, Brezinka, C, et al. 2 year neurodevelopmental and intermediate perinatal outcomes in infants with very preterm fetal growth restriction (TRUFFLE): a randomised trial. Lancet. 2015; 385: 2162–72.Google Scholar
Ting, JY, Kingdom, JC, Shah, PS. Antenatal glucocorticoids, magnesium sulfate, and mode of birth in preterm fetal small for gestational age. Am J Obstet Gynecol. 2018; 218: S818–28.Google Scholar
Lee, HC, Gould, JB. Survival rates and mode of delivery for vertex preterm neonates according to small- or appropriate-for-gestational-age status. Pediatrics. 2006; 118: e1836–44.Google Scholar
Boers, KE, Vijgen, SM, Bijlenga, D, van der Post, JA, Bekedam, DJ, Kwee, A, et al. Induction versus expectant monitoring for intrauterine growth restriction at term: randomised equivalence trial (DIGITAT). BMJ. 2010; 341: c7087.Google Scholar
van Wyk, L, Boers, KE, van der Post, JA, van Pampus, MG, van Wassenaer, AG, van Baar, AL, et al. Effects on (neuro)developmental and behavioral outcome at 2 years of age of induced labor compared with expectant management in intrauterine growth-restricted infants: long-term outcomes of the DIGITAT trial. Am J Obstet Gynecol. 2012; 206: 406. e1–7.Google Scholar
Meher, S, Duley, L, Hunter, K, Askie, L. Antiplatelet therapy before or after 16 weeks’ gestation for preventing preeclampsia: an individual participant data meta-analysis. Am J Obstet Gynecol. 2017; 216: 121–8. e2.Google Scholar
Groom, KM, David, AL. The role of aspirin, heparin, and other interventions in the prevention and treatment of fetal growth restriction. Am J Obstet Gynecol. 2018; 218: S829–40.Google Scholar
Rolnik, DL, Wright, D, Poon, LC, O’Gorman, N, Syngelaki, A, de Paco Matallana, C, et al. Aspirin versus placebo in pregnancies at high risk for preterm preeclampsia. New Engl J Med. 2017; 377: 613–22.Google Scholar
Wat, JM, Audette, MC, Kingdom, JC. Molecular actions of heparin and their implications in preventing pre-eclampsia. J Thromb Haemost. 2018 [ePub ahead of print].Google Scholar
National Health and Medical Research Council (Australia). (2010). Antenatal magnesium sulphate prior to preterm birth for neuroprotection of the fetus, infant and child. National Clinical Practice Guidelines. https://www.clinicalguidelines.gov.au/register/antenatal-magnesium-sulphate-prior-preterm-birth-neuroprotection-fetus-infant-and-childGoogle Scholar
Stockley, EL, Ting, JY, Kingdom, JC, McDonald, SD, Barrett, JF, Synnes, AR, et al. Intrapartum magnesium sulfate is associated with neuroprotection in growth-restricted fetuses. Am J Obstet Gynecol. 2018; 219: 606. e1–e8.Google Scholar
Nawathe, A, David, AL. Prophylaxis and treatment of foetal growth restriction. Best Pract Res Clin Obstet Gynaecol. 2018; 49: 6678.Google Scholar
Wright, E, Audette, MC, Ye, XY, Keating, S, Hoffman, B, Lye, SJ, et al. Maternal vascular malperfusion and adverse perinatal outcomes in low-risk nulliparous women. Obstet Gynecol. 2017; 130: 1112–20.Google Scholar

References

Resnik, R. Intrauterine growth restriction. Obstet Gynecol. 2002; 99: 490–6.Google Scholar
Levine, TA, Grunau, RE, McAuliffe, FM, Pinnamaneni, R, Foran, A, Alderdice, FA. Early childhood neurodevelopment after intrauterine growth restriction: a systematic review. Pediatrics. 2015; 135: 126–41.Google Scholar
Barker, DJP. Adult consequences of fetal growth restriction. Clin Obstet Gynecol. 2006; 49: 270–83.Google Scholar
Gaillard, R, Steegers, EAP, de Jongste, JC, Hofman, A, Jaddoe, VWV. Tracking of fetal growth characteristics during different trimesters and the risks of adverse birth outcomes. Int J Epidemiol. 2014; 43: 1140–53.Google Scholar
Gardosi, J, Kady, SM, McGeown, P, Francis, A, Tonks, A. Classification of stillbirth by relevant condition at death (ReCoDe): population based cohort study. BMJ. 2005; 331: 1113.Google Scholar
Partap, U, Sovio, U, Smith, GC. Fetal growth and the risk of spontaneous preterm birth in a prospective cohort study of nulliparous women. Am J Epidemiol. 2016; 184: 110–19.Google Scholar
Lees, C, Marlow, N, Arabin, B, Bilardo, CM, Brezinka, C, et al. Perinatal morbidity and mortality in early-onset fetal growth restriction: cohort outcomes of the Trial of Randomized Umbilical and Fetal Flow in Europe (TRUFFLE). Ultrasound Obstet Gynecol. 2013; 42: 400–8.Google Scholar
Walker, DM, Marlow, N. Neurocognitive outcome following fetal growth restriction. Arch Dis Child Fetal Neonatal Ed. 2008; 93: F322–5.Google Scholar
Barker, DJP, Godfrey, KM, Gluckman, PD, Harding, JE, Owens, JA, Robinson, JS. Fetal nutrition and cardiovascular disease in adult life. Lancet. 1993; 341: 938–41.Google Scholar
Smith, GC, Wood, AM, White, IR, Pell, JP, Hattie, J. Birth weight and the risk of cardiovascular disease in the maternal grandparents. Am J Epidemiol. 2010; 171: 736–44.Google Scholar
Smith, GCS, Smith, MFS, McNay, MB, Fleming, JEE. First-trimester growth and the risk of low birth weight. N Engl J Med. 1998; 339: 1817–22.Google Scholar
Hadlock, FP, Harrist, RB, Sharman, RS, Deter, RL, Park, SK. Estimation of fetal weight with the use of head, body and femur measurements: A prospective study. Am J Obstet Gynecol. 1985; 151: 333–7.Google Scholar
Royal College of Obstetricians and Gynaecologists. (2014). The Investigation and Management of the Small-for-Gestational-Age Fetus. Green-top Guideline No. 31. www.rcog.org.uk/globalassets/documents/guidelines/gtg_31.pdfGoogle Scholar
Sovio, U, White, IR, Dacey, A, Pasuparthy, D, Smith, GCS. Screening for fetal growth restriction with universal third trimester ultrasonography in nulliparous women in the Pregnancy Outcome Prediction (POP) study: a prospective cohort study. Lancet. 2015; 386: 2089–97.Google Scholar
Clausson, B, Gardosi, J, Francis, A, Cnattingius, S. Perinatal outcome in SGA births defined by customised versus population-based birthweight standards. BJOG. 2001; 108 : 830–4.Google Scholar
Carberry, AE, Gordon, A, Bond, DM, Hyett, J, Raynes-Greenow, CH, Jeffery, HE. Customised versus population-based growth charts as a screening tool for detecting small for gestational age infants in low-risk pregnant women. Cochrane Database Syst Rev. 2014; 5: CD008549.Google Scholar
Iliodromiti, S, Mackay, DF, Smith, GCS, Pell, JP, Sattar, N, Lawlor, DA, et al. Customised and noncustomised birth weight centiles and prediction of stillbirth and infant mortality and morbidity: a cohort study of 979,912 term singleton pregnancies in Scotland. PLoS Med. 2017; 14: e1002228Google Scholar
Martin, AM, Bindra, R, Curcio, P, Cicero, S, Nicolaides, KH. Screening for pre-eclampsia and fetal growth restriction by uterine artery Doppler at 11-14 weeks of gestation. Ultrasound Obstet Gynecol. 2001; 18: 583–6.Google Scholar
Smith, GC, Yu, CK, Papageorghiou, AT, Cacho, AM, Nicolaides, KH, Fetal Medicine Foundation Second Trimester Screening Group. Maternal uterine Doppler flow velocimetry and the risk of stillbirth. Obstet Gynecol. 2007; 109: 144–51.Google Scholar
Figueras, F, Gratacós, E. Update on the diagnosis and classification of fetal growth restriction and proposal of a stage-based management protocol. Fetal Diagn Ther. 2014; 36: 8698.Google Scholar
Eixarch, E, Meler, E, Iraola, A, Illa, M, Crispi, F, Hernandez-Andrade, E, et al. Neurodevelopmental outcome in 2-year-old infants who were small-for-gestational age term fetuses with cerebral blood flow redistribution. Ultrasound Obstet Gynecol. 2008; 32: 894–9.Google Scholar
Khalil, A, Morales-Roselló, J, Townsend, R, Morlando, M, Papageorghiou, A, Bhide, A, et al. Value of third-trimester cerebroplacental ratio and uterine artery Doppler indices as predictors of stillbirth and perinatal loss. Ultrasound Obstet Gynecol. 2016; 47: 7480.Google Scholar
Vollgraff Heidweiller-Schreurs, CA, De Boer, MA, Heymans, MW, Schoonmade, LJ, Bossuyt, PMM, Mol, BWJ, et al. Prognostic accuracy of cerebroplacental ratio and middle cerebral artery Doppler for adverse perinatal outcome: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2017; 51: 313–22.Google Scholar
Morris, RK, Selman, TJ, Verma, M, Robson, SC, Kleijnen, J, Khan, KS. Systematic review and meta-analysis of the test accuracy of ductus venosus Doppler to predict compromise of fetal/neonatal wellbeing in high risk pregnancies with placental insufficiency. Eur J Obstet Gynecol Reprod Biol. 2010; 152: 312.Google Scholar
Lees, CC, Marlow, N, vas Wassenaer-Leemhuis, A, Arabin, B, Bilardo, CM, Brezinka, C, et al. 2 year neurodevelopmental and intermediate perinatal outcomes in infants with very preterm fetal growth restriction (TRUFFLE): a randomised trial. Lancet. 2015; 385: 2162–72.Google Scholar
Gaccioli, F, Aye, ILMH, Sovio, U, Charnock-Jones, DS, Smith, GCS. Screening for fetal growth restriction using fetal biometry combined with maternal biomarkers. Am J Obstet Gynecol. 2018; 218: S725–37.Google Scholar
Smith, GC, Stenhouse, EJ, Crossley, JA, Aitken, DA, Cameron, AD, Connor, JM. Early pregnancy levels of pregnancy-associated plasma protein A and the risk of intrauterine growth restriction, premature birth, preeclampsia and stillbirth. J Clin Endocrinol Metab. 2002; 87: 1762–7.Google Scholar
Dugoff, L, Hobbins, JC, Malone, FD, Vidaver, J, Sullivan, L, Canick, JA, et al. Quad screen as a predictor of adverse pregnancy outcome. Obstet Gynecol. 2005; 106: 260–7.Google Scholar
McCowan, LME, Thompson, JMD, Taylor, RS, Baker, PN, North, RA, Poston, L, et al. Prediction of small for gestational age infants in healthy nulliparous women using clinical and ultrasound risk factors combined with early pregnancy biomarkers. PLoS One. 2017; 12: e0169311.Google Scholar
Smith, GC, Shah, I, Crossley, JA, Aitken, DA, Pell, JP, Nelson, SM, et al. Pregnancy-associated plasma protein A and alpha-fetoprotein and prediction of adverse perinatal outcome. Obstet Gynecol. 2006; 107: 161–6.Google Scholar
Lesmes, C, Gallo, DM, Gonzalez, R, Poon, LC, Nicolaides, KH. Prediction of small-for-gestational-age neonates: screening by maternal serum biochemical markers at 19–24 weeks. Ultrasound Obstet Gynecol. 2015; 46: 341–9.Google Scholar
Karagiannis, G, Akolekar, R, Sarquis, R, Wright, D, Nicolaides, KH. Prediction of small-for-gestation neonates from biophysical and biochemical markers at 11–13 weeks. Fetal Diagn Ther. 2011; 29: 148–54.Google Scholar
Valiño, N, Giunta, G, Gallo, DM, Akolekar, R, Nicolaides, KH. Biophysical and biochemical markers at 30–34 weeks’ gestation in the prediction of adverse perinatal outcome. Ultrasound Obstet Gynecol. 2016; 47: 194202.Google Scholar
Valiño, N, Giunta, G, Gallo, DM, Akolekar, R, Nicolaides, KH. Biophysical and biochemical markers at 35–37 weeks’ gestation in the prediction of adverse perinatal outcome. Ultrasound Obstet Gynecol. 2016; 47: 203–9.Google Scholar
Cleaton, MAM, Dent, CL, Howard, M, Corish, JA, Gutteridge, I, Sovio, U, et al. Fetus-derived DLK1 is required for maternal metabolic adaptations to pregnancy and is associated with fetal growth restriction. Nat Genet. 2016; 48: 1473–80.Google Scholar
Hutcheon, JA, Zhang, X, Platt, RW, Cnattingius, S, Kramer, MS. The case against customised birthweight standards. Paediatr Perinat Epidemiol. 2011; 25: 1116.Google Scholar
Gaccioli, F, Sovio, U, Cook, E, Hund, M, Charnock-Jones, DS, Smith, GCS. Screening for fetal growth restriction using ultrasound and the sFLT1:PlGF ratio in a prospective cohort study of nulliparous women. Lancet Child Adolesc Health. 2018; 2: 569–81.Google Scholar
Duley, L, Henderson-Smart, DJ, Meher, S, King, JF. Antiplatelet agents for preventing pre-eclampsia and its complications. Cochrane Database Syst Rev. 2007; 2: CD004659.Google Scholar
Groom, KM, McCowan, LM, Mackay, LK, Lee, AC, Said, JM, Kane, SC, et al. Enoxaparin for the prevention of preeclampsia and intrauterine growth restriction in women with a history: a randomized trial. Am J Obstet Gynecol. 2017; 216: e1296.Google Scholar
Sharp, A, Cornforth, C, Jackson, R, Harrold, J, Turner, MA, Kenny, LC, et al. Maternal sildenafil for severe fetal growth restriction (STRIDER): a multicentre, randomised, placebo-controlled, double-blind trial. Lancet Child Adolesc Health. 2018; 2: 93102.Google Scholar
Wyrwoll, CS, Noble, J, Thomson, A, Tesic, D, Miller, MR, Rog-Zielinska, EA, et al. Pravastatin ameliorates placental vascular defects, fetal growth, and cardiac function in a model of glucocorticoid excess. Proc Natl Acad Sci U S A. 2016; 113: 6265–70.Google Scholar
Brownfoot, FC, Tong, S, Hannan, NJ, Binder, NK, Walker, SP, Cannon, P, et al. Effects of pravastatin on human placenta, endothelium, and women with severe preeclampsia. Hypertension. 2015; 66: 687–97.Google Scholar
Brownfoot, FC, Hastie, R, Hannan, NJ, Cannon, P, Tuohey, L, Parry, LJ, et al. Metformin as a prevention and treatment for preeclampsia: effects on soluble fms-like tyrosine kinase 1 and soluble endoglin secretion and endothelial dysfunction. Am J Obstet Gynecol. 2016; 214: e1356.Google Scholar
Nawaz, FH, Khalid, R, Naru, T, Rizvi, J. Does continuous use of metformin throughout pregnancy improve pregnancy outcomes in women with polycystic ovarian syndrome? J Obstet Gynecol Res. 2008; 34: 832–7.Google Scholar
Onda, K, Tong, S, Beard, S, Binder, N, Muto, M, Senadheera, SN, et al. Proton pump inhibitors decrease soluble fms-like tyrosine kinase-1 and soluble endoglin secretion, decrease hypertension and rescue endothelial dysfunction. Hypertension. 2017; 69: 457–68.Google Scholar
Miller, SL, Yawno, T, Alers, NO, Castillo-Melendez, M, Supramaniam, VG, van Zyl, N, et al. Antenatal antioxidant treatment with melatonin to decrease newborn neurodevelopmental deficits and brain injury caused by fetal growth restriction. J Pineal Res. 2014; 56: 283–94.Google Scholar
Chang, EY, Barbosa, E, Paintila, MK, Singh, A, Singh, I. The use of N-acetylcysteine for the prevention of hypertension in the reduced uterine perfusion pressure model for preeclampsia in Sprague-Dawley rats. Am J Obstet Gynecol. 2005; 193: 952–6.Google Scholar
Roes, EM, Raijmakers, MT, de Boo, TM, Zusterzeel, PL, Merkus, HM, Peters, WH, et al. Oral N-acetylcysteine administration does not stabilise the process of established severe preeclampsia. Eur J Obstet Gynecol. 2006; 127: 61–7.Google Scholar
Soothill, PW, Nicolaides, KH, Bilardo, CM, Campbell, S. Relation of fetal hypoxia in growth retardation to mean blood velocity in the fetal aorta. Lancet. 1986; 2: 1118–20.Google Scholar
GRIT Study Group. A randomised trial of timed delivery for the compromised preterm fetus: short-term outcomes and Bayesian interpretation. BJOG. 2003; 110: 2732.Google Scholar
Walker, DM, Marlow, N, Upstone, L, Gross, H, Hornbuckle, J, Vail, A, et al. The Growth Restriction Intervention Trial: long-term outcomes in a randomized trial of timing of delivery in fetal growth restriction. Am J Obstet Gynecol. 2011; 2014: e1–9.Google Scholar
Morris, RK, Malin, G, Robson, SC, Kleijnen, J, Zamora, J, Khan, KS. Fetal umbilical artery Doppler to predict compromise of fetal/neonatal wellbeing in a high-risk population: systematic review and bivariate meta-analysis. Ultrasound Obstet Gynecol. 2011; 37: 135–42.Google Scholar
American College of Obstetricians and Gynecologists. ACOG Practice Bulletin no. 134: fetal growth restriction. Obstet Gynecol. 2013; 121: 1122–33.Google Scholar
Whitehead, CL, Walker, SP, Mendis, S, Lappas, M, Tong, S. Quantifying mRNA coding growth genes in the maternal circulation to detect fetal growth restriction. Am J Obstet Gynecol. 2013; 209: e1–9.Google Scholar
Wӧlter, M, Rӧwer, C, Koy, C, Rath, W, Pecks, U, Glocker, MO. Proteoform profiling of peripheral blood serum proteins from pregnant women provides a molecular IUGR signature. J Proteomics. 2016; 149: 4452.Google Scholar
Maitre, L, Fthenou, E, Athersuch, T, Coen, M, Toledano, MB, Holmes, E, et al. Urinary metabolic profiles in early pregnancy are associated with preterm birth and fetal growth restriction in the Rhea mother–child cohort study. BMC Med. 2014; 12: 10.Google Scholar
Pantichob, N, Widdows, KL, Crocker, IP, Johnstone, ED, Please, CP, Sibley, CP, et al. Computational modelling of placental amino acid transfer as an integrated system. Biochim Biophys Acta. 2016; 1858: 1451–61.Google Scholar
Chappell, LC, David, AL. Improving the Pipeline for Developing and Testing Pharmacological Treatments in Pregnancy. PLoS Med. 2016; 13: e1002161.Google Scholar
King, A, Ndifon, C, Lui, S, Widdows, K, Kotamraju, VR, Agemy, L, et al. Tumor-homing peptides as tools for targeted delivery of payloads to the placenta. Sci Adv. 2016; 2: e1600349.Google Scholar
Cureton, N, Korotkova, I, Baker, B, Greenwood, S, Wareing, M, Kotamraji, VR, et al. Selective targeting of a novel vasodilator to the uterine vasculature to treat impaired uteroplacental perfusion in pregnancy. Theranostics. 2017; 7: 3715–31.Google Scholar
Spencer, R, Ambler, G, Brodszki, J, Diemert, A, Figueras, F, Gratacós, E, et al. EVERREST prospective study: a 6-year prospective study to define the clinical and biological characteristics of pregnancies affected by severe early onset fetal growth restriction. BMC Pregnancy Childbirth. 2017; 17: 43.Google Scholar

References

Brosens, I, Pijnenborg, R, Benagiano, G. Defective myometrial spiral artery remodelling as a cause of major obstetrical syndromes in endometriosis and adenomyosis. Placenta. 2013; 34: 100–5.Google Scholar
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: 1046–54.Google Scholar
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: 301–5.Google Scholar
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: 1668–73.Google Scholar
Timmermans, S, Steegers-Theunissen, RP, Vujkovic, M, den Breeijen, H, Russcher, H, Lindemans, J, et al. The Mediterranean diet and fetal size parameters: the Generation R Study. Br J Nutr. 2012; 108: 1399–409.Google Scholar
Mahon, P, Harvey, N, Crozier, S, Inskip, H, Robinson, S, Arden, NK, et al. Low maternal vitamin D status and fetal bone development: cohort study. J Bone Min Res. 2010; 25: 1419.Google Scholar
Stephenson, J, Heslehurst, N, Hall, J, Schoenaker, DAJM, Hutchinson, J, Cade, JE, et al. Before the beginning: nutrition and lifestyle in the preconception period and its importance for future health. Lancet. 2018; 391: 1830–41.Google Scholar
Fleming, TP, Watkins, AJ, Velazquez, MA, Mathers, JC, Prentice, AM, Stephenson, J, et al. Origins of lifetime health around the time of conception: causes and consequences. Lancet. 2018; 391: 1842–52.Google Scholar
Hillman, S, Peebles, DM, Williams, DJ. Paternal metabolic and cardiovascular risk factors for fetal growth restriction: a case–control study. Diabetes Care. 2013; 36: 1675–80.Google Scholar
Barker, M, Dombrowski, SU, Colbourn, T, Fall, CHD, Kriznik, NM, Lawrence, WT, et al. Intervention strategies to improve nutrition and health behaviours before conception. Lancet. 2018; 391: 1853–64.Google Scholar
Knudsen, VK, Orozova-Bekkevold, IM, Mikkelsen, TB, Wolff, S, Olsen, SF. Major dietary patterns in pregnancy and fetal growth. Eur J Clin Nutr. 2008; 62: 463–70.Google Scholar
Ota, E, Hori, H, Mori, R, Tobe-Gai, R, Farrar, D. Antenatal dietary education and supplementation to increase energy and protein intake. Cochrane Database Syst Rev. 2015; 6: CD000032.Google Scholar
Haider, BA, Bhutta, ZA. Multiple-micronutrient supplementation for women during pregnancy. Cochrane Database Syst Rev. 2017; 4: CD004905.Google Scholar
Peña-Rosas, JP, De-Regil, LM, Gomez Malave, H, Flores-Urrutia, MC, Dowswell, T. Intermittent oral iron supplementation during pregnancy. Cochrane Database Syst Rev. 2015; 11: CD009997.Google Scholar
Peña-Rosas, JP, De-Regil, LM, Garcia-Casal, MN, Dowswell, T. Daily oral iron supplementation during pregnancy. Cochrane Database Syst Rev. 2015; 7: CD004736.Google Scholar
Hofmeyr, GJ, Lawrie, TA, Atallah, ÁN, Duley, L, Torloni, MR. Calcium supplementation during pregnancy for preventing hypertensive disorders and related problems. Cochrane Database Syst Rev. 2014; 6: CD001059.Google Scholar
Rumbold, A, Duley, L, Crowther, CA, Haslam, RR. Antioxidants for preventing pre-eclampsia. Cochrane Database Syst Rev. 2008; 1: CD004227.Google Scholar
Han, Z, Mulla, S, Beyene, J, Liao, G, McDonald, SD, Knowledge Synthesis Group. Maternal underweight and the risk of preterm birth and low birth weight: a systematic review and meta-analyses. Int J Epidemiol. 2011; 40: 65101.Google Scholar
Yaktine, AL, Rasmussen, KM (eds.). Weight Gain During Pregnancy: Reexamining the Guidelines. Washington, DC: National Academies Press, 2009.Google Scholar
Radulescu, L, Munteanu, O, Popa, F, Cirstoiu, M. The implications and consequences of maternal obesity on fetal intrauterine growth restriction. J Med Life. 2013; 6: 292–8.Google Scholar
CARE Study Group. Maternal caffeine intake during pregnancy and risk of fetal growth restriction: a large prospective observational study. BMJ. 2008; 337: a2332.Google Scholar
Wills, R-A, Coory, MD. Effect of smoking among Indigenous and non-Indigenous mothers on preterm birth and full-term low birthweight. Med J Aust. 2008; 189: 490–4.Google Scholar
Riisinen, S, Sankilampi, U, Gissler, M, Kramer, MR, Hakulinen-Viitanen, T, Saari, J, et al. Smoking cessation in the first trimester reduces most obstetric risks, but not the risks of major congenital anomalies and admission to neonatal care: a population-based cohort study of 1,164,953 singleton pregnancies in Finland. J Epidemiol Community Health. 2014; 68: 159–64.Google Scholar
Suzuki, K, Sato, M, Zheng, W, Shinohara, R, Yokomichi, H, Yamagata, Z. Effect of maternal smoking cessation before and during early pregnancy on fetal and childhood growth. J Epidemiol. 2014; 24: 60–6.Google Scholar
Lumley, J, Chamberlain, C, Dowswell, T, Oliver, S, Oakley, L, Watson, L. Interventions for promoting smoking cessation during pregnancy. Cochrane Database Syst Rev. 2009; 3: CD001055.Google Scholar
Abraham, M, Alramadhan, S, Iniguez, C, Duijts, L, Jaddoe, VW, Den Dekker, HT, et al. A systematic review of maternal smoking during pregnancy and fetal measurements with meta-analysis. PLoS ONE. 2017; 12: e0170946.Google Scholar
Fergusson, DM, Horwood, LJ, Northstone, K. Maternal use of cannabis and pregnancy outcome. BJOG. 2002; 109: 21–7.Google Scholar
Gouin, K, Murphy, K, Shah, PS. Effects of cocaine use during pregnancy on low birthweight and preterm birth: Systematic review and metaanalyses. Am J Obstet Gynecol. 2011; 204: 340. e1–12.Google Scholar
Ladhani, NNN, Shah, PS, Murphy, KE. Prenatal amphetamine exposure and birth outcomes: a systematic review and metaanalysis. Am J Obstet Gynecol. 2011; 205: 219. e1–219. e7.Google Scholar
Whitehead, N, Lipscomb, L. Patterns of alcohol use before and during pregnancy and the risk of small-for-gestational-age birth. Am J Epidemiol. 2003; 158: 654–62.Google Scholar
Patra, J, Bakker, R, Irving, H, Jaddoe, V, Malini, S, Rehm, J. Dose-response relationship between alcohol consumption before and during pregnancy and the risks of low birthweight, preterm birth and small for gestational age (SGA) – a systematic review and meta-analyses. BJOG. 2011; 118: 1411–21.Google Scholar
Abalos, E, Duley, L, Steyn, DW. Antihypertensive drug therapy for mild to moderate hypertension during pregnancy. Cochrane Database Syst Rev. 2014; 1: CD002252.Google Scholar
Magee, LA, von Dadelszen, P, Rey, E, Ross, S, Asztalos, E, Murphy, KE, et al. Less-Tight versus Tight Control of Hypertension in Pregnancy. N Engl J Med. 2015; 372: 407–17.Google Scholar
Groom, KM, David, AL. The role of aspirin, heparin, and other interventions in the prevention and treatment of fetal growth restriction. Am J Obstet Gynecol. 2018; 218: S829–40.Google Scholar
Taubert, D, Berkels, R, Grosser, N, Schröder, H, Gründemann, D, Schömig, E. Aspirin induces nitric oxide release from vascular endothelium: a novel mechanism of action. Br J Pharmacol. 2004; 143: 159–65.Google Scholar
Grosser, N, Abate, A, Oberle, S, Vreman, HJ, Dennery, PA, Becker, JC, et al. Heme oxygenase-1 induction may explain the antioxidant profile of aspirin. Biochem Biophys Res Commun. 2003; 308: 956–60.Google Scholar
Roberge, S, Nicolaides, K, Demers, S, Hyett, J, Chaillet, N, Bujold, E. The role of aspirin dose on the prevention of preeclampsia and fetal growth restriction: systematic review and meta-analysis. Am J Obstet Gynecol. 2017; 216: 110120. e6.Google Scholar
Meher, S, Duley, L, Hunter, K, Askie, L. Antiplatelet therapy before or after 16 weeks’ gestation for preventing preeclampsia: an individual participant data meta-analysis. Am J Obstet Gynecol. 2017; 216: 121128. e2.Google Scholar
Ayala, DE, Ucieda, R, Hermida, RC. Chronotherapy with low-dose aspirin for prevention of complications in pregnancy. Chronobiol Int. 2013; 30: 260–79.Google Scholar
Hermida, RC, Ayala, DE, Iglesias, M. Administration time-dependent influence of aspirin on blood pressure in pregnant women. Hypertension. 2003; 41: 651–6.Google Scholar
McCowan, LM, Figueras, F, Anderson, NH. Evidence-based national guidelines for the management of suspected fetal growth restriction: comparison, consensus, and controversy. Am J Obstet Gynecol. 2018: 218: S855–68.Google Scholar
Rolnik, DL, Wright, D, Poon, LC, O’Gorman, N, Syngelaki, A, de Paco Matallana, C, et al. Aspirin versus Placebo in Pregnancies at High Risk for Preterm Preeclampsia. N Engl J Med. 2017; 377: 613–22.Google Scholar
Omri, A, Delaloye, JF, Andersen, H, Bachmann, F. Low molecular weight heparin Novo (LHN-1) does not cross the placenta during the second trimester of pregnancy. Thromb Haemost. 1989; 61: 55–6.Google Scholar
Mousavi, S, Moradi, M, Khorshidahmad, T, Motamedi, M. Anti-Inflammatory effects of heparin and its derivatives: a systematic review. Adv Pharmacol Sci. 2015; 2015: 507151.Google Scholar
Oberkersch, R, Attorresi, AI, Calabrese, GC. Low-molecular-weight heparin inhibition in classical complement activation pathway during pregnancy. Thromb Res. 2010; 125: e240–5.Google Scholar
Mousa, SA, Petersen, LJ. Anti-cancer properties of low-molecular-weight heparin: preclinical evidence. Thromb Haemost. 2009; 102: 258–67.Google Scholar
Bose, P, Black, S, Kadyrov, M, Bartz, C, Shlebak, A, Regan, L, et al. Adverse effects of lupus anticoagulant positive blood sera on placental viability can be prevented by heparin in vitro. Am J Obstet Gynecol. 2004; 191: 2125–31.Google Scholar
Sobel, ML, Kingdom, J, Drewlo, S. Angiogenic response of placental villi to heparin. Obstet Gynecol. 2011; 117: 1375–83.Google Scholar
Di Simone, N, Di Nicuolo, F, Sanguinetti, M, Ferrazzani, S, D’Alessio, MC, Castellani, R, et al. Low-molecular weight heparin induces in vitro trophoblast invasiveness: role of matrix metalloproteinases and tissue inhibitors. Placenta. 2007; 28: 298304.Google Scholar
Yinon, Y, Ben Meir, E, Margolis, L, Lipitz, S, Schiff, E, Mazaki-Tovi, S, et al. Low molecular weight heparin therapy during pregnancy is associated with elevated circulatory levels of placental growth factor. Placenta. 2015; 36: 121–4.Google Scholar
McLaughlin, K, Baczyk, D, Potts, A, Hladunewich, M, Parker, JD, Kingdom, JCP. Low molecular weight heparin improves endothelial function in pregnant women at high risk of Preeclampsia. Hypertension. 2016; 1–9.Google Scholar
Torricelli, M, Reis, FM, Florio, P, Severi, FM, Bocchi, C, Picciolini, E, et al. Low-molecular-weight heparin improves the performance of uterine artery Doppler velocimetry to predict preeclampsia and small-for-gestational age infant in women with gestational hypertension. Ultrasound Med Biol. 2006; 32: 1431–5.Google Scholar
Abheiden, CNH, Van Hoorn, ME, Hague, WM, Kostense, PJ, Van Pampus, MG, De Vries, JIP. Does low-molecular-weight heparin influence fetal growth or uterine and umbilical arterial Doppler in women with a history of early-onset uteroplacental insufficiency and an inheritable thrombophilia? Secondary randomised controlled trial results. BJOG. 2016; 123: 797805.Google Scholar
de Vries, JIP, Hague, WM, van Pampus, MG. Low-molecular-weight heparin added to aspirin in the prevention of recurrent early-onset pre-eclampsia in women with inheritable thrombophilia: the FRUIT-RCT: a reply to a rebuttal. J Thromb Haemost. 2012; 10: 1196.Google Scholar
Rey, E, Garneau, P, David, M, Gauthier, R, Leduc, L, Michon, N, et al. Dalteparin for the prevention of recurrence of placental-mediated complications of pregnancy in women without thrombophilia: a pilot randomized controlled trial. J Thromb Haemost. 2009; 7: 5864.Google Scholar
Mastrolia, SA, Novack, L, Thachil, J, Rabinovich, A, Pikovsky, O, Klaitman, V, et al. LMWH in the prevention of preeclampsia and fetal growth restriction in women without thrombophilia: a systematic review and meta-analysis. Thromb Haemost. 2016; 116: 868–78.Google Scholar
Say, L, Gülmezoglu, AM, Hofmeyr, GJ. Maternal oxygen administration for suspected impaired fetal growth. Cochrane Database Syst Rev. 2003; 1: CD000137.Google Scholar
David, AL, Thornton, S, Sutcliffe, A, Williams, P. (2015). Developing New Pharmaceutical Treatments for Obstetric Conditions. Royal College of Obstetricians & Gynaecologists. www.rcog.org.uk/globalassets/documents/guidelines/scientific-impact-papers/sip-50.pdfGoogle Scholar
Learmont, JG, Poston, L. Nitric oxide is involved in flow-induced dilation of isolated human small fetoplacental arteries. Am J Obstet Gynecol. 1996; 174: 583–8.Google Scholar
Duvekot, JJ, Cheriex, EC, Pieters, FAA, Menheere, PPCA, Schouten, HJA, Peeters, LLH. Maternal volume homeostasis in early pregnancy in relation to fetal growth restriction. Obstet Gynecol. 1995; 85: 361–7.Google Scholar
Duvekot, JJ, Cheriex, EC, Pieters, FA, Peeters, LL. Severely impaired fetal growth is preceded by maternal hemodynamic maladaptation in very early pregnancy. Acta Obstet Gynecol Scand. 1995; 74: 693–7.Google Scholar
Krause, BJ, Carrasco-Wong, I, Caniuguir, A, Carvajal, J, Farías, M, Casanello, P. Endothelial eNOS/arginase imbalance contributes to vascular dysfunction in IUGR umbilical and placental vessels. Placenta. 2013; 34: 20–8.Google Scholar
Kulandavelu, S, Whiteley, KJ, Bainbridge, SA, Qu, D, Adamson, SL. Endothelial NO synthase augments fetoplacental blood flow, placental vascularization, and fetal growth in mice. Hypertension. 2013; 61: 259–66.Google Scholar
Kähler, C, Schleußner, E, Möller, A, Seewald, HJ. Nitric oxide donors: effects on fetoplacental blood flow. Eur J Obstet Gynecol Reprod Biol. 2004; 115: 1014.Google Scholar
Meher, S, Duley, L. Nitric oxide for preventing pre-eclampsia and its complications. Cochrane Database Syst Rev. 2007; 2: CD006490.Google Scholar
Tiralongo, G, Pisani, I, Vasapollo, B, Khalil, A, Thilaganathan, B, Valensise, H. Nitric oxide (NO) donors and haemodynamic changes in fetal growth restriction. Ultrasound Obstet Gynecol. 2018; 4: 514–18.Google Scholar
Ormesher, L, Myers, JE, Chmiel, C, Wareing, M, Greenwood, SL, Tropea, T, et al. Effects of dietary nitrate supplementation, from beetroot juice, on blood pressure in hypertensive pregnant women: a randomised, double-blind, placebo-controlled feasibility trial. Nitric Oxide. 2018; 80: 3744.Google Scholar
Schleussner, E, Lehmann, T, Kähler, C, Schneider, U, Schlembach, D, Groten, T. Impact of the nitric oxide-donor pentaerythrityltetranitrate on perinatal outcome in risk pregnancies: a prospective, randomized, double-blinded trial. J Perinat Med. 2014; 42: 507–14.Google Scholar
Wareing, M, Myers, JE, O’Hara, M, Kenny, LC, Warren, AY, Taggart, MJ, et al. Effects of a phosphodiesterase-5 (PDE5) inhibitor on endothelium-dependent relaxation of myometrial small arteries. Am J Obstet Gynecol. 2004; 190: 1283–90.Google Scholar
Wareing, M, Myers, JE, O’Hara, M, Baker, PN. Sildenafil citrate (Viagra) enhances vasodilatation in fetal growth restriction. J Clin Endocrinol Metab. 2005; 90: 2550–5.Google Scholar
Russo, FM, Conings, S, Allegaert, K, van Mieghem, T, Toelen, J, van Calsteren, K, et al. Sildenafil crosses the placenta at therapeutic levels in a dually perfused human cotyledon model. Am J Obstet Gynecol. 2018; 219: 619. e1-619. e10.Google Scholar
Walton, RB, Reed, LC, Estrada, SM, Schmiedecke, SS, Villazana-Kretzer, DL, Napolitano, PG, et al. Evaluation of sildenafil and tadalafil for reversing constriction of fetal arteries in a human placenta perfusion model. Hypertension. 2018; 72: 167–76.Google Scholar
Samangaya, RA, Mires, G, Shennan, A, Skillern, L, Howe, D, McLeod, A, et al. A randomised, double-blinded, placebo-controlled study of the phosphodiesterase type 5 inhibitor sildenafil for the treatment of preeclampsia. Hypertens Pregnancy. 2009; 28: 369–82.Google Scholar
Oyston, C, Stanley, JL, Oliver, MH, Bloomfield, FH, Baker, PN. Maternal administration of sildenafil citrate alters fetal and placental growth and fetal-placental vascular resistance in the growth-restricted ovine fetus. Hypertension. 2016; 68: 760–7.Google Scholar
Miller, SL, Loose, JM, Jenkin, G, Wallace, EM. The effects of sildenafil citrate (Viagra) on uterine blood flow and well being in the intrauterine growth-restricted fetus. Am J Obstet Gynecol. 2009; 200: 102. e1–7.Google Scholar
Von Dadelszen, P, Dwinnell, S, Magee, LA, Carleton, BC, Gruslin, A, Lee, B, et al. Sildenafil citrate therapy for severe early-onset intrauterine growth restriction. BJOG. 2011; 118: 624–8.Google Scholar
Adel El-Sayed, M, Abdel-Aty Saleh, S, Ahmed Maher, M, Khidre, AM. Utero-placental perfusion Doppler indices in growth restricted fetuses: effect of sildenafil citrate. J Matern Fetal Neonatal Med. 2018; 31: 1045–50.Google Scholar
Ganzevoort, W, Alfirevic, Z, von Dadelszen, P, Kenny, L, Papageorghiou, A, van Wassenaer-Leemhuis, A, et al. STRIDER: Sildenafil therapy in dismal prognosis early-onset intrauterine growth restriction? A protocol for a systematic review with individual participant data and aggregate data meta-analysis and trial sequential analysis. Syst Rev. 2014; 3: 23.Google Scholar
Pels, A, Kenny, LC, Alfirevic, Z, Baker, PN, von Dadelszen, P, Gluud, C, et al. STRIDER (Sildenafil TheRapy in dismal prognosis early onset fetal growth restriction): an international consortium of randomised placebo-controlled trials. BMC Pregnancy Childbirth. 2017; 17: 440.Google Scholar
Sharp, A, Cornforth, C, Jackson, R, Harrold, J, Turner, MA, Kenny, LC, et al. Maternal sildenafil for severe fetal growth restriction (STRIDER ): a multicentre, randomised, placebo-controlled, double-blind trial. Lancet Child Adolesc Health. 2018; 2: 93102.Google Scholar
Groom, KM, Ganzevoort, W, Alfirevic, Z, Lim, K, Papageorghiou, AT. Clinicians should stop prescribing sildenafil for fetal growth restriction (FGR): comment from the STRIDER Consortium. Ultrasound Obstet Gynecol. 2018; 52: 295–6.Google Scholar
Tanaka, K, Tanaka, H, Maki, S, Kubo, M, Nii, M, Magawa, S, et al. Cardiac function and tadalafil used for treating fetal growth restriction in pregnant women without cardiovascular disease. J Matern Neonatal Med. 2018; 20: 13.Google Scholar
Kubo, M, Umekawa, T, Maekawa, Y, Tanaka, H, Nii, M, Murabayashi, N, et al. Retrospective study of tadalafil for fetal growth restriction: impact on maternal and perinatal outcomes. J Obstet Gynaecol Res. 2017; 43: 291–7.Google Scholar
Tanaka, H, Kubo, M, Nii, M, Maki, S, Umekawa, T, Ikeda, T. Treatment using tadalafil for severe pre-eclampsia with fetal growth restriction. J Obstet Gynaecol Res. 2017; 43: 1205–8.Google Scholar
Sakamoto, M, Osato, K, Kubo, M, Nii, M, Tanaka, H, Murabayashi, N, et al. Early-onset fetal growth restriction treated with the long-acting phosphodiesterase-5 inhibitor tadalafil: a case report. J Med Case Rep. 2016; 10: 15.Google Scholar
Kubo-Kaneda, M, Tanaka, H, Maki, S, Nii, M, Umekawa, T, Osato, K, et al. Placental growth factor as a predictor of the efficacy of tadalafil treatment for fetal growth restriction. J Matern Neonatal Med. 2018; 0: 14.Google Scholar
Belo, L, Caslake, M, Santos-Silva, A, Castro, EMB, Pereira-Leite, L, Quintanilha, A, et al. LDL size, total antioxidant status and oxidised LDL in normal human pregnancy: a longitudinal study. Atherosclerosis. 2004; 177: 391–9.Google Scholar
Bauer, AJ, Banek, CT, Needham, K, Gillham, H, Capoccia, S, Regal, JF, et al. Pravastatin attenuates hypertension, oxidative stress, and angiogenic imbalance in rat model of placental ischemia-induced hypertension. Hypertension. 2013; 61: 1103–10.Google Scholar
Yang, M-Y, Diao, Z-Y, Wang, Z-Y, Yan, G-J, Zhao, G-F, Zheng, M-M, et al. Pravastatin alleviates lipopolysaccharide-induced placental TLR4 over-activation and promotes uterine arteriole remodeling without impairing fetal development of rats. J Biomed Res. 2018; 32: 288–97.Google Scholar
Ofori, B, Oraichi, D, Blais, L, Rey, E, Berard, A. Risk of congenital anomalies in pregnant users of non-steroidal anti-inflammatory drugs: a nested case-control study. Birth Defects Res B Dev Reprod Toxicol. 2006; 77: 268–79.Google Scholar
Kane, AD, Herrera, EA, Hansell, JA, Giussani, DA. Statin treatment depresses the fetal defence to acute hypoxia via increasing nitric oxide bioavailability. J Physiol. 2012; 590: 323–34.Google Scholar
Costantine, MM, Cleary, K. Pravastatin for the prevention of preeclampsia in high-risk pregnant women. Obstet Gynecol. 2013; 121: 349–53.Google Scholar
Lefkou, E, Mamopoulos, A, Dagklis, T, Vosnakis, C, Rousso, D, Girardi, G. Pravastatin improves pregnancy outcomes in obstetric antiphospholipid syndrome refractory to antithrombotic therapy. J Clin Invest. 2016; 126: 2933–40.Google Scholar
Kaitu’u-Lino, TJ, Brownfoot, FC, Beard, S, Cannon, P, Hastie, R, Nguyen, TV, et al. Combining metformin and esomeprazole is additive in reducing sFlt-1 secretion and decreasing endothelial dysfunction – implications for treating preeclampsia. PLoS ONE. 2018; 13: 114.Google Scholar
Cluver, CA, Walker, SP, Mol, BW, Theron, GB, Hall, DR, Hiscock, R, et al. Double blind, randomised, placebo-controlled trial to evaluate the efficacy of esomeprazole to treat early onset pre-eclampsia (PIE Trial): a study protocol. BMJ Open. 2015; 5: e008211.Google Scholar
Wang, M-J, Cai, W-J, Li, N, Ding, Y-J, Chen, Y, Zhu, Y-C. The hydrogen sulfide donor NaHS promotes angiogenesis in a rat model of hind limb ischemia. Antioxid Redox Signal. 2010; 12: 1065–77.Google Scholar
Cindrova-Davies, T, Herrera, EA, Niu, Y, Kingdom, J, Giussani, DA, Burton, GJ. Reduced cystathionine γ-lyase and increased miR-21 expression are associated with increased vascular resistance in growth-restricted pregnancies: hydrogen sulfide as a placental vasodilator. Am J Pathol. 2013; 182: 1448–58.Google Scholar
Holwerda, KM, Burke, SD, Faas, MM, Zsengeller, Z, Stillman, IE, Kang, PM, et al. Hydrogen sulfide attenuates sFlt1-induced hypertension and renal damage by upregulating vascular endothelial growth factor. J Am Soc Nephrol. 2014; 25: 717–25.Google Scholar
Wang, Y, Walli, AK, Schulze, A, Blessing, F, Fraunberger, P, Thaler, C, et al. Heparin-mediated extracorporeal low density lipoprotein precipitation as a possible therapeutic approach in preeclampsia. Transfus Apher Sci. 2006; 35: 103–10.Google Scholar
David, AL, Torondel, B, Zachary, I, Wigley, V, Nader, KA, 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: 1344–50.Google Scholar
Mehta, V, Abi-Nader, K, Peebles, D, 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: 925–35.Google Scholar
Carr, DJ, Aitken, RP, Milne, JS, Peebles, DM, Martin, JF, Zachary, IC, et al. Prenatal Ad.VEGF gene therapy – a promising new treatment for fetal growth restriction. Hum Gene Ther. 2011; 22: A128.Google Scholar
Carr, DJ, Wallace, JM, Aitken, R, Milne, JS, Martin, JF, Zachary, IZ, et al. Peri- and postnatal effects of prenatal adenoviral VEGF gene therapy in growth-restricted sheep. Biol Reprod. 2016; 94: 142.Google Scholar
Swanson, AM, Rossi, CA, Ofir, K, Mehta, V, Boyd, M, Barker, H, et al. Maternal therapy with Ad.VEGF-A165 increases fetal weight at term in a guinea pig model of fetal growth restriction. Hum Gene Ther. 2016; 27: 9971007.Google Scholar
Vaughan, OR, Rossi, CA, Ginsberg, Y, White, A, Hristova, M, Sebire, NJ, et al. Perinatal and long-term effects of maternal uterine artery adenoviral VEGF-A165 gene therapy in the growth-restricted guinea pig fetus. Am J Physiol Regul Integr Comp Physiol. 2018; 315: R344–53.Google Scholar
David, AL. Maternal uterine artery VEGF gene therapy for treatment of intrauterine growth restriction. Placenta. 2017; 59 (Suppl. 1): S44–50.Google Scholar
Sheppard, M, Spencer, RN, Ashcroft, R, David, AL. Ethics and social acceptability of a proposed clinical trial using maternal gene therapy to treat severe early-onset fetal growth restriction. Ultrasound Obstet Gynecol. 2016; 47: 484–91.Google Scholar
Spencer, R, Ambler, G, Brodszki, J, Diemert, A, Figueras, F, Gratacós, E, et al. EVERREST prospective study: a 6-year prospective study to define the clinical and biological characteristics of pregnancies affected by severe early onset fetal growth restriction. BMC Pregnancy Childbirth. 2017; 17: 43.Google Scholar
Royal College of Obstetricians and Gynaecologists. (2007). The Role of Emergency and Elective Interventional Radiology in Postpartum Haemorrhage. www.rcog.org.uk/globalassets/documents/guidelines/goodpractice6roleemergency2007.pdf.Google Scholar
Camacho-Hübner, C, Woods, KA, Clark, AJL, Savage, MO. Insulin-like growth factor (IGF)-I gene deletion. Rev Endocr Metab Disord. 2002; 3: 357–61.Google Scholar
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: e74632.Google Scholar
Harris, LK. Could peptide-decorated nanoparticles provide an improved approach for treating pregnancy complications? Nanomedicine. 2016; 11: 2235–8.Google Scholar
King, A, Ndifon, C, Lui, S, Widdows, K, Kotamraju, VR, Agemy, L, et al. Tumor-homing peptides as tools for targeted delivery of payloads to the placenta. Sci Adv. 2016; 2: e1600349.Google Scholar
Cureton, N, Korotkova, I, Baker, B, Greenwood, S, Wareing, M, Kotamraju, R, et al. Selective targeting of a novel vasodilator to the uterine vasculature to treat impaired uteroplacental perfusion in pregnancy. Theranostics. 2017; 7: 3715–31.Google Scholar
Phillips, TJ, Scott, H, Menassa, DA, Bignell, AL, Sood, A, Morton, JS, et al. Treating the placenta to prevent adverse effects of gestational hypoxia on fetal brain development. Sci Rep. 2017; 7: 9079.Google Scholar
Beards, F, Jones, LE, Charnock, J, Forbes, K, Harris, LK. Placental homing peptide-microRNA inhibitor conjugates for targeted enhancement of intrinsic placental growth signaling. Theranostics. 2017; 7: 2940–55.Google Scholar
Vasapollo, B, Lo Presti, D, Gagliardi, G, Farsetti, D, Tiralongo, GM, Pisani, I, et al. Restricted physical activity in pregnancy reduces maternal vascular resistance and improves fetal growth. Ultrasound Obstet Gynecol. 2018; 51: 672–6.Google Scholar
Renshall, LJ, Morgan, HL, Moens, H, Cansfield, D, Finn-Sell, SL, Tropea, T, et al. Melatonin increases fetal weight in wild-type mice but not in mouse models of fetal growth restriction. Front Physiol. 2018; 9: 111.Google Scholar
Castillo-Melendez, M, Yawno, T, Sutherland, A, Jenkin, G, Wallace, EM, Miller, SL. Effects of antenatal melatonin treatment on the cerebral vasculature in an ovine model of fetal growth restriction. Dev Neurosci. 2017; 39: 323–37.Google Scholar
Chang, EY, Barbosa, E, Paintlia, MK, Singh, A, Singh, I. The use of N-acetylcysteine for the prevention of hypertension in the reduced uterine perfusion pressure model for preeclampsia in Sprague-Dawley rats. Am J Obstet Gynecol. 2005; 193: 952–6.Google Scholar
Hashimoto, K, Pinkas, G, Evans, L, Liu, H, Al-Hasan, Y, Thompson, LP. Protective effect of N-acetylcysteine on liver damage during chronic intrauterine hypoxia in fetal guinea pig. Reprod Sci. 2012; 19: 1001–9.Google Scholar
Roes, EM, Raijmakers, MT, Boo, TM, de Zusterzeel, PL, Merkus, HM, Peters, WH, et al. Oral N-acetylcysteine administration does not stabilise the process of established severe preeclampsia. Eur J Obstet Gynecol Reprod Biol. 2006; 127: 61–7.Google Scholar
LaRosa, DA, Ellery, SJ, Parkington, HC, Snow, RJ, Walker, DW, Dickinson, H. Maternal creatine supplementation during pregnancy prevents long-term changes in diaphragm muscle structure and function after birth asphyxia. Pediatr Res. 2016; 80: 852–60.Google Scholar
Dickinson, H, Davies-Tuck, M, Ellery, S, Grieger, J, Wallace, E, Snow, R, et al. Maternal creatine in pregnancy: a retrospective cohort study. BJOG. 2016; 123: 1830–8.Google Scholar
Dickinson, H, Bain, E, Wilkinson, D, Middleton, P, Crowther, CA, Walker, DW. Creatine for women in pregnancy for neuroprotection of the fetus. Cochrane Database Syst Rev. 2014; 12: CD010846.Google Scholar
Rodger, MA, Gris, JC, de Vries, JIP, et al. Low-molecular-weight heparin and recurrent placenta-mediated pregnancy complications: a meta-analysis of individual patient data from randomised controlled trials. Lancet. 2016; 388: 2629–41.Google Scholar

References

Blencowe, H, Cousens, S, Chou, D, Oestergaard, M, Say, L, Moller, AB, et al. Born too soon: the global epidemiology of 15 million preterm births. Reprod Health. 2013; 10: (Suppl. 1): S2.Google Scholar
Gravett, MG, Rubens, CE, Nunes, TM, GAPPS Review Group. Global report on preterm birth and stillbirth (2 of 7): discovery science. BMC Pregnancy Childbirth. 2010; 10: (Suppl. 1): S2.Google Scholar
Romero, R, Kuivaniemi, H, Tromp, G. Functional genomics and proteomics in term and preterm parturition. J Clin Endocrinol Metab. 2002; 87: 2431–4.Google Scholar
Bezold, KY, Karjalainen, MK, Hallman, M, Teramo, K, Muglia, LJ. The genomics of preterm birth: from animal models to human studies. Genome Med. 2013; 5: 34.Google Scholar
Liggins, GC. Initiation of parturition. Br Med Bull. 1979; 35: 145–50.Google Scholar
Smith, R. Parturition. N Engl J Med. 2007; 356: 271–83.Google Scholar
Phillips, JB, Abbot, P, Rokas, A. Is preterm birth a human-specific syndrome? Evol Med Public Health. 2015; 2015: 136–48.Google Scholar
Fischer, B, Mitteroecker, P. Covariation between human pelvis shape, stature, and head size alleviates the obstetric dilemma. Proc Natl Acad Sci U S A. 2015; 112: 5655–60.Google Scholar
de Bernis, L, Kinney, MV, Stones, W, Ten Hoope-Bender, P, Vivio, D, Leisher, SH, et al. Stillbirths: ending preventable deaths by 2030. Lancet. 2016; 387: 703–16.Google Scholar
Phillippe, M. Cell-Free Fetal DNA, Telomeres, and the Spontaneous Onset of Parturition. Reprod Sci. 2015; 22: 1186–201.Google Scholar
Menon, R, Mesiano, S, Taylor, RN. Programmed fetal membrane senescence and exosome-mediated signaling: a mechanism associated with timing of human parturition. Front Endocrinol (Lausanne). 2017; 8: 196.Google Scholar
Menon, R, Bonney, EA, Condon, J, Mesiano, S, Taylor, RN. Novel concepts on pregnancy clocks and alarms: redundancy and synergy in human parturition. Hum Reprod Update. 2016; 22: 535–60.Google Scholar
McLean, M, Bisits, A, Davies, J, Woods, R, Lowry, P, Smith, R. A placental clock controlling the length of human pregnancy. Nat Med. 1995; 1: 460–3.Google Scholar
Gaccioli, F, Sovio, U, Cook, E, Hund, M, Charnock-Jones, DS, Smith, GCS. Screening for fetal growth restriction using ultrasound and the sFLT1/PlGF ratio in nulliparous women: a prospective cohort study. Lancet Child Adolesc Health. 2018; 2: 569–81.Google Scholar
Kim, YM, Bujold, E, Chaiworapongsa, T, Gomez, R, Yoon, BH, Thaler, HT, et al. Failure of physiologic transformation of the spiral arteries in patients with preterm labor and intact membranes. Am J Obstet Gynecol. 2003; 189: 1063–9.Google Scholar
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
Elovitz, MA, Saunders, T, Ascher-Landsberg, J, Phillippe, M. Effects of thrombin on myometrial contractions in vitro and in vivo. Am J Obstet Gynecol. 2000; 183: 799804.Google Scholar
Shynlova, O, Lee, YH, Srikhajon, K, Lye, SJ. Physiologic uterine inflammation and labor onset: integration of endocrine and mechanical signals. Reprod Sci. 2013; 20: 154–67.Google Scholar
Lindstrom, TM, Bennett, PR. The role of nuclear factor kappa B in human labour. Reproduction. 2005; 130: 569–81.Google Scholar
Lindstrom, T, Bennett, P. Transcriptional regulation of genes for enzymes of the prostaglandin biosynthetic pathway. Prostaglandins Leukot Essent Fatty Acids. 2004; 70: 115–35.Google Scholar
Mendelson, CR, Montalbano, AP, Gao, L. Fetal-to-maternal signaling in the timing of birth. J Steroid Biochem Mol Biol. 2017; 170: 1927.Google Scholar
Mesiano, S, Wang, Y, Norwitz, ER. Progesterone receptors in the human pregnancy uterus: do they hold the key to birth timing? Reprod Sci. 2011; 18: 619.Google Scholar
Mendelson, CR. Minireview: fetal-maternal hormonal signaling in pregnancy and labor. Mol Endocrinol. 2009; 23: 947–54.Google Scholar
Nadeem, L, Shynlova, O, Matysiak-Zablocki, E, Mesiano, S, Dong, X, Lye, S. Molecular evidence of functional progesterone withdrawal in human myometrium. Nat Commun. 2016; 7: 11565.Google Scholar
Ridout, AE, Ibeto, L, Ross, G, Cook, JR, Sykes, L, David, AL, et al. Cervical Length and quantitative fetal fibronectin in the prediction of spontaneous preterm birth in asymptomatic women with congenital uterine anomaly. Am J Obstet Gynecol. 2019; pii: S0002-9378(19)30704-5 [Epub ahead of print]Google Scholar
Zhang, X, Zhivaki, D, Lo-Man, R. Unique aspects of the perinatal immune system. Nat Rev Immunol. 2017; 17: 495507.Google Scholar
Rinaldi, SF, Hutchinson, JL, Rossi, AG, Norman, JE. Anti-inflammatory mediators as physiological and pharmacological regulators of parturition. Expert Rev Clin Immunol. 2011; 7: 675–96.Google Scholar
Sykes, L, MacIntyre, DA, Yap, XJ, Teoh, TG, Bennett, PR. The Th1:th2 dichotomy of pregnancy and preterm labour. Mediators Inflamm. 2012; 2012: 967629.Google Scholar
Chen, SJ, Liu, YL, Sytwu, HK. Immunologic regulation in pregnancy: from mechanism to therapeutic strategy for immunomodulation. Clin Dev Immunol. 2012; 2012: 258391.Google Scholar
Aghaeepour, N, Ganio, EA, McIlwain, D, Tsai, AS, Tingle, M, Van Gassen, S, et al. An immune clock of human pregnancy. Sci Immunol. 2017; 2: eaan2946.Google Scholar
Rinaldi, SF, Hutchinson, JL, Rossi, AG, Norman, JE. Anti-inflammatory mediators as physiological and pharmacological regulators of parturition. Expert Rev Clin Immu. 2011; 7: 675–96.Google Scholar
Migale, R, MacIntyre, DA, Cacciatore, S, Lee, YS, Hagberg, H, Herbert, BR, et al. Modeling hormonal and inflammatory contributions to preterm and term labor using uterine temporal transcriptomics. BMC Med. 2016; 14: 86.Google Scholar
Kelly, RW. Pregnancy maintenance and parturition: the role of prostaglandin in manipulating the immune and inflammatory response. Endocr Rev. 1994; 15: 684706.Google Scholar
Aguilar, HN, Mitchell, BF. Physiological pathways and molecular mechanisms regulating uterine contractility. Hum Reprod Update. 2010; 16: 725–44.Google Scholar
Arulkumaran, S, Kandola, MK, Hoffman, B, Hanyaloglu, AC, Johnson, MR, Bennett, PR. The roles of prostaglandin EP 1 and 3 receptors in the control of human myometrial contractility. J Clin Endocrinol Metab. 2012; 97: 489–98.Google Scholar
Kandola, MK, Sykes, L, Lee, YS, Johnson, MR, Hanyaloglu, AC, Bennett, PR. EP2 receptor activates dual G protein signaling pathways that mediate contrasting proinflammatory and relaxatory responses in term pregnant human myometrium. Endocrinology. 2014; 155: 605–17.Google Scholar
Reinebrant, HE, Pileggi-Castro, C, Romero, CL, Dos Santos, RA, Kumar, S, Souza, JP, et al. Cyclo-oxygenase (COX) inhibitors for treating preterm labour. Cochrane Database Syst Rev. 2015; 6: CD001992.Google Scholar
Kim, SH, Bennett, PR, Terzidou, V. Advances in the role of oxytocin receptors in human parturition. Mol Cell Endocrinol. 2017; 449: 5663.Google Scholar
Kim, SH, Pohl, O, Chollet, A, Gotteland, JP, Fairhurst, AD, Bennett, PR, et al. Differential effects of oxytocin receptor antagonists, atosiban and nolasiban, on oxytocin receptor-mediated signaling in human amnion and myometrium. Mol Pharmacol. 2017; 91: 403–15.Google Scholar
Lager, S, de Goffau, MC, Sovio, U, Peacock, SJ, Parkhill, J, Charnock-Jones, DS, et al. Detecting eukaryotic microbiota with single-cell sensitivity in human tissue. Microbiome. 2018; 6: 151.Google Scholar
Perez-Munoz, ME, Arrieta, MC, Ramer-Tait, AE, Walter, J. A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: implications for research on the pioneer infant microbiome. Microbiome. 2017; 5: 48.Google Scholar
Romero, R, Dey, SK, Fisher, SJ. Preterm labor: one syndrome, many causes. Science. 2014; 345: 760–5.Google Scholar
MacIntyre, DA, Sykes, L, Bennett, PR. The human female urogenital microbiome: complexity in normality. Emerg Top Life Sci. 2017; 1: 363–72.Google Scholar
Kindinger, LM, Bennett, PR, Lee, YS, Marchesi, JR, Smith, A, Cacciatore, S, et al. The interaction between vaginal microbiota, cervical length, and vaginal progesterone treatment for preterm birth risk. Microbiome. 2017; 5: 6.Google Scholar
Callahan, BJ, DiGiulio, DB, Goltsman, DSA, Sun, CL, Costello, EK, Jeganathan, P, et al. Replication and refinement of a vaginal microbial signature of preterm birth in two racially distinct cohorts of US women. Proc Natl Acad Sci U S A. 2017; 114: 9966–71.Google Scholar
Brown, RG, Marchesi, JR, Lee, YS, Smith, A, Lehne, B, Kindinger, LM, et al. Vaginal dysbiosis increases risk of preterm fetal membrane rupture, neonatal sepsis and is exacerbated by erythromycin. BMC Med. 2018; 16: 9.Google Scholar
Chandiramani, M, Seed, PT, Orsi, NM, Ekbote, UV, Bennett, PR, Shennan, AH, et al. Limited relationship between cervico-vaginal fluid cytokine profiles and cervical shortening in women at high risk of spontaneous preterm birth. PLoS ONE. 2012; 7: e52412.Google Scholar
Brown, RG, Chan, D, Terzidou, V, Lee, YS, Smith, A, Marchesi, JR, et al. Prospective observational study of vaginal microbiota pre- and post-rescue cervical cerclage. BJOG. 2019; 126: 916925.Google Scholar
Kyrgiou, M, Athanasiou, A, Kalliala, IEJ, Paraskevaidi, M, Mitra, A, Martin-Hirsch, PP, et al. Obstetric outcomes after conservative treatment for cervical intraepithelial lesions and early invasive disease. Cochrane Database Syst Rev. 2017; 11: CD012847.Google Scholar
Cook, JR, Chatfield, S, Chandiramani, M, Kindinger, L, Cacciatore, S, Sykes, L, et al. Cerclage position, cervical length and preterm delivery in women undergoing ultrasound indicated cervical cerclage: a retrospective cohort study. PLoS ONE. 2017; 12: e0178072.Google Scholar
Kindinger, LM, MacIntyre, DA, Lee, YS, Marchesi, JR, Smith, A, McDonald, JA, et al. Relationship between vaginal microbial dysbiosis, inflammation, and pregnancy outcomes in cervical cerclage. Sci Transl Med. 2016; 8: 350ra102.Google Scholar
Dunsworth, HM, Warrener, AG, Deacon, T, Ellison, PT, Pontzer, H. Metabolic hypothesis for human altriciality. Proc Natl Acad Sci U S A. 2012; 109: 15212–16.Google Scholar
Muglia, LJ, Katz, M. The enigma of spontaneous preterm birth. N Engl J Med. 2010; 362: 529–35.Google Scholar
Monangi, NK, Brockway, HM, House, M, Zhang, G, Muglia, LJ. The genetics of preterm birth: Progress and promise. Semin Perinatol. 2015; 39: 574–83.Google Scholar
Zhang, G, Feenstra, B, Bacelis, J, Liu, X, Muglia, LM, Juodakis, J, et al. Genetic Associations with Gestational Duration and Spontaneous Preterm Birth. N Engl J Med. 2017; 377: 1156–67.Google Scholar

References

World Health Organization. (2017). Preterm Birth. www.who.int/news-room/fact-sheets/detail/preterm-birthGoogle Scholar
Liu, L, Oza, S, Hogan, D, Chu, Y, Perin, J, Zhu, J, et al. Global, regional, and national causes of under-5 mortality in 2000-15: an updated systematic analysis with implications for the Sustainable Development Goals. Lancet. 2016; 388: 3027–35.Google Scholar
Rubens, CE, Sadovsky, Y, Muglia, L, Gravett, MG, Lackritz, E, Gravett, C. Prevention of preterm birth: Harnessing science to address the global epidemic. Sci Transl Med. 2014; 6: 262sr5.Google Scholar
Shapiro-Mendoza, CK, Lackritz, EM. Epidemiology of late and moderate preterm birth. Semin Fetal Neonatal Med. 2012; 17: 120–5.Google Scholar
Moster, D, Lie, RT, Markestad, T. Long-term medical and social consequences of preterm birth. N Engl J Med. 2008; 359: 262–73.Google Scholar
Murray, SR, Stock, SJ, Norman, JE. Long-term childhood outcomes after interventions for prevention and management of preterm birth. Semin Perinatol. 2017; 41: 519–27.Google Scholar
Boyle, AK, Rinaldi, SF, Norman, JE, Stock, SJ. Preterm birth: inflammation, fetal injury and treatment strategies. J Reprod Immunol. 2017; 119: 62–6.Google Scholar
Goldenberg, RL, Culhane, JF, Iams, JD, Romero, R. Epidemiology and causes of preterm birth. Lancet. 2008; 371: 7584.Google Scholar
Kemp, MW. Preterm birth, intrauterine infection, and fetal inflammation. Front Immunol. 2014; 5: 574.Google Scholar
Fuchs, F, Senat, M-V. Multiple gestations and preterm birth. Semin Fetal Neonatal Med. 2016; 21: 113–20.Google Scholar
Esplin, MS, O’Brien, E, Fraser, A, Kerber, RA, Clark, E, Simonsen, SE, et al. Estimating recurrence of spontaneous preterm delivery. Obstet Gynecol. 2008; 112: 516–23.Google Scholar
Dekker, GA, Lee, SY, North, RA, McCowan, LM, Simpson, NAB, Roberts, CT, et al. Risk factors for preterm birth in an international prospective cohort of nulliparous women. PLoS ONE. 2012; 7: e39154.Google Scholar
Mercer, BM, Goldenberg, RL, Moawad, AH, Meis, PJ, Iams, JD, Das, AF, et al. The preterm prediction study: effect of gestational age and cause of preterm birth on subsequent obstetric outcome. National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Am J Obstet Gynecol. 1999; 181: 1216–21.Google Scholar
Malouf, R, Redshaw, M. Specialist antenatal clinics for women at high risk of preterm birth: a systematic review of qualitative and quantitative research. BMC Pregnancy Childbirth. 2017; 17: 51.Google Scholar
Georgiou, HM, Di Quinzio, MKW, Permezel, M, Brennecke, SP. Predicting preterm labour: current status and future prospects. Dis Markers. 2015; 2015: 435014.Google Scholar
Honest, H, Bachmann, LM, Gupta, JK, Kleijnen, J, Khan, KS. Accuracy of cervicovaginal fetal fibronectin test in predicting risk of spontaneous preterm birth: systematic review. BMJ. 2002; 325: 301.Google Scholar
Abbott, DS, Hezelgrave, NL, Seed, PT, Norman, JE, David, AL, Bennett, PR, et al. Quantitative fetal fibronectin to predict preterm birth in asymptomatic women at high risk. Obstet Gynecol. 2015; 125: 1168–76.Google Scholar
Berghella, V, Baxter, JK, Hendrix, NW. Cervical assessment by ultrasound for preventing preterm delivery. Cochrane Database Syst Rev. 2013; 3: CD007235.Google Scholar
Iams, JD, Goldenberg, RL, Meis, PJ, Mercer, BM, Moawad, A, Das, A, et al. The length of the cervix and the risk of spontaneous premature delivery. N Engl J Med. 1996; 334: 567–73.Google Scholar
Berghella, V, Roman, A, Daskalakis, C, Ness, A, Baxter, JK. Gestational age at cervical length measurement and incidence of preterm birth. Obstet Gynecol. 2007; 110: 311–17.Google Scholar
Society for Maternal-Fetal Medicine, McIntosh, J, Feltovich, H, Berghella, V, Manuck, T. The role of routine cervical length screening in selected high- and low-risk women for preterm birth prevention. Am J Obstet Gynecol. 2016; 215: B2–7.Google Scholar
Alfirevic, Z, Allen-Coward, H, Molina, F, Vinuesa, CP, Nicolaides, K. Targeted therapy for threatened preterm labor based on sonographic measurement of the cervical length: a randomized controlled trial. Ultrasound Obstet Gynecol. 2007; 29: 4750.Google Scholar
Boots, AB, Sanchez-Ramos, L, Bowers, DM, Kaunitz, AM, Zamora, J, Schlattmann, P. The short- term prediction of preterm birth: a systematic review and diagnostic metaanalysis. Am J Obstet Gynecol. 2014; 210: 54. e1–54. e10.Google Scholar
Berghella, V, Hayes, E, Visintine, J, Baxter, JK. Fetal fibronectin testing for reducing the risk of preterm birth. Cochrane Database Syst Rev. 2008; 4: CD006843.Google Scholar
National Institute for Health and Care Excellence. (2015). Preterm Labour and Birth. https://www.nice.org.uk/guidance/ng25.Google Scholar
Sarri, G, Davies, M, Gholitabar, M, Norman, JE, Guideline Development Group. Preterm labour: summary of NICE guidance. BMJ. 2015; 351: h6283.Google Scholar
Watson, HA, Carter, J, Seed, PT, Tribe, RM, Shennan, AH. The QUiPP App: a safe alternative to a treat-all strategy for threatened preterm labor. Ultrasound Obstet Gynecol. 2017; 50: 342–6.Google Scholar
Palacio, M, Kühnert, M, Berger, R, Larios, CL, Marcellin, L. Meta-analysis of studies on biochemical marker tests for the diagnosis of premature rupture of membranes: comparison of performance indexes. BMC Pregnancy Childbirth. 2014; 14: 183.Google Scholar
Norwitz, ER, Robinson, JN, Challis, JRG. The Control of Labor. N Engl J Med. 1999; 341: 660–6.Google Scholar
Anderson, L, Martin, W, Higgins, C, Nelson, SM, Norman, JE. The Effect of Progesterone on Myometrial Contractility, Potassium Channels, and Tocolytic Efficacy. Reprod Sci. 2009; 16: 1052–61.Google Scholar
Kumar, D, Springel, E, Moore, RM, Mercer, BM, Philipson, E, Mansour, JM, et al. Progesterone inhibits in vitro fetal membrane weakening. Am J Obstet Gynecol. 2015; 213: 520. e1–520. e9.Google Scholar
Keirse, MJ. Progestogen administration in pregnancy may prevent preterm delivery. BJOG. 1990; 97: 149–54.Google Scholar
Dodd, JM, Jones, L, Flenady, V, Cincotta, R, Crowther, CA. Prenatal administration of progesterone for preventing preterm birth in women considered to be at risk of preterm birth. Cochrane Database Syst Rev. 2013; 7: CD004947.Google Scholar
Norman, JE, Marlow, N, Messow, C-M, Shennan, A, Bennett, PR, Thornton, S, et al. Vaginal progesterone prophylaxis for preterm birth (the OPPTIMUM study): a multicentre, randomised, double-blind trial. Lancet. 2016; 387: 2106–16.Google Scholar
Crowther, CA, Ashwood, P, McPhee, AJ, Flenady, V, Tran, T, Dodd, JM, et al. Vaginal progesterone pessaries for pregnant women with a previous preterm birth to prevent neonatal respiratory distress syndrome (the PROGRESS Study): a multicentre, randomised, placebo-controlled trial. PLOS Med. 2017; 14: e1002390.Google Scholar
FIGO Working Group On Best Practice In Maternal-Fetal Medicine, International Federation of Gynecology and Obstetrics. Best practice in maternal-fetal medicine. Int J Gynecol Obstet. 2015; 128: 80–2.Google Scholar
Northen, AT, Norman, GS, Anderson, K, Moseley, L, Divito, M, Cotroneo, M, et al. Follow-up of children exposed in utero to 17 alpha-hydroxyprogesterone caproate compared with placebo. Obstet Gynecol. 2007; 110: 865–72.Google Scholar
Willing, J, Wagner, CK. Exposure to the synthetic progestin, 17α-hydroxyprogesterone caproate during development impairs cognitive flexibility in adulthood. Endocrinology. 2016; 157: 7782.Google Scholar
Shirodkar, V. A new method of operative treatment for habitual abortion in the second trimester of pregnancy. Antiseptic. 1955; 52: 299300.Google Scholar
McDonald, IA. Suture of the cervix for inevitable miscarriage. J Obstet Gynaecol Br Emp. 1957; 64: 346–50.Google Scholar
Suhag, A, Saccone, G, Bisulli, M, Seligman, N, Berghella, V. Trends in cerclage use. Acta Obstet Gynecol Scand. 2015; 94: 1188–94.Google Scholar
American College of Obstetrics and Gynecologists. ACOG practice bulletin. Cervical insufficiency. Int J Gynaecol Obstet. 2004; 85: 81–9.Google Scholar
Alfirevic, Z, Stampalija, T, Medley, N. Cervical stitch (cerclage) for preventing preterm birth in singleton pregnancy. Cochrane Database Syst Rev. 2017; 6: CD008991.Google Scholar
Royal College of Obstetricians & Gynecologists. (2011). Cervical Cerclage.Green-top Guideline No. 60. https://www.rcog.org.uk/en/guidelines-research-services/guidelines/gtg60Google Scholar
Berghella, V, Rafael, TJ, Szychowski, JM, Rust, OA, Owen, J. Cerclage for short cervix on ultrasonography in women with singleton gestations and previous preterm birth: a meta-analysis. Obs Gynecol. 2011; 117: 663–71.Google Scholar
Berghella, V, Ciardulli, A, Rust, OA, To, M, Otsuki, K, Althuisius, S, et al. Cerclage for sonographic short cervix in singleton gestations without prior spontaneous preterm birth: systematic review and meta-analysis of randomized controlled trials using individual patient-level data. Ultrasound Obstet Gynecol. 2017; 50: 569–77.Google Scholar
Simcox, R, Seed, PT, Bennett, P, Teoh, TG, Poston, L, Shennan, AH. A randomized controlled trial of cervical scanning vs history to determine cerclage in women at high risk of preterm birth (CIRCLE trial). Am J Obstet Gynecol. 2009; 200: 623. e1–623. e6.Google Scholar
Sinkey, RG, Garcia, MR, Odibo, AO. Does adjunctive use of progesterone in women with cerclage improve prevention of preterm birth? J Matern Neonatal Med. 2018; 31: 202–8.Google Scholar
Ehsanipoor, RM, Seligman, NS, Saccone, G, Szymanski, LM, Wissinger, C, Werner, EF, et al. Physical Examination–indicated cerclage. Obstet Gynecol. 2015; 126: 125–35.Google Scholar
Benson, RC, Durfee, RB. Transabdominal cervico uterine cerclage during pregnancy for the treatment of cervical incompetency. Obstet Gynecol. 1965; 25: 145–55.Google Scholar
Shiber, L-D, Lang, T, Pasic, R. First trimester laparoscopic cerclage. J Minim Invasive Gynecol. 2015; 22: 715–16.Google Scholar
Menderes, G, Clark, M, Clark-Donat, L, Azodi, M. Robotic-assisted abdominal cerclage placement during pregnancy and its challenges. J Minim Invasive Gynecol. 2015; 22: 713–14.Google Scholar
Moawad, GN, Tyan, P, Bracke, T, Abi Khalil, ED, Vargas, V, Gimovsky, A, et al. Systematic review of transabdominal cerclage placed via laparoscopy for the prevention of preterm birth. J Minim Invasive Gynecol. 2018; 25: 277–86.Google Scholar
Arabin, B, Alfirevic, Z. Cervical pessaries for prevention of spontaneous preterm birth: past, present and future. Ultrasound Obstet Gynecol. 2013; 42: 390–9.Google Scholar
Society for Maternal-Fetal Medicine (SMFM) Publications Committee. The role of cervical pessary placement to prevent preterm birth in clinical practice. Am J Obstet Gynecol. 2017; 216: B8–10.Google Scholar
Goya, M, Pratcorona, L, Merced, C, Rodó, C, Valle, L, Romero, A, et al. Cervical pessary in pregnant women with a short cervix (PECEP): an open-label randomised controlled trial. Lancet. 2012; 379: 1800–6.Google Scholar
Hui, A, Lao, TT, Ting, Y, Leung, T. Cervical pessary in pregnant women with a short cervix. Lancet. 2012; 380: 887.Google Scholar
Liem, SMS, van Pampus, MG, Mol, BWJ, Bekedam, DJ. Cervical pessaries for the prevention of preterm birth: a systematic review. Obstet Gynecol Int. 2013; 2013: 576723.Google Scholar
Nicolaides, KH, Syngelaki, A, Poon, LC, Picciarelli, G, Tul, N, Zamprakou, A, et al. A Randomized trial of a cervical pessary to prevent preterm singleton birth. N Engl J Med. 2016; 374: 1044–52.Google Scholar
Alfirevic, Z, Owen, J, Carreras Moratonas, E, Sharp, AN, Szychowski, JM, Goya, M. Vaginal progesterone, cerclage or cervical pessary for preventing preterm birth in asymptomatic singleton pregnant women with a history of preterm birth and a sonographic short cervix. Ultrasound Obstet Gynecol. 2013; 41: 146–51.Google Scholar
Van 't Hooft, J, Cuijpers, C, van der Lee, JH, Liem, S, Schuit, E, Opmeer, BC, et al. Long-term effects of cervical pessary for preterm birth prevention in twin pregnancy with short cervix: a 3 years follow-up of the ProTwin trial. Am J Obstet Gynecol. 2016; 214 (Suppl. 1): S287.Google Scholar
Hor, K, Norman, JE. Strategies for prevention of preterm birth. Fetal Matern Med Rev. 2013; 24: 3169.Google Scholar
Gyetvai, K, Hannah, ME, Hodnett, ED, Ohlsson, A. Tocolytics for preterm labor: a systematic review. Obstet Gynecol. 1999; 94: 869–77.Google Scholar
Haas, DM, Caldwell, DM, Kirkpatrick, P, McIntosh, JJ, Welton, NJ. Tocolytic therapy for preterm delivery: systematic review and network meta-analysis. BMJ. 2012; 345: e6226.Google Scholar
van Vliet, E, Seinen, L, Roos, C, Schuit, E, Scheepers, H, Bloemenkamp, K, et al. Maintenance tocolysis with nifedipine in threatened preterm labour: 2-year follow up of the offspring in the APOSTEL II trial. BJOG. 2016; 123: 1107–14.Google Scholar
Kenyon, S, Boulvain, M, Neilson, JP. Antibiotics for preterm rupture of membranes. Cochrane Database Syst Rev. 2013; 12: CD001058.Google Scholar
Kenyon, SL, Taylor, DJ, Tarnow-Mordi, W, ORACLE Collaborative Group. Broad-spectrum antibiotics for preterm, prelabour rupture of fetal membranes: the ORACLE I randomised trial. ORACLE Collaborative Group. Lancet. 2001; 357: 979–88.Google Scholar
Kenyon, S, Pike, K, Jones, D, Brocklehurst, P, Marlow, N, Salt, A, et al. Childhood outcomes after prescription of antibiotics to pregnant women with spontaneous preterm labour: 7-year follow-up of the ORACLE II trial. Lancet. 2008; 372: 1319–27.Google Scholar
Flenady, V, Hawley, G, Stock, OM, Kenyon, S, Badawi, N. Prophylactic antibiotics for inhibiting preterm labour with intact membranes. Cochrane Database Syst Rev. 2013: 4: CD000246.Google Scholar
Kenyon, SL, Taylor, DJ, Tarnow-Mordi, W, ORACLE Collaborative Group. Broad-spectrum antibiotics for spontaneous preterm labour: the ORACLE II randomised trial. ORACLE Collaborative Group. Lancet. 2001; 357: 989–94.Google Scholar
Othman, M, Alfirevic, Z, Neilson, JP. Probiotics for preventing preterm labour. Cochrane Database Syst Rev. 2007; 1: CD005941.Google Scholar
Reid, G, Bruce, AW, Fraser, N, Heinemann, C, Owen, J, Henning, B. Oral probiotics can resolve urogenital infections. FEMS Immunol Med Microbiol. 2001; 30: 4952.Google Scholar
Englund-Ögge, L, Brantsæter, AL, Sengpiel, V, Haugen, M, Birgisdottir, BE, Myhre, R, et al. Maternal dietary patterns and preterm delivery: results from large prospective cohort study. BMJ. 2014; 348: g1446.Google Scholar
Sosa, CG, Althabe, F, Belizán, JM, Bergel, E. Bed rest in singleton pregnancies for preventing preterm birth. Cochrane Database Syst Rev. 2015; 1: CD003581.Google Scholar
Hagberg, H, Mallard, C, Ferriero, DM, Vannucci, SJ, Levison, SW, Vexler, ZS, et al. The role of inflammation in perinatal brain injury. Nat Rev Neurol. 2015; 11: 192208.Google Scholar
FineSmith, R, Roche, K, Yellin, P, Walsh, K, Shen, C, Zeglis, M, et al. Effect of magnesium sulfate on the development of cystic periventricular leukomalacia in preterm infants. Am J Perinatol. 1997; 14: 303–7.Google Scholar
Royal College of Obstetricians & Gynaecologists. (2011). Magnesium Sulphate to Prevent Cerebral Palsy Following Preterm Birth. Scientific Impact Paper No. 29. www.rcog.org.uk/globalassets/documents/guidelines/scientific-impact-papers/sip_29.pdfGoogle Scholar
American College of Obstetricians and Gynecologists. Committee Opinion No 652: Magnesium sulfate use in obstetrics. Obstet Gynecol. 2016; 127: e52–3.Google Scholar
Macdonald, RL, Curry, DJ, Aihara, Y, Zhang, ZD, Jahromi, BS, Yassari, R. Magnesium and experimental vasospasm. J Neurosurg. 2004; 100: 106–10.Google Scholar
Oddie, S, Tuffnell, DJ, McGuire, W. Antenatal magnesium sulfate: Neuro-protection for preterm infants. Arch Dis Child Fetal Neonatal Ed. 2015; 100: F553–7.Google Scholar
Hallak, M, Hotra, JW, Kupsky, WJ. Magnesium sulfate protection of fetal rat brain from severe maternal hypoxia. Obstet Gynecol. 2000; 96: 124–8.Google Scholar
World Health Organization. (2015). WHO Recommendations on Interventions to Improve Preterm Birth Outcomes. https://apps.who.int/iris/bitstream/handle/10665/183037/9789241508988_eng.pdf;jsessionid=EA58641EA182B8839EAFB90BF86428F2?sequence=1.Google Scholar
Asztalos, EV, Murphy, KE, Willan, AR, Matthews, SG, Ohlsson, A, Saigal, S, et al. Multiple courses of antenatal corticosteroids for preterm birth study. JAMA Pediatr. 2013; 167: 1102–10.Google Scholar
Crowther, CA, Haslam, RR, Hiller, JE, Doyle, LW, Robinson, JS, Australasian Collaborative Trial of Repeat Doses of Steroids (ACTORDS) Study Group. Neonatal respiratory distress syndrome after repeat exposure to antenatal corticosteroids: a randomised controlled trial. Lancet. 2006; 367: 1913–19.Google Scholar
Crowther, CA, Anderson, PJ, McKinlay, CJD, Harding, JE, Ashwood, PJ, Haslam, RR, et al. Mid-childhood outcomes of repeat antenatal corticosteroids: a randomized controlled trial. Pediatrics. 2016; 138: e20160947.Google Scholar
Stutchfield, P, Whitaker, R, Russell, I, Antenatal Steroids for Term Elective Caesarean Section (ASTECS) Research Team. Antenatal betamethasone and incidence of neonatal respiratory distress after elective caesarean section: pragmatic randomised trial. BMJ. 2005; 331: 662.Google Scholar
Stutchfield, PR, Whitaker, R, Gliddon, AE, Hobson, L, Kotecha, S, Doull, IJM. Behavioural, educational and respiratory outcomes of antenatal betamethasone for term caesarean section (ASTECS trial). Arch Dis Child Fetal Neonatal Ed. 2013; 98: F195–200.Google Scholar

References

Hall, JG. Twinning. Lancet. 2003; 362: 735–43.Google Scholar
Lee, YM, Cleary-Goldman, J, Thaker, HM, Simpson, LL. Antenatal sonographic prediction of twin chorionicity. Am J Obstet Gynecol. 2006; 195: 863–7.Google Scholar
Kulkarni, AD, Jamieson, DJ, Jones, HW Jr., Kissin, DM, Gallo, MF, Macaluso, M, et al. Fertility treatments and multiple births in the United States. New Engl J Med. 2013; 369: 2218–25.Google Scholar
Martin, JA, Hamilton, BE, Osterman, MJK, Driscoll, AK, Drake, P. Births: Final Data for 2016. Natl Vital Stat Rep. 2018; 67: 155.Google Scholar
Ventura, SJ, Martin, JA, Curtin, SC, Mathews, TJ, Park, MM. Births: Final Data for 1998. Natl Vital Stat Rep. 2000; 48: 1100.Google Scholar
Heino, A, Gissler, M, Hindori-Mohangoo, AD, Blondel, B, Klungsoyr, K, Verdenik, I, et al. Variations in multiple birth rates and impact on perinatal outcomes in Europe. PLoS ONE. 2016; 11: e0149252.Google Scholar
Bodeau-Livinec, F, Zeitlin, J, Blondel, B, Arnaud, C, Fresson, J, Burguet, A, et al. Do very preterm twins and singletons differ in their neurodevelopment at 5 years of age? Arch Dis Child Fetal Neonatal Ed. 2013; 98: F480–7.Google Scholar
Donovan, EF, Ehrenkranz, RA, Shankaran, S, Stevenson, DK, Wright, LL, Younes, N, et al. Outcomes of very low birth weight twins cared for in the National Institute of Child Health and Human Development Neonatal Research Network’s intensive care units. Am J Obstet Gynecol. 1998; 179: 742–9.Google Scholar
Smith, GC, Pell, JP, Dobbie, R. Birth order, gestational age, and risk of delivery related perinatal death in twins: retrospective cohort study. BMJ. 2002; 325: 1004.Google Scholar
Luo, ZC, Ouyang, F, Zhang, J, Klebanoff, M. Perinatal mortality in second- vs firstborn twins: a matter of birth size or birth order? Am J Obstet Gynecol. 2014; 211: 153. e1–8.Google Scholar
Goldenberg, RL, Culhane, JF, Iams, JD, Romero, R. Epidemiology and causes of preterm birth. Lancet. 2008; 371: 7584.Google Scholar
James, S, Gil, KM, Myers, NA, Stewart, J. Effect of parity on gestational age at delivery in multiple gestation pregnancies. J Perinatol. 2009; 29: 1319.Google Scholar
Schaaf, JM, Mol, BW, Abu-Hanna, A, Ravelli, AC. Trends in preterm birth: singleton and multiple pregnancies in the Netherlands, 2000–2007. BJOG. 2011; 118: 1196–204.Google Scholar
Blondel, B, Kogan, MD, Alexander, GR, Dattani, N, Kramer, MS, Macfarlane, A, et al. The impact of the increasing number of multiple births on the rates of preterm birth and low birthweight: an international study. Am J Public Health. 2002; 92: 1323–30.Google Scholar
To, MS, Fonseca, EB, Molina, FS, Cacho, AM, Nicolaides, KH. Maternal characteristics and cervical length in the prediction of spontaneous early preterm delivery in twins. Am J Obstet Gynecol. 2006; 194: 1360–5.Google Scholar
Kahn, B, Lumey, LH, Zybert, PA, Lorenz, JM, Cleary-Goldman, J, D’Alton, ME, et al. Prospective risk of fetal death in singleton, twin, and triplet gestations: implications for practice. Obstet Gynecol. 2003; 102: 685–92.Google Scholar
Breathnach, FM, McAuliffe, FM, Geary, M, Daly, S, Higgins, JR, Dornan, J, et al. Optimum timing for planned delivery of uncomplicated monochorionic and dichorionic twin pregnancies. Obstet Gynecol. 2012; 119: 50–9.Google Scholar
Sullivan, AE, Hopkins, PN, Weng, HY, Henry, E, Lo, JO, Varner, MW, et al. Delivery of monochorionic twins in the absence of complications: analysis of neonatal outcomes and costs. Am J Obstet Gynecol. 2012; 206: 257. e1–7.Google Scholar
Burgess, JL, Unal, ER, Nietert, PJ, Newman, RB. Risk of late-preterm stillbirth and neonatal morbidity for monochorionic and dichorionic twins. Am J Obstet Gynecol. 2014; 210: 578. e1–9.Google Scholar
Cheong-See, F, Schuit, E, Arroyo-Manzano, D, Khalil, A, Barrett, J, Joseph, KS, et al. Prospective risk of stillbirth and neonatal complications in twin pregnancies: systematic review and meta-analysis. BMJ. 2016; 354: i4353.Google Scholar
Ortibus, E, Lopriore, E, Deprest, J, Vandenbussche, FP, Walther, FJ, Diemert, A, et al. The pregnancy and long-term neurodevelopmental outcome of monochorionic diamniotic twin gestations: a multicenter prospective cohort study from the first trimester onward. Am J Obstet Gynecol. 2009; 200: 494. e1–8.Google Scholar
Barigye, O, Pasquini, L, Galea, P, Chambers, H, Chappell, L, Fisk, NM. High risk of unexpected late fetal death in monochorionic twins despite intensive ultrasound surveillance: a cohort study. PLoS Med. 2005; 2: e172.Google Scholar
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
Ou, CW, Orsino, A, Lye, SJ. Expression of connexin-43 and connexin-26 in the rat myometrium during pregnancy and labor is differentially regulated by mechanical and hormonal signals. Endocrinology. 1997; 138: 5398–407.Google Scholar
Lyall, F, Lye, S, Teoh, T, Cousins, F, Milligan, G, Robson, S. Expression of Gsalpha, connexin-43, connexin-26, and EP1, 3, and 4 receptors in myometrium of prelabor singleton versus multiple gestations and the effects of mechanical stretch and steroids on Gsalpha. J Soc Gynecol Investig. 2002; 9: 299307.Google Scholar
Emery, SP, Bahtiyar, MO, Dashe, JS, Wilkins-Haug, LE, Johnson, A, Paek, BW, et al. The North American Fetal Therapy Network Consensus Statement: prenatal management of uncomplicated monochorionic gestations. Obstet Gynecol. 2015; 125: 1236–43.Google Scholar
Committee on Practice Bulletins—Obstetrics, Society for Maternal–Fetal Medicine. Practice Bulletin No. 169: Multifetal Gestations: Twin, Triplet, and Higher-Order Multifetal Pregnancies. Obstet Gynecol. 2016; 128: e131–46.Google Scholar
Cleary-Goldman, J, D’Alton, ME. Uncomplicated monochorionic diamniotic twins and the timing of delivery. PLoS Med. 2005; 2: e180.Google Scholar
Simoes, T, Amaral, N, Lerman, R, Ribeiro, F, Dias, E, Blickstein, I. Prospective risk of intrauterine death of monochorionic-diamniotic twins. Am J Obstet Gynecol. 2006; 195: 134–9.Google Scholar
Beasley, E, Megerian, G, Gerson, A, Roberts, NS. Monoamniotic twins: case series and proposal for antenatal management. Obstet Gynecol. 1999; 93: 130–4.Google Scholar
Rodis, JF, McIlveen, PF, Egan, JF, Borgida, AF, Turner, GW, Campbell, WA. Monoamniotic twins: improved perinatal survival with accurate prenatal diagnosis and antenatal fetal surveillance. Am J Obstet Gynecol. 1997; 177: 1046–9.Google Scholar
Rolett, A, Kiely, JL. Maternal sociodemographic characteristics as risk factors for preterm birth in twins versus singletons. Paediatr Perinat Epidemiol. 2000; 14: 211–18.Google Scholar
Kazemier, BM, Buijs, PE, Mignini, L, Limpens, J, de Groot, CJ, Mol, BW. Impact of obstetric history on the risk of spontaneous preterm birth in singleton and multiple pregnancies: a systematic review. BJOG. 2014; 121: 1197–208; discussion 209.Google Scholar
McCarthy, FP, Khashan, AS, North, RA, Rahma, MB, Walker, JJ, Baker, PN, et al. Pregnancy loss managed by cervical dilatation and curettage increases the risk of spontaneous preterm birth. Hum Reprod. 2013; 28: 3197–206.Google Scholar
Lemmers, M, Verschoor, MA, Hooker, AB, Opmeer, BC, Limpens, J, Huirne, JA, et al. Dilatation and curettage increases the risk of subsequent preterm birth: a systematic review and meta-analysis. Hum Reprod. 2016; 31: 3445.Google Scholar
Goldenberg, RL, Iams, JD, Miodovnik, M, Van Dorsten, JP, Thurnau, G, Bottoms, S, et al. The preterm prediction study: Risk factors in twin gestations. Am J Obstet Gynecol. 1996; 175: 1047–53.Google Scholar
Conde-Agudelo, A, Romero, R, Hassan, SS, Yeo, L. Transvaginal sonographic cervical length for the prediction of spontaneous preterm birth in twin pregnancies: a systematic review and meta-analysis. Am J Obstet Gynecol. 2010; 203: 128. e1–12.Google Scholar
Conde-Agudelo, A, Romero, R. Cervicovaginal fetal fibronectin for the prediction of spontaneous preterm birth in multiple pregnancies: a systematic review and meta-analysis. J Matern Fetal Neonatal Med. 2010; 23: 1365–76.Google Scholar
da Silva Lopes, K, Takemoto, Y, Ota, E, Tanigaki, S, Mori, R. Bed rest with and without hospitalisation in multiple pregnancy for improving perinatal outcomes. Cochrane Database Syst Rev. 2017; 3: CD012031.Google Scholar
Urquhart, C, Currell, R, Harlow, F, Callow, L. Home uterine monitoring for detecting preterm labour. Cochrane Database Syst Rev. 2017; 2: CD006172.Google Scholar
Ellings, JM, Newman, RB, Hulsey, TC, Bivins, HA Jr., Keenan, A. Reduction in very low birth weight deliveries and perinatal mortality in a specialized, multidisciplinary twin clinic. Obstet Gynecol. 1993; 81: 387–91.Google Scholar
Henry, A, Lees, N, Bein, KJ, Hall, B, Lim, V, Chen, KQ, et al. Pregnancy outcomes before and after institution of a specialised twins clinic: a retrospective cohort study. BMC Pregnancy Childbirth. 2015; 15: 217.Google Scholar
Rafael, TJ, Berghella, V, Alfirevic, Z. Cervical stitch (cerclage) for preventing preterm birth in multiple pregnancy. Cochrane Database Syst Rev. 2014; 9: CD009166.Google Scholar
Jarde, A, Lutsiv, O, Park, CK, Barrett, J, Beyene, J, Saito, S, et al. Preterm birth prevention in twin pregnancies with progesterone, pessary, or cerclage: a systematic review and meta-analysis. BJOG. 2017; 124: 1163–73.Google Scholar
Saccone, G, Rust, O, Althuisius, S, Roman, A, Berghella, V. Cerclage for short cervix in twin pregnancies: systematic review and meta-analysis of randomized trials using individual patient-level data. Acta Obstet Gynecol Scand. 2015; 94: 352–8.Google Scholar
Liem, S, Schuit, E, Hegeman, M, Bais, J, de Boer, K, Bloemenkamp, K, et al. Cervical pessaries for prevention of preterm birth in women with a multiple pregnancy (ProTWIN): a multicentre, open-label randomised controlled trial. Lancet. 2013; 382: 1341–9.Google Scholar
Goya, M, de la Calle, M, Pratcorona, L, Merced, C, Rodo, C, Munoz, B, et al. Cervical pessary to prevent preterm birth in women with twin gestation and sonographic short cervix: a multicenter randomized controlled trial (PECEP-Twins). Am J Obstet Gynecol. 2016; 214: 145–52.Google Scholar
Nicolaides, KH, Syngelaki, A, Poon, LC, de Paco Matallana, C, Plasencia, W, Molina, FS, et al. Cervical pessary placement for prevention of preterm birth in unselected twin pregnancies: a randomized controlled trial. Am J Obstet Gynecol. 2016; 214: 3. e1–9.Google Scholar
Murthy, YS, Arronet, GH, Parekh, MC. Luteal phase inadequacy: its significance in infertility. Obstet Gynecol. 1970; 36: 758–61.Google Scholar
Romero, R, Conde-Agudelo, A, El-Refaie, W, Rode, L, Brizot, ML, Cetingoz, E, et al. Vaginal progesterone decreases preterm birth and neonatal morbidity and mortality in women with a twin gestation and a short cervix: an updated meta-analysis of individual patient data. Ultrasound Obstet Gynecol. 2017; 49: 303–14.Google Scholar
Yamasmit, W, Chaithongwongwatthana, S, Tolosa, JE, Limpongsanurak, S, Pereira, L, Lumbiganon, P. Prophylactic oral betamimetics for reducing preterm birth in women with a twin pregnancy. Cochrane Database Syst Rev. 2015; 12: CD004733.Google Scholar
Palas, D, Ehlinger, V, Alberge, C, Truffert, P, Kayem, G, Goffinet, F, et al. Efficacy of antenatal corticosteroids in preterm twins: the EPIPAGE-2 cohort study. BJOG. 2018; 125: 1164–70.Google Scholar

References

Blencowe, H, Cousens, S, Oestergaard, MZ, Chou, D, Moller, AB, Narwal, R, et al. National, regional, and worldwide estimates of preterm birth rates in the year 2010 with time trends since 1990 for selected countries: a systematic analysis and implications. Lancet. 2012; 379: 2162–72.Google Scholar
Liu, L, Johnson, HL, Cousens, S, Perin, J, Scott, S, Lawn, JE, et al. Global, regional, and national causes of child mortality: an updated systematic analysis for 2010 with time trends since 2000. Lancet. 2012; 379: 2151–61.Google Scholar
Mwaniki, MK, Atieno, M, Lawn, JE, Newton, CR. Long-term neurodevelopmental outcomes after intrauterine and neonatal insults: a systematic review. Lancet. 2012; 379: 445–52.Google Scholar
Moore, T, Hennessy, EM, Myles, J, Johnson, SJ, Draper, ES, Costeloe, KL, et al. Neurological and developmental outcome in extremely preterm children born in England in 1995 and 2006: the EPICure studies. BMJ. 2012; 345: e7961.Google Scholar
Shepherd, E, Salam, RA, Middleton, P, Makrides, M, McIntyre, S, Badawi, N, et al. Antenatal and intrapartum interventions for preventing cerebral palsy: an overview of Cochrane systematic reviews. Cochrane Database Syst Rev. 2017; 8: CD012077.Google Scholar
Villar, J, Abalos, E, Carroli, G, Giordano, D, Wojdyla, D, Piaggio, G, et al. Heterogeneity of perinatal outcomes in the preterm delivery syndrome. Obstet Gynecol. 2004; 104: 7887.Google Scholar
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
Goldenberg, RL, Gravett, MG, Iams, J, Papageorghiou, AT, Waller, SA, Kramer, M, et al. The preterm birth syndrome: issues to consider in creating a classification system. Am J Obstet Gynecol. 2012; 206: 113–18.Google Scholar
Barros, FC, Papageorghiou, AT, Victora, CG, Noble, JA, Pang, R, Iams, J, et al. The distribution of clinical phenotypes of preterm birth syndrome: implications for prevention. JAMA Pediatr. 2015; 169: 220–9.Google Scholar
Menon, R. Spontaneous preterm birth, a clinical dilemma: etiologic, pathophysiologic and genetic heterogeneities and racial disparity. Acta Obstet Gynecol Scand. 2008; 87: 590600.Google Scholar
Blondel, B, Macfarlane, A, Gissler, M, Breart, G, Zeitlin, J, Group, PS. Preterm birth and multiple pregnancy in European countries participating in the PERISTAT project. BJOG. 2006; 113: 528–35.Google Scholar
Lee, SE, Romero, R, Park, CW, Jun, JK, Yoon, BH. The frequency and significance of intraamniotic inflammation in patients with cervical insufficiency. Am J Obstet Gynecol. 2008; 198: 633. e1–8.Google Scholar
Blencowe, H, Cousens, S, Chou, D, Oestergaard, M, Say, L, Moller, AB, et al. Born too soon: the global epidemiology of 15 million preterm births. Reprod Health. 2013; 10 (Suppl. 1): S2.Google Scholar
Gyamfi Bannerman, C. Late preterm birth: can be reduced. Am J Obstet Gynecol. 2011; 204: 459–60.Google Scholar
Newnham, JP, White, SW, Meharry, S, Lee, HS, Pedretti, MK, Arrese, CA, et al. Reducing preterm birth by a statewide multifaceted program: an implementation study. Am J Obstet Gynecol. 2017; 216: 434–42.Google Scholar
Bierstone, D, Wagenaar, N, Gano, DL, Guo, T, Georgio, G, Groenendaal, F, et al. Association of Histologic Chorioamnionitis With Perinatal Brain Injury and Early Childhood Neurodevelopmental Outcomes Among Preterm Neonates. JAMA Pediatr. 2018; 172: 534–41.Google Scholar
Woodward, LJ, Anderson, PJ, Austin, NC, Howard, K, Inder, TE. Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. New Engl J Med. 2006; 355: 685–94.Google Scholar
Behrman, R, Stith Butler, A (eds.). Preterm Birth: Causes, Consequences, and Prevention. Washington, DC: National Academies Press, 2007.Google Scholar
Rosenbaum, P, Paneth, N, Leviton, A, Goldstein, M, Bax, M, Damiano, D, et al. A report: the definition and classification of cerebral palsy April 2006. Dev Med Child Neurol Suppl. 2007; 109: 814.Google Scholar
Graham, HK, Rosenbaum, P, Paneth, N, Dan, B, Lin, JP, Damiano, DL, et al. Cerebral palsy. Nat Rev Dis Primers. 2016; 2: 15082.Google Scholar
MacLennan, AH, Thompson, SC, Gecz, J. Cerebral palsy: causes, pathways, and the role of genetic variants. Am J Obstet Gynecol. 2015; 213: 779–88.Google Scholar
Himpens, E, Van den Broeck, C, Oostra, A, Calders, P, Vanhaesebrouck, P. Prevalence, type, distribution, and severity of cerebral palsy in relation to gestational age: a meta-analytic review. Dev Med Child Neurol. 2008; 50: 334–40.Google Scholar
Colver, A, Fairhurst, C, Pharoah, PO. Cerebral palsy. Lancet. 2014; 383: 1240–9.Google Scholar
Bos, AF, Van Braeckel, KN, Hitzert, MM, Tanis, JC, Roze, E. Development of fine motor skills in preterm infants. Dev Med Child Neurol. 2013; 55 (Suppl. 4): 14.Google Scholar
Sommer, C, Urlesberger, B, Maurer-Fellbaum, U, Kutschera, J, Muller, W. Neurodevelopmental outcome at 2 years in 23 to 26 weeks old gestation infants. Klin Padiatr. 2007; 219: 23–9.Google Scholar
Linsell, L, Malouf, R, Morris, J, Kurinczuk, JJ, Marlow, N. Prognostic Factors for Poor Cognitive Development in Children Born Very Preterm or With Very Low Birth Weight: A Systematic Review. JAMA Pediatr. 2015;169: 1162–72.Google Scholar
Salmaso, N, Jablonska, B, Scafidi, J, Vaccarino, FM, Gallo, V. Neurobiology of premature brain injury. Nat Neurosci. 2014; 17: 341–6.Google Scholar
Nijman, TA, van Vliet, EO, Koullali, B, Mol, BW, Oudijk, MA. Antepartum and intrapartum interventions to prevent preterm birth and its sequelae. Semin Fetal Neonatal Med. 2016; 21: 121–8.Google Scholar
Rolnik, DL, Wright, D, Poon, LC, O’Gorman, N, Syngelaki, A, de Paco Matallana, C, et al. Aspirin versus Placebo in Pregnancies at High Risk for Preterm Preeclampsia. New Engl J Med. 2017; 377: 613–22.Google Scholar
Stock, SJ, Ismail, KM. Which intervention reduces the risk of preterm birth in women with risk factors? BMJ. 2016; 355: i5206.Google Scholar
McNamara, HC, Wood, R, Chalmers, J, Marlow, N, Norrie, J, MacLennan, G, et al. STOPPIT Baby Follow-up Study: the effect of prophylactic progesterone in twin pregnancy on childhood outcome. PLoS ONE. 2015; 10: e0122341.Google Scholar
Dodd, JM, Jones, L, Flenady, V, Cincotta, R, Crowther, CA. Prenatal administration of progesterone for preventing preterm birth in women considered to be at risk of preterm birth. Cochrane Database Syst Rev. 2013; 7: CD004947.Google Scholar
Cespedes Rubio, AE, Perez-Alvarez, MJ, Lapuente Chala, C, Wandosell, F. Sex steroid hormones as neuroprotective elements in ischemia models. J Endocrinol. 2018; 237: R65–81.Google Scholar
Flenady, V, Hawley, G, Stock, OM, Kenyon, S, Badawi, N. Prophylactic antibiotics for inhibiting preterm labour with intact membranes. Cochrane Database Syst Rev. 2013; 12: CD000246.Google Scholar
Neilson, JP, West, HM, Dowswell, T. Betamimetics for inhibiting preterm labour. Cochrane Database Syst Rev. 2014; 2: CD004352.Google Scholar
Haas, DM, Caldwell, DM, Kirkpatrick, P, McIntosh, JJ, Welton, NJ. Tocolytic therapy for preterm delivery: systematic review and network meta-analysis. BMJ. 2012; 345: e6226.Google Scholar
Stock, SJ, Bricker, L, Norman, JE, West, HM. Immediate versus deferred delivery of the preterm baby with suspected fetal compromise for improving outcomes. Cochrane Database Syst Rev. 2016; 7: CD008968.Google Scholar
Lees, CC, Marlow, N, van Wassenaer-Leemhuis, A, Arabin, B, Bilardo, CM, Brezinka, C, et al. 2 year neurodevelopmental and intermediate perinatal outcomes in infants with very preterm fetal growth restriction (TRUFFLE): a randomised trial. Lancet. 2015; 385: 2162–72.Google Scholar
Ting, JY, Kingdom, JC, Shah, PS. Antenatal glucocorticoids, magnesium sulfate, and mode of birth in preterm fetal small for gestational age. Am J Obstet Gynecol. 2018; 218: S818–28.Google Scholar
Grabovac, M, Karim, JN, Isayama, T, Liyanage, SK, McDonald, SD. What is the safest mode of birth for extremely preterm breech singleton infants who are actively resuscitated? A systematic review and meta-analyses. BJOG. 2018; 125: 652–63.Google Scholar
Liggins, GC, Howie, RN. A controlled trial of antepartum glucocorticoid treatment for prevention of the respiratory distress syndrome in premature infants. Pediatrics. 1972; 50: 515–25.Google Scholar
Roberts, D, Brown, J, Medley, N, Dalziel, SR. Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev. 2017; 3: CD004454.Google Scholar
Brownfoot, FC, Gagliardi, DI, Bain, E, Middleton, P, Crowther, CA. Different corticosteroids and regimens for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev. 2013; 8: CD006764.Google Scholar
Grether, J, Hirtz, D, McNellis, D, Nelson, K, Rouse, DJ. Tocolytic magnesium sulphate and paediatric mortality. Lancet. 1998; 351: 292; author reply 3.Google Scholar
Mittendorf, R, Covert, R, Boman, J, Khoshnood, B, Lee, KS, Siegler, M. Is tocolytic magnesium sulphate associated with increased total paediatric mortality? Lancet. 1997; 350: 1517–18.Google Scholar
Magpie Trial Follow-Up Study Collaborative Group. The Magpie Trial: a randomised trial comparing magnesium sulphate with placebo for pre-eclampsia. Outcome for children at 18 months. BJOG. 2007; 114: 289–99.Google Scholar
Crowther, CA, Hiller, JE, Doyle, LW, Haslam, RR, Australasian Collaborative Trial of Magnesium Sulphate Collaborative Group. Effect of magnesium sulfate given for neuroprotection before preterm birth: a randomized controlled trial. JAMA. 2003; 290: 2669–76.Google Scholar
Rouse, DJ, Hirtz, DG, Thom, E, Varner, MW, Spong, CY, Mercer, BM, et al. A randomized, controlled trial of magnesium sulfate for the prevention of cerebral palsy. New Engl J Med. 2008; 359: 895905.Google Scholar
Marret, S, Marpeau, L, Zupan-Simunek, V, Eurin, D, Leveque, C, Hellot, MF, et al. Magnesium sulphate given before very-preterm birth to protect infant brain: the randomised controlled PREMAG trial. BJOG. 2007; 114: 310–18.Google Scholar
Doyle, LW, Crowther, CA, Middleton, P, Marret, S, Rouse, D. Magnesium sulphate for women at risk of preterm birth for neuroprotection of the fetus. Cochrane Database Syst Rev. 2009; 1: CD004661.Google Scholar
Australian National Health and Medical Research Council. (2010). Antenatal magnesium sulphate prior to preterm birth for neuroprotection of the fetus, infant and child. National Clinical Practice Guidelines. https://www.clinicalguidelines.gov.au/register/antenatal-magnesium-sulphate-prior-preterm-birth-neuroprotection-fetus-infant-and-child.Google Scholar
National Institute for Health and Care Excellence. (2015). Preterm labour and Birth. NICE Guideline NG25. https://www.nice.org.uk/guidance/ng25.Google Scholar
Magee, L, Sawchuck, D, Synnes, A, von Dadelszen, P, Magnesium Sulphate For Fetal Neuroprotection Consensus Committee, Maternal Fetal Medicine Committee. SOGC Clinical Practice Guideline: Magnesium sulphate for fetal neuroprotection. J Obstet Gynaecol Can. 2011; 33: 516–29.Google Scholar
American College of Obstetricians and Gynecologists Committee on Obstetric Practice; Society for Maternal-Fetal Medicine. Committee Opinion No. 455: Magnesium Sulfate before anticipated preterm birth for neuroprotection. Obstet Gynecol. 2016; 115: 669–71.Google Scholar
Crowther, CA, Middleton, PF, Voysey, M, Askie, L, Duley, L, Pryde, PG, et al. Assessing the neuroprotective benefits for babies of antenatal magnesium sulphate: an individual participant data meta-analysis. PLoS Med. 2017; 14: e1002398.Google Scholar
Chang, E. Preterm birth and the role of neuroprotection. BMJ. 2015; 350: g6661.Google Scholar
Colella, M, Biran, V, Baud, O. Melatonin and the newborn brain. Early Hum Dev. 2016; 102: 13.Google Scholar
Dickinson, H, Bain, E, Wilkinson, D, Middleton, P, Crowther, CA, Walker, DW. Creatine for women in pregnancy for neuroprotection of the fetus. Cochrane Database Syst Rev. 2014; 12: CD010846.Google Scholar
Jenkins, DD, Wiest, DB, Mulvihill, DM, Hlavacek, AM, Majstoravich, SJ, Brown, TR, et al. Fetal and neonatal effects of N-acetylcysteine when used for neuroprotection in maternal chorioamnionitis. J Pediatr. 2016; 168: 6776. e6.Google Scholar
Ranchhod, SM, Gunn, KC, Fowke, TM, Davidson, JO, Lear, CA, Bai, J, et al. Potential neuroprotective strategies for perinatal infection and inflammation. Int J Dev Neurosci. 2015; 45: 4454.Google Scholar
Paton, MCB, McDonald, CA, Allison, BJ, Fahey, MC, Jenkin, G, Miller, SL. Perinatal brain injury as a consequence of preterm birth and intrauterine inflammation: designing targeted stem cell therapies. Front Neurosci. 2017; 11: 200.Google Scholar
Costantine, MM, Weiner, SJ, Eunice Kennedy Shriver National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Effects of antenatal exposure to magnesium sulfate on neuroprotection and mortality in preterm infants: a meta-analysis. Obstet Gynecol. 2009; 114: 354–64.Google Scholar
Conde-Agudelo, A, Romero, R. Antenatal magnesium sulfate for the prevention of cerebral palsy in preterm infants less than 34 weeks’ gestation: a systematic review and meta-analysis. Am J Obstet Gynecol. 2009; 200: 595609.Google Scholar

References

Schatz, F. Klinische Beiträge zur Physiologie des Fetus. Berlin: Hirschwald, 1900.Google Scholar
Berghella, V, Kaufmann, M. Natural history of twin-twin transfusion syndrome. J Reprod Med. 2001; 46: 480–4.Google Scholar
Lewi, L, Jani, J, Blickstein, I, Huber, A, Gucciardo, L, Van Mieghem, T, et al. The outcome of monochorionic diamniotic twin gestations in the era of invasive fetal therapy: a prospective cohort study. Am J Obstet Gynecol. 2008; 199: 514. e1–8.Google Scholar
Akkermans, J, Peeters, S, Klumper, F, Lopriore, E, Middeldorp, J, Oepkes, D. Twenty-five years of fetoscopic laser coagulation in twin–twin transfusion syndrome: a systematic review. Fetal Diagn Ther. 2015; 38: 241–53.Google Scholar
Peeters, S, Devlieger, R, Middeldorp, J, DeKoninck, P, Deprest, J, Lopriore, E, et al. Fetal surgery in complicated monoamniotic pregnancies: case series and systematic review of the literature. Prenat Diagn. 2014; 34: 586–91.Google Scholar
Denbow, M, Welsh, A, Taylor, M, Blomley, M, Cosgrove, D, Fisk, N. Twin fetuses: intravascular microbubble US contrast agent administration – early experience. Radiology. 2000; 214: 724–8.Google Scholar
Gardiner, HM. Early changes in vascular dynamics in relation to twin-twin transfusion syndrome. Twin Res. 2001; 4: 371–7.Google Scholar
Charnock-Jones, DS, Kaufmann, P, Mayhew, T. Aspects of human fetoplacental vasculogenesis and angiogenesis. I. Molecular regulation. Placenta. 2004; 25: 103–13.Google Scholar
Mayhew, T, Charnock-Jones, D, Kaufmann, P. Aspects of human fetoplacental vasculogenesis and angiogenesis. III. Changes in complicated pregnancies. Placenta. 2004; 25: 127–39.Google Scholar
Bdolah, Y, Lam, C, Rajakumar, A, Shivalingappa, V, Mutter, W, Sachs, BP, et al. Twin pregnancy and the risk of preeclampsia: bigger placenta or relative ischemia? Am J Obstet Gynecol. 2008; 198: 428. e1–6.Google Scholar
Machin, G, Still, K, Lalani, T. Correlations of placental vascular anatomy and clinical outcomes in 69 monochorionic twin pregnancies. Am J Med Genet. 1996; 61: 229–36.Google Scholar
Lewi, L, Deprest, J, Hecher, K. The vascular anastomoses in monochorionic twin pregnancies and their clinical consequences. Am J Obstet Gynecol. 2013; 208: 1930.Google Scholar
Bajoria, R, Wigglesworth, J, Fisk, NM. Angioarchitecture of monochorionic placentas in relation to the twin-twin transfusion syndrome. Am J Obstet Gynecol. 1995; 172: 856–63.Google Scholar
Zhao, DP, de Villiers, SF, Slaghekke, F, Walther, FJ, Middeldorp, JM, Oepkes, D, et al. Prevalence, size, number and localization of vascular anastomoses in monochorionic placentas. Placenta. 2013; 34: 589–93.Google Scholar
Umur, A, Van Gemert, M, Nikkels, P, Ross, M. Monochorionic twins and twin-twin transfusion syndrome: the protective role of arterio-arterial anastomoses. Placenta. 2002; 23: 201–9.Google Scholar
de Villiers, S, Slaghekke, F, Middeldorp, J, Walther, F, Oepkes, D, Lopriore, E. Arterio-arterial vascular anastomoses in monochorionic placentas with and without twin-twin transfusion syndrome. Placenta. 2012; 33: 652–4.Google Scholar
Taylor, M, Denbow, M, Duncan, K, Overton, T, Fisk, N. Antenatal factors at diagnosis that predict outcome in twin-twin transfusion syndrome. Am J Obstet Gynecol. 2000; 183: 1023–8.Google Scholar
De Villiers, SF, Zhao, DP, Cohen, D, Van Zwet, EW, Duan, T, Oepkes, D, et al. Correlation between veno-venous anastomoses, TTTS and perinatal mortality in monochorionic twin pregnancies. Placenta. 2015; 36: 603–6.Google Scholar
Zhao, DP, Cohen, D, Middeldorp, JM, Klumper, FJ, Haak, MC, Oepkes, D, et al. The role of veno-venous anastomoses in twin-twin transfusion syndrome. Placenta. 2014; 5: 334–6.Google Scholar
Kalafat, E, Thilaganathan, B, Papageorghiou, A, Bhide, A, Khalil, A. The significance of placental cord insertion site in twin pregnancy. Ultrasound Obstet Gynecol. 2018; 52: 378–84.Google Scholar
Chmait, R, Kontopoulos, E, Korst, L, Llanes, A, Petisco, I, Quintero, R. Stage-based outcomes of 682 consecutive cases of twin-twin transfusion syndrome treated with laser surgery: the USFetus experience. Am J Obstet Gynecol. 2011; 204: 393. e1–6.Google Scholar
De Paepe, M, Shapiro, S, Greco, D, Luks, V, Abellar, R, Luks, C, et al. Placental markers of twin-to twin transfusion syndrome in diamniotic-monochorionic twins: a morphometric analysis of deep artery-to vein anastomoses. Placenta. 2010; 31: 269–76.Google Scholar
Fries, MH, Goldstein, RB, Kilpatrick, SJ, Golbus, MS, Callen, PW, Filly, RA. The role of velamentous cord insertion in the etiology of twin-twin transfusion syndrome. Obstet Gynecol. 1993; 81: 569–74.Google Scholar
Machin, GA. Velamentous cord insertion in monochorionic twin gestation: An added risk factor. J Reprod Med. 1997; 42: 785–9.Google Scholar
Lopriore, E, Sueters, M, Middeldorp, JM, Oepkes, D, Walther, FJ, Vandenbussche, FPHA. Velamentous cord insertion and unequal placental territories in monochorionic twins with and without twin-to-twin-transfusion syndrome. Am J Obstet Gynecol. 2007; 196: 159. e1–5.Google Scholar
Tammela, T, Enholm, B, Alitalo, K, Paavonen, K. The biology of vascular endothelial growth factors. Cardiovasc Res. 2005; 65: 550–63.Google Scholar
Zhang, E, Smith, S, Baker, P, Charnock-Jones, D. The regulation and localization of angiopoietin-1, -2, and their receptor Tie2 in normal and pathologic human placentae. Mol Med. 2001; 7: 624–35.Google Scholar
Maisonpierre, P, Suri, C, Jones, P, Bartunkova, S, Wiegand, S, Radziejewski, C, et al. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science. 1997; 277: 5560.Google Scholar
St-Jacques, S, Forte, M, Lye, S, Letarte, M. Localization of endoglin, a transforming growth factor-beta binding protein, and of CD44 and integrins in placenta during the first trimester of pregnancy. Biol Reprod. 1994; 51: 405–13.Google Scholar
Venkatesha, S, Toporsian, M, Lam, C, Hanai, J, Mammoto, T, Kim, Y, et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med. 2006; 12: 642–9.Google Scholar
Kaufmann, P, Mayhew, T, Charnock-Jones, D. Aspects of human fetoplacental vasculogenesis and angiogenesis. II. Changes during normal pregnancy. Placenta. 2004; 25: 114–26.Google Scholar
Burton, G, Charnock-Jones, D, Jauniaux, E. Regulation of vascular growth and function in the human placenta. Reproduction. 2009; 138: 895902.Google Scholar
Karumanchi, S, Bdolah, Y. Hypoxia and sFlt-1 in preeclampsia: the “chicken-and-egg” question. Endocrinology. 2004; 145: 4835–7.Google Scholar
Romero, R, Nien, JK, Espinoza, J, Todem, D, Fu, W, Chung, H, et al. A longitudinal study of angiogenic (placental growth factor) and anti-angiogenic (soluble endoglin and soluble VEGF receptor-1) factors in normal pregnancy and patients destined to develop preeclampsia and deliver a small-for-gestational-age neonate. J Matern Fetal Neonatal Med. 2008; 21: 923.Google Scholar
Levine, RJ, Maynard, SE, Qian, C, Lim, K-H, England, LJ, Yu, KF, et al. Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med. 2004; 350: 672–83.Google Scholar
Crispi, F, Domínguez, C, Llurba, E, Martín-Gallán, P, Cabero, L, Gratacós, E. Placental angiogenic growth factors and uterine artery Doppler findings for characterization of different subsets in preeclampsia and in isolated intrauterine growth restriction. Am J Obstet Gynecol. 2006; 195: 201–7.Google Scholar
Kumazaki, K, Nakayama, M, Suehara, N, Wada, Y. Expression of vascular endothelial growth factor, placental growth factor, and their receptors Flt-1 and KDR in human placenta under pathologic conditions. Hum Pathol. 2002; 33: 1069–77.Google Scholar
Kusanovic, JP, Romero, R, Espinoza, J, Nien, JK, Kim, CJ, Mittal, P, et al. Twin-to-twin transfusion syndrome: an antiangiogenic state? Am J Obstet Gynecol. 2008; 198: 382. e1–8.Google Scholar
Fox, CE, Lash, GE, Pretlove, SJ, Chan, BC, Holder, R, Kilby, MD. Maternal plasma and amniotic fluid angiogenic factors and their receptors in monochorionic twin pregnancies complicated by twin-to-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2010; 35: 695701.Google Scholar
Mackie, FL, Whittle, R, Morris, RK, Hyett, J, Riley, R, Kilby, MD. First trimester ultrasound measurements and maternal serum biomarkers as prognostic factors in monochorionic twins: a cohort study. Diagn Progn Res. 2019; 3: 9.Google Scholar
Galea, P, Barigye, O, Wee, L, Jain, V, Sullivan, M, Fisk, NM. The placenta contributes to activation of the renin angiotensin system in twin-twin transfusion syndrome. Placenta. 2008; 29: 734–42.Google Scholar
Barrea, C, Alkazaleh, F, Ryan, G, McCrindle, BW, Roberts, A, Bigras, JL, et al. Prenatal cardiovascular manifestations in the twin-to-twin transfusion syndrome recipients and the impact of therapeutic amnioreduction. Am J Obstet Gynecol. 2005; 192: 892902.Google Scholar
Habli, M, Michelfelder, E, Livingston, J, Harmon, J, Lim, F, Polzin, W, et al. Acute effects of selective fetoscopic laser photocoagulation on recipient cardiac function in twin-twin transfusion syndrome. Am J Obstet Gynecol. 2008; 199: 412. e1–6.Google Scholar
Tei, C, Ling, L, Hodge, D, Bailey, K, Oh, J, Rodeheffer, R, et al. New index of combined systolic and diastolic myocardial performance: a simple and reproducible measure of cardiac function—a study in normals and dilated. J Cardiol. 1995; 26: 357–66.Google Scholar
Rychik, J, Tian, Z, Bebbington, M, Xu, F, McCann, M, Mann, S, et al. The twin-twin transfusion syndrome: spectrum of cardiovascular abnormality and development of a cardiovascular score to assess severity of disease. Am J Obstet Gynecol. 2007; 197: 392. e1–8.Google Scholar
Van Mieghem, T, Klaritsch, P, Doné, E, Gucciardo, L, Lewi, P, Verhaeghe, J, et al. Assessment of fetal cardiac function before and after therapy for twin-to-twin transfusion syndrome. Am J Obstet Gynecol. 2009; 200: 400. e1–7.Google Scholar
Gapp-Born, E, Sananes, N, Weingertner, AS, Guerra, F, Kohler, M, Fritz, G, et al. Predictive value of cardiovascular parameters in twin-to-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2014; 44: 427–33.Google Scholar
Habli, M, Michelfelder, E, Cnota, J, Wall, D, Polzin, W, Lewis, D, et al. Prevalence and progression of recipient-twin cardiomyopathy in early stage twin-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2012; 39: 63–8.Google Scholar
Ortiz, J, Torres, X, Eixarch, E, Bennasar, M, Cruz-Lemini, M, Gómez, O, et al. Differential changes in myocardial performance index and its time intervals in donors and recipients of twin-to-twin transfusion syndrome before and after laser therapy. Fetal Diagn Ther. 2018; 44: 305–10.Google Scholar
Karatza, AA, Wolfenden, JL, Taylor, MJO, Wee, L, Fisk, NM, Gardiner, HM. Influence of twin-twin transfusion syndrome on fetal cardiovascular structure and function: Prospective case-control study of 136 monochorionic twin pregnancies. Heart. 2002; 88: 271–7.Google Scholar
Wohlmuth, C, Boudreaux, D, Moise, KJ Jr., Johnson, A, Papanna, R, Bebbington, M, et al. Cardiac pathophysiology in twin-twin transfusion syndrome: new insights into its evolution. Ultrasound Obstet Gynecol. 2017: 51: 341–8.Google Scholar
Gardiner, H, Matsui, H, Roughton, M, Greenwald, S, Diemert, A, Taylor, M, et al. Cardiac function in 10-year-old twins following different fetal therapies for twin-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2014; 43: 652–7.Google Scholar
Gardiner, H, Taylor, M, Karatza, A, Vanderheyden, T, Huber, A, Greenwald, S, et al. Twin-twin transfusion syndrome: the influence of intrauterine laser photocoagulation on arterial distensibility in childhood. Circulation. 2003; 107: 1906–11.Google Scholar
Gardiner, H, Barlas, A, Matsui, H, Diemert, A, Taylor, M, Preece, J, et al. Vascular programming in twins: the effects of chorionicity and fetal therapy for twin-to-twin transfusion syndrome. J Dev Orig Health Dis. 2012; 3: 182–9.Google Scholar
Mahieu-Caputo, D, Salomon, L, Le Bidois, J, Fermont, L, Brunhes, A, Jouvet, P, et al. Fetal hypertension: An insight into the pathogenesis of the twin-twin transfusion syndrome. Prenat Diagn. 2003; 23: 640–5.Google Scholar
Stagnati, V, Zanardini, C, Fichera, A, Pagani, G, Quintero, RA, Bellocco, R, et al. Early prediction of twin-to-twin transfusion syndrome: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2017; 49: 573–82.Google Scholar
Sebire, NJ, Souka, A, Skentou, H, Geerts, L, Nicolaides, KH. Early prediction of severe twin-to-twin transfusion syndrome. Hum Reprod. 2000; 15: 2008–10.Google Scholar
Dunn, WB, Shek, N, Fox, CE, Mackie, F, van Mieghem, T, Kilby, M. Non-targeted metabolomics in recipient amniotic fluid of MC twin pregnancies complicated by severe twin to twin transfusion syndrome (TTTS) and treated by fetoscopic laser ablation (FLC). BJOG. 2015; 122(Suppl. 8): 8.Google Scholar
Taylor, G, Peart, W, Porter, K, Zondek, L, Zondek, T. Concentration and molecular forms of active and inactive renin in human fetal kidney, amniotic fluid and adrenal gland: evidence for renin-angiotensin system hyperactivity in 2nd trimester of pregnancy. J Hypertens. 1986; 4: 121–9.Google Scholar
Paul, M, Poyan, M, Kreutz, R. Physiology of local renin-angiotensin systems. Physiol Rev. 2006; 86: 747803.Google Scholar
Kilby, M, Platt, C, Whittle, M, Oxley, J, Lindop, G. Renin-gene expression in fetal kidneys of pregnancies complicated by twin-twin transfusion syndrome. Pediatr Dev Pathol. 2001; 4: 175–9.Google Scholar
Wee, L, Sullivan, M, Humphries, K, Fisk, N. Longitudinal blood flow in shared (arteriovenous anastomoses) and non-shared cotyledons in monochorionic placentae. Placenta. 2007; 28: 516–22.Google Scholar
Mahieu-Caputo, D, Meulemans, A, Martinovic, J, Gubler, M, Delezoide, A, Muller, F, et al. Paradoxic activation of the renin-angiotensin system in twin-twin transfusion syndrome: an explanation for cardiovascular disturbances in the recipient. Pediatr Res. 2005; 58: 685–8.Google Scholar
Verbeek, L, Joemmanbaks, F, Quak, J, Sukhai, R, Middeldorp, J, Oepkes, D, et al. Renal function in neonates with twin-twin transfusion syndrome treated with or without fetoscopic laser surgery. Eur J Pediatr. 2017; 176: 1209–15.Google Scholar
Bajoria, R, Ward, S, Chatterjee, R. Natriuretic peptides in the pathogenesis of cardiac dysfunction in the recipient fetus of twin-twin transfusion syndrome. Am J Obstet Gynecol. 2002; 186: 121–7.Google Scholar
Hynynen, M, Khalil, R. The vascular endothelin system in hypertension – recent patents and discoveries. Recent Pat Cardiovasc Drug Discov. 2006; 1: 95108.Google Scholar
Bajoria, R, Sullivan, M, Fisk, NM. Endothelin concentrations in monochorionic twins with severe twin-twin transfusion syndrome. Hum Reprod. 1999; 14: 1614–18.Google Scholar
Bajoria, R, Ward, S, Chatterjee, R. Brain natriuretic peptide and endothelin-1 in the pathogenesis of polyhydramnios-oligohydramnios in monochorionic twins. Am J Obstet Gynecol. 2003; 189: 189–94.Google Scholar
Wilkins, M, Redondo, J, Brown, L. The natriuretic-peptide family. Lancet. 1997; 349: 1307–10.Google Scholar
Chen, H, Burnett, J. Natriuretic peptides in the pathophysiology of congestive heart failure. Curr Cardiol Rep. 2000; 2: 198205.Google Scholar
Butt, R, Laurent, G, Bishop, J. Mechanical load and polypeptide growth factors stimulate cardiac fibroblast activity. Ann N Y Acad Sci. 1995; 752: 387–93.Google Scholar
Cao, L, Gardner, D. Natriuretic peptides inhibit DNA synthesis in cardiac fibroblasts. Hypertension. 1995; 25: 227–34.Google Scholar
Omland, T, Aakvaag, A, Bonarjee, V, Caidahl, K, Lie, R, Nilsen, D, et al. Plasma brain natriuretic peptide as an indicator of left ventricular systolic function and long-term survival after acute myocardial infarction. Comparison with plasma atrial natriuretic peptide and N-terminal proatrial natriuretic peptide. Circulation. 1996; 93: 1963–9.Google Scholar
Bajoria, R, Ward, S, Sooranna, SR. Atrial natriuretic peptide mediated polyuria: pathogenesis of polyhydramnios in the recipient twin of twin-twin transfusion syndrome. Placenta. 2001; 22: 716–24.Google Scholar
Kiowski, W, Sütsch, G, Hunziker, P, Müller, P, Kim, J, Oechslin, E, et al. Evidence for endothelin-1-mediated vasoconstriction in severe chronic heart failure. Lancet. 1995; 346: 732–6.Google Scholar
Yorikane, R, Sakai, S, Miyauchi, T, Sakurai, T, Sugishita, Y, Goto, K. Increased production of endothelin-1 in the hypertrophied rat heart due to pressure overload. FEBS Lett. 1993; 332: 31–4.Google Scholar
Garcia, R, Lachance, D, Thibault, G. Positive inotropic action, natriuresis and atrial natriuretic factor release induced by endothelin in the conscious rat. J Hypertens. 1990; 8: 725–31.Google Scholar
Kanno, K, Hirata, Y, Tsujino, M, Imai, T, Shichiri, M, Ito, H, et al. Up-regulation of ETB receptor subtype mRNA by angiotensin II in rat cardiomyocytes. Biochem Biophys Res Commun. 1993; 194: 1282–7.Google Scholar

References

Salomon, LJ, Stirnemann, JJ, Bernard, JP, et al. Prenatal management of uncomplicated monochorionic biamniotic pregnancies. J Gynecol Obstet Biol Reprod. 2009; 38: S45–50.Google Scholar
Chalouhi, GE, Stirnemann, JJ, Salomon, LJ, et al. Specific complications of monochorionic twin pregnancies: twin-twin transfusion syndrome and twin reversed arterial perfusion sequence. Semin Fetal Neonatal Med. 2010; 15: 349–56.Google Scholar
Bebbington, M. Twin-to-twin transfusion syndrome: current understanding of pathophysiology, in-utero therapy and impact for future development. Semin Fetal Neonatal Med. 2010; 15: 1520.Google Scholar
Lopriore, E, Middeldorp, JM, Oepkes, D, et al. Twin anemia-polycythemia sequence in two monochorionic twin pairs without oligo-polyhydramnios sequence. Placenta. 2007; 28: 4751.Google Scholar
Slaghekke, F, Kist, WJ, Oepkes, D, et al. Twin anemia-polycythemia sequence: diagnostic criteria, classification, perinatal management and outcome. Fetal Diagn Ther. 2010; 27: 181–90.Google Scholar
Lewi, L, Gucciardo, L, Huber, A, et al. Clinical outcome and placental characteristics of monochorionic diamniotic twin pairs with early- and late-onset discordant growth. Am J Obstet Gynecol. 2008; 199: 511. e1–7.Google Scholar
Sebire, NJ, Snijders, RJ, Hughes, K, Sepulveda, W, Nicolaides, KH. The hidden mortality of monochorionic twin pregnancies. BJOG. 1997; 104: 1203–7.Google Scholar
Hack, K, Derks, J, Elias, S, et al. Increased perinatal mortality and morbidity in monochorionic versus dichorionic twin pregnancies: clinical implications of a large Dutch cohort study. BJOG. 2008; 115: 5867.Google Scholar
Lewi, L, Van Schoubroeck, D, Gratacós, E, et al. Monochorionic diamniotic twins: complications and management options. Curr Opin Obstet Gynecol. 2003; 15: 177–94.Google Scholar
Hamilton, EF, Platt, RW, Morin, L, Usher, R, Kramer, M. How small is too small in a twin pregnancy? Am J Obstet Gynecol. 1998; 179: 682–5.Google Scholar
Ville, Y. Monochorionic twin pregnancies: ‘les liaisons dangereuses’. Ultrasound Obstet Gynecol. 1997; 10: 82–5.Google Scholar
Lewi, L, Jani, J, Cannie, M, et al. Intertwin anastomoses in monochorionic placentas after fetoscopic laser coagulation for twin-to-twin transfusion syndrome: is there more than meets the eye? Am J Obstet Gynecol. 2006; 194; 790–5.Google Scholar
Quintero, RA, Morales, WJ, Allen, MH, et al. Staging of twin-twin transfusion syndrome. J Perinatol. 1999; 19: 550–5.Google Scholar
Wee, LY, Fisk, NM. The twin-twin transfusion syndrome. Semin Neonatol. 2002; 7; 187202.Google Scholar
Weir, PE, Ratten, GJ, Beischer, NA. Acute polyhydramnios: a complication of monozygous twin pregnancy. BJOG. 1979; 86: 849–53.Google Scholar
Robyr, R, Lewi, L, Salomon, LJ, et al. Prevalence and management of late fetal complications following successful selective laser coagulation of chorionic plate anastomoses in twin-to-twin transfusion syndrome. Am J Obstet Gynecol. 2006; 194: 796803.Google Scholar
De Lia, JE. Surgery of the placenta and umbilical cord. Clin Obstet Gynecol. 1996; 39: 607–25.Google Scholar
Haverkamp, F, Lex, C, Hanisch, C, Fahnenstich, H, Zerres, K. Neurodevelopmental risks in twin-to-twin transfusion syndrome: preliminary findings. Eur J Paediatr Neurol. 2001; 5; 21–7.Google Scholar
Senat, MV, Deprest, J, Boulvain, M, et al. A randomized trial of endoscopic laser surgery versus serial amnioreduction for severe twin-to-twin transfusion syndrome at midgestation. N Engl J Med. 2004; 351: 136–44.Google Scholar
De Lia, J, Fisk, N, Hecher, K, et al. Twin-to-twin transfusion syndrome – debates on the etiology, natural history and management. Ultrasound Obstet Gynecol. 2000; 16: 210–13.Google Scholar
Saade, GR, Belfort, MA, Berry, DL, et al. Amniotic septostomy for the treatment of twin oligohydramnios-polyhydramnios sequence. Fetal Diagn Ther. 1998; 13: 8693.Google Scholar
Garry, D, Lysikiewicz, A, Mays, J, Canterino, J, Tejani, N. Intra-amniotic pressure reduction in twin-twin transfusion syndrome. J Perinatol. 1998; 18: 284–6.Google Scholar
Moise, KJJ, Dorman, K, Lamvu, G, et al. A randomized trial of amnioreduction versus septostomy in the treatment of twin-twin transfusion syndrome. Am J Obstet Gynecol. 2005; 193: 701–7.Google Scholar
Johnson, JR, Rossi, KQ, O’Shaughnessy, RW. Amnioreduction versus septostomy in twin-twin transfusion syndrome. Am J Obstet Gynecol. 2001; 185: 1044–7.Google Scholar
Gilbert, WM, Davis, SE, Kaplan, C, et al. Morbidity associated with prenatal disruption of the dividing membrane in twin gestations. Obstet Gynecol. 1991; 78: 623–30.Google Scholar
Feldman, DM, Odibo, A, Campbell, WA, Rodis, JF. Iatrogenic monoamniotic twins as a complication of therapeutic amniocentesis. Obstet Gynecol. 1998; 91: 815–16.Google Scholar
Rujiwetpongstorn, J, Tongsong, T. Amniotic band syndrome following septostomy in management of twin-twin transfusion syndrome: a case report. J Perinatol. 2008; 28: 377–9.Google Scholar
Cruz-Martinez, R, Van Mieghem, T, Lewi, L, et al. Incidence and clinical implications of early inadvertent septostomy after laser therapy for twin-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2011; 37: 458–62.Google Scholar
Winer, N, Salomon, LJ, Essaoui, M, et al. Pseudoamniotic band syndrome: a rare complication of monochorionic twins with fetofetal transfusion syndrome treated by laser coagulation. Am J Obstet Gynecol. 2008; 198: 393. e1–5.Google Scholar
Hecher, K, Diehl, W, Zikulnig, L, et al. Endoscopic laser coagulation of placental anastomoses in 200 pregnancies with severe mid-trimester twin-to-twin transfusion syndrome. Eur J Obstet Gynecol Reprod Biol. 2000; 92: 135–9.Google Scholar
Gray, PH, Cincotta, R, Chan, FY, Soong, B. Perinatal outcomes with laser surgery for twin-twin transfusion syndrome. Twin Res Hum Genet. 2006; 9: 438–43.Google Scholar
Lenclen, R, Paupe, A, Ciarlo, G, et al. Neonatal outcome in preterm monochorionic twins with twin-to-twin transfusion syndrome after intrauterine treatment with amnioreduction or fetoscopic laser surgery: comparison with dichorionic twins. Am J Obstet Gynecol. 2007; 196; 450. e1–7.Google Scholar
Roberts, D, Neilson, JP, Kilby, M, et al. Interventions for the treatment of twin-twin transfusion syndrome. Cochrane Database Syst Rev. 2008; 1: CD002073.Google Scholar
Rossi, AC, D’Addario, V. Laser therapy and serial amnioreduction as treatment for twin-twin transfusion syndrome: a meta-analysis and review of literature. Am J Obstet Gynecol. 2008; 198: 147–52.Google Scholar
Salomon, LJ, Ortqvist, L, Aegerter, P, et al. Long-term developmental follow-up of infants who participated in a randomized clinical trial of amniocentesis versus laser photocoagulation for the treatment of twin-to-twin transfusion syndrome (TTTS). Am J Obstet Gynecol. 2010; 203; 444. e1–7.Google Scholar
Benirschke, K, Kim, CK. Multiple pregnancy. N Engl J Med. 1973; 288: 1276–84.Google Scholar
De Vore, G, Dixon, J, Hobbins, JC. Fetoscope-directed Nd:YAG laser: a potential tool for fetal surgery. Am J Obstet Gynecol. 1983; 143: 379–80.Google Scholar
De Lia, JE, Cruikshank, DP, Keye, WR. Fetoscopic neodymium:YAG laser occlusion of placental vessels in severe twin-twin transfusion syndrome. Obstet Gynecol. 1990; 75: 1046–53.Google Scholar
Ville, Y, Hyett, JA, Vandenbussche, FP, Nicolaides, KH. Endoscopic laser coagulation of umbilical cord vessels in twin reversed arterial perfusion sequence. Ultrasound Obstet Gynecol. 1994; 4: 396–8.Google Scholar
Ville, Y, Hyett, J, Hecher, K, Nicolaides, K. Preliminary experience with endoscopic laser surgery for severe twin-twin transfusion syndrome. N Engl J Med. 1995; 332: 224–7.Google Scholar
Ville, Y, Hecher, K, Gagnon, A, et al. Endoscopic laser coagulation in the management of severe twin-to-twin transfusion syndrome. BJOG. 1998; 105: 446–53.Google Scholar
Klaritsch, P, Albert, K, Van Mieghem, T, et al. Instrumental requirements for minimal invasive fetal surgery. BJOG. 2009; 116: 188–97.Google Scholar
Nizard, J, Barbet, JP, Ville, Y. Does the source of laser energy influence the coagulation of chorionic plate vessels? Comparison of Nd:YAG and Diode Laser on an ex-vivo placental model. Fetal Diagn Ther. 2007; 22: 33–7.Google Scholar
Chalouhi, GE, Essaoui, M, Stirnemann, J, et al. Laser therapy for twin-to-twin transfusion syndrome (TTTS). Prenat Diagn. 2011; 31: 637–46.Google Scholar
Salomon, LJ, Nasr, B, Nizard, J, et al. Emergency cerclage in cases of twin-to-twin transfusion syndrome with a short cervix at the time of surgery and relationship to perinatal outcome. Prenat Diagn. 2008; 28: 1256–61.Google Scholar
Van Peborgh, P, Rambaud, C, Ville, Y. Effect of laser coagulation on placental vessels: histological aspects. Fetal Diagn Ther. 1997; 12: 32–5.Google Scholar
Deprest, JA, Van Schoubroeck, D, Van Ballaer, PP, et al. Alternative technique for Nd:YAG laser coagulation in twin-to-twin transfusion syndrome with anterior placenta. Ultrasound Obstet Gynecol. 1998; 11: 347–52.Google Scholar
Huber, A, Baschat, AA, Bregenzer, T, et al. Laser coagulation of placental anastomoses with a 30 degrees fetoscope in severe mid-trimester twin-twin transfusion syndrome with anterior placenta. Ultrasound Obstet Gynecol. 2008; 31: 412–16.Google Scholar
Quintero, RA, Bornick, PW, Allen, MH, Johson, PK. Selective laser photocoagulation of communicating vessels in severe twin-twin transfusion syndrome in women with an anterior placenta. Obstet Gynecol. 2001; 97: 477–81.Google Scholar
Middeldorp, JM, Lopriore, E, Sueters, M, et al. Laparoscopically guided uterine entry for fetoscopy in twin-to-twin transfusion syndrome with completely anterior placenta: a novel technique. Fetal Diagn Ther. 2007; 22: 409–15.Google Scholar
De Lia, JE, Kuhlmann, RS, Harstad, TW, Cruikshank, DP. Fetoscopic laser ablation of placental vessels in severe previable twin-twin transfusion syndrome. Am J Obstet Gynecol. 1995; 172: 1202–8.Google Scholar
Stirnemann, JJ, Nasr, B, Quarello, E, et al. A definition of selectivity in laser coagulation of chorionic plate anastomoses in twin-to-twin transfusion syndrome and its relationship to perinatal outcome. Am J Obstet Gynecol. 2008; 198: 62. e1–6.Google Scholar
Quintero, RA, Comas, C, Bornick, PW, et al. Selective versus non-selective laser photocoagulation of placental vessels in twin-to-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2000; 16: 230–6.Google Scholar
Martinez, JM, Bermudez, C, Becerra, C, et al. The role of Doppler studies in predicting individual intrauterine fetal demise after laser therapy for twin–twin transfusion syndrome. Ultrasound Obstet Gynecol. 2003; 22: 246–51.Google Scholar
Quintero, RA, Martinez, JM, Lopez, J, et al. Individual placental territories after selective laser photocoagulation of communicating vessels in twin-twin transfusion syndrome. Am J Obstet Gynecol. 2005; 192: 1112–18.Google Scholar
Crisan, LS, Kontopoulos, EV, Quintero, RA. Appraisal of the selectivity index in a cohort of patients treated with laser surgery for twin-twin transfusion syndrome. Am J Obstet Gynecol. 2010; 202: 157. e1–5.Google Scholar
Lopriore, E, Slaghekke, F, Middeldorp, JM, et al. Residual anastomoses in twin-to-twin transfusion syndrome treated with selective fetoscopic laser surgery: localization, size, and consequences. Am J Obstet Gynecol. 2009; 201: 66. e1–4.Google Scholar
Sago, H, Hayashi, S, Saito, M, et al. The outcome and prognostic factors of twin-twin transfusion syndrome following fetoscopic laser surgery. Prenat Diagn. 2010; 30: 1185–91.Google Scholar
Thilaganathan, B, Gloeb, DJ, Sairam, S, Tekay, A. Sono-endoscopic delineation of the placental vascular equator prior to selective fetoscopic laser ablation in twin-to-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2000; 16: 226–9.Google Scholar
Ierullo, AM, Papageorghiou, AT, Bhide, A, et al. Severe twin-twin transfusion syndrome: outcome after fetoscopic laser ablation of the placental vascular equator. BJOG. 2007; 114: 689–93.Google Scholar
Morris, RK, Selman, TJ, Harbidge, A, Martin, WL, Kilby, MD. Fetoscopic laser coagulation for severe twin-to-twin transfusion syndrome: factors influencing perinatal outcome, learning curve of the procedure and lessons for new centres. BJOG. 2010; 117; 1350–7.Google Scholar
Sepulveda, W, Wong, AE, Dezerega, V, Devoto, JC, Alcalde, JL. Endoscopic laser surgery in severe second-trimester twin-twin transfusion syndrome: a three-year experience from a Latin American center. Prenat Diagn. 2007; 27: 1033–8.Google Scholar
Johnston, TA, Cameron, AD. Atosiban for the primary prevention of preterm labour. BJOG. 2001; 108: 886–7.Google Scholar
Yamamoto, M, Barki, G, Ville, Y. Direct visual control on cord coagulation using a fetoscopy-guided bipolar forceps. Description of a new technique. Prenat Diagn. 2010; 30: 156–8.Google Scholar
Cheong-See, F, Schuit, E, Arroyo-Manzano, D, et al. Prospective risk of stillbirth and neonatal complications in twin pregnancies: systematic review and meta-analysis. BMJ. 2016; 354: i4353.Google Scholar
Robyr, R, Lewi, L, Yamamoto, M, Deprest, J, Ville, Y. Permanent feto-fetal transfusion from the recipient to the donor twin: a complication of laser surgery in twin-to-twin transfusion syndrome. Am J Obstet Gynecol. 2004; 191: 42.Google Scholar
Ville, Y, Hecher, K, Gagnon, A, et al. Endoscopic laser coagulation in the management of severe twin-to-twin transfusion syndrome. BJOG. 1998; 105: 446–53.Google Scholar
Yamamoto, M, El Murr, L, Robyr, R, et al. Incidence and impact of perioperative complications in 175 fetoscopy-guided laser coagulation of chorionic plate anastomoses in fetofetal transfusion syndrome before 26 weeks of gestation. Am J Obstet Gynecol. 2005; 193: 1110–16.Google Scholar
Merz, W, Tchatcheva, K, Gembruch, U, Kohl, T. Maternal complications of fetoscopic laser photocoagulation (FLP) for treatment of twin-twin transfusion syndrome (TTTS). J Perinat Med. 2010; 38: 439–43.Google Scholar
Habli, M, Bombrys, A, Lewis, D, et al. Incidence of complications in twin-twin transfusion syndrome after selective fetoscopic laser photocoagulation: a single-center experience. Am J Obstet Gynecol. 2009; 201: 417. e1–7.Google Scholar
Grannum, PA, Copel, JA. Invasive fetal procedures. Radiol Clin North Am. 1990; 28: 217–26.Google Scholar
Deprest, JA, Van Ballaer, PP, Evrard, VA, et al. Experience with fetoscopic cord ligation. Eur J Obstet Gynecol Reprod Biol. 1998; 81: 157–64.Google Scholar
Quintero, RA. Treatment of previable premature ruptured membranes. Clin Perinatol. 2003; 30: 573–89.Google Scholar
Harrison, MR, Mychaliska, GB, Albanese, CT, et al. Correction of congenital diaphragmatic hernia in utero. IX: Fetuses with poor prognosis (liver herniation and low lung-to-head ratio) can be saved by fetoscopic temporary tracheal occlusion. J Pediatr Surg. 1998; 33: 1017–22.Google Scholar
Bilic, G, Brubaker, C, Messersmith, PB, et al. Injectible candidate sealants for fetal membrane repair: bonding and toxicity in vitro. Am J Obstet Gynecol. 2010; 202: 85. e1–85. e9.Google Scholar
Liekens, D, Lewi, L, Jani, J, et al. Enrichment of collagen plugs with platelets and amniotic fluid cells increases cell proliferation in sealed iatrogenic membrane defects in the foetal rabbit model. Prenat Diagn. 2008; 28: 503–7.Google Scholar
Devlieger, R, Millar, LK, Bryant-Greenwood, G, et al. Fetal membrane healing after spontaneous and iatrogenic membrane rupture: a review of current evidence. Am J Obstet Gynecol. 2006; 195; 1512–20.Google Scholar
Lopriore, E, Lewi, L, Oepkes, D, et al. In utero acquired limb ischemia in monochorionic twins with and without twin-to-twin trnsfusion syndrome. Prenat Diagn. 2008; 28: 800–4.Google Scholar
Akkermans, J, Peeters, SH, Klumper, FJ, Lopriore, E, Middeldorp, JM, Oepkes, D. Twenty-five years of fetoscopic laser coagulation in twinetwin transfusion syndrome: a systematic review. Fetal Diagn Ther. 2015; 38: 241. e53.Google Scholar
Djaafri, F, Stirnemann, J, Mediouni, I, Colmant, C, Ville, Y. Twin-to-twin transfusion syndrome: what we have learned from clinical trials ? Sem Fetal Neonatal Med. 2017; 22: 367. e375Google Scholar
Stirnemann, JJ, Nasr, B, Proulx, F, Essaoui, M, Ville, Y. Evaluation of the CHOP cardiovascular score as a prognostic predictor of outcome in twin–twin transfusion syndrome after laser coagulation of placental vessels in a prospective cohort. Ultrasound Obstet Gynecol. 2010; 36: 52–7.Google Scholar
Taylor, MJ, Govender, L, Jolly, M, et al. Validations of the Quintero staging system for twin–twin transfusion syndrome. Obstet Gynecol. 2002; 100: 1257–65.Google Scholar
Dickinson, JE, Evans, S. The progression of disease stage in twin–twin transfusion syndrom. J Matern Fetal Neonatal Med. 2004; 16: 95101.Google Scholar
O’Donoghue, K, Cartwright, E, Galea, P, Fisk, NM. Stage I twin-twin transfusion syndrome: rates of progression and regression in relation to outcome. Ultrasound Obstet Gynecol. 2007; 30: 958–66.Google Scholar
Wagner, MM, Lopriore, E, Klumper, FJ, Oepkes, D, Vandenbussche, FP, Middeldorp, JM. Short- and long-term outcome in stage 1 twin-to-twin transfusion syndrome treated with laser surgery compared with conservative management. Am J Obstet Gynecol. 2009; 201: 286. e1–6.Google Scholar
Khalil, A, Cooper, E, Townsend, R, Thilaganathan, B. Evolution of stage 1 twin-to-twin transfusion syndrome (TTTS): systematic review and meta-analysis. Twin Res Hum Genet. 2016; 19: 207–16.Google Scholar
Molina, S, Papanna, R, Moise, KJ Jr., Johnson, A. Management of Stage I twin-to-twin transfusion syndrome: an international survey. Ultrasound Obstet Gynecol. 2010; 36: 42–7.Google Scholar
Stirnemann, JDjaafri, FKim, A, et al. Preterm premature rupture of membranes is a collateral effect of improvement in perinatal outcomes following fetoscopic coagulation of chorionic vessels for twin-twin transfusion syndrome: a retrospective observational study of 1092 cases. BJOG2018; 125: 1154–62.Google Scholar
Vayssiere, C, Benoist, G, Blondel, B, et al. Twin pregnancies: guidelines for clinical practice from the French College of Gynaecologists and Obstetricians (CNGOF). Eur J Obstet Gynecol Reprod Biol. 2011; 156: 1217.Google Scholar
Kilby, MD, Bricker, L. Management of monochorionic twin pregnancies. BJOG. 2016; 124: e145.Google Scholar

References

Dickinson, JE, Evans, SF. Obstetric and perinatal outcomes from The Australian and New Zealand twin-twin transfusion syndrome registry. Am J Obstet Gynecol. 2000; 182: 706–12.Google Scholar
Van Mieghem, T, Martin, AMM, Weber, R, Barrea, C, Windrim, R, Hornberger, LKK, Jaeggi, E, Ryan, G. Fetal cardiac function in recipient twins undergoing fetoscopic laser ablation of placental anastomoses for Stage IV twin-twin transfusion syndrome. Ultrasound Obs Gynecol. 2013; 42: 64–9.Google Scholar
Van Mieghem, T, Lewi, L, Gucciardo, L, Dekoninck, P, Van Schoubroeck, D, Devlieger, R, Deprest, J. The fetal heart in twin-to-twin transfusion syndrome. Int J Pediatr. 2010; 2010: 18.Google Scholar
Senat, MV, Deprest, J, Boulvain, M, Paupe, A, Winer, N, Ville, Y. Endoscopic laser surgery versus serial amnioreduction for severe twin-to-twin transfusion syndrome. N Engl J Med. 2004; 351: 136–44.Google Scholar
Baud, D, Windrim, R, Keunen, J, Kelly, EN, Shah, P, Van Mieghem, T, Seaward, PGR, Ryan, G. Fetoscopic laser therapy for twin-twin transfusion syndrome before 17 and after 26 weeks’ gestation. Am J Obstet Gynecol. 2013; 208: 197. e1–7.Google Scholar
Lecointre, L, Sananes, N, Weingertner, AS, Kohler, M, Guerra, F, Fritz, G, Viville, B, Langer, B, Nisand, I, Favre, R. Fetoscopic laser coagulation for twin-twin transfusion syndrome before 17 weeks’ gestation: laser data, complications and neonatal outcome. Ultrasound Obstet Gynecol. 2014; 44: 299303.Google Scholar
Middeldorp, JM, Lopriore, E, Sueters, M, Klumper, FJCM, Kanhai, HHH, Vandenbussche, FPHA, Oepkes, D. Twin-to-twin transfusion syndrome after 26 weeks of gestation: is there a role for fetoscopic laser surgery? BJOG. 2007; 114: 694–8.Google Scholar
Touboul, C, Boulvain, M, Picone, O, Levaillant, J-M, Frydman, R, Senat, M-V. Normal fetal urine production rate estimated with 3-dimensional ultrasonography using the rotational technique (virtual organ computer-aided analysis). Am J Obstet Gynecol. 2008; 199: 57. e1–57. e5.Google Scholar
Dekoninck, P, Deprest, J, Lewi, P, Richter, J, Galjaard, S, Van Keirsbilck, J, Van Calsteren, K, Lewi, L. Gestational age-specific reference ranges for amniotic fluid assessment in monochorionic diamniotic twin pregnancies. Ultrasound Obstet Gynecol. 2013; 41: 649–52.Google Scholar
Emery, SP, Bahtiyar, MO, Dashe, JS, Wilkins-Haug, LE, Johnson, A, Paek, BW, et al. The North American Fetal Therapy Network Consensus Statement. Obstet Gynecol. 2015; 125: 1236–43.Google Scholar
Van Mieghem, T, Eixarch, E, Gucciardo, L, Done, E, Gonzales, I, Van Schoubroeck, D, Lewi, L, Gratacos, E, Deprest, J. Outcome prediction in monochorionic diamniotic twin pregnancies with moderately discordant amniotic fluid. Ultrasound Obstet Gynecol. 2011; 37: 1521.Google Scholar
Slaghekke, F, Lopriore, E, Lewi, L, Middeldorp, JM, Van Zwet, EW, Weingertner, AS, et al. Fetoscopic laser coagulation of the vascular equator versus selective coagulation for twin-to-twin transfusion syndrome: an open-label randomised controlled trial. Lancet. 2014; 383: 2144–51.Google Scholar
Petersen, SG, Gibbons, KS, Luks, FI, Lewi, L, Diemert, A, Hecher, K, et al. The impact of entry technique and access diameter on prelabour rupture of membranes following primary fetoscopic laser treatment for twin-twin transfusion syndrome. Fetal Diagn Ther. 2016; 40: 100–9.Google Scholar
Rossi, AC, D’Addario, V. Umbilical cord occlusion for selective feticide in complicated monochorionic twins: a systematic review of literature. Am J Obstet Gynecol. 2009; 200: 123–9.Google Scholar
Emery, SP, Hasley, SK, Catov, JM, Miller, RS, Moon-Grady, AJ, Baschat, AA, et al. North American Fetal Therapy Network: intervention vs expectant management for stage I twin-twin transfusion syndrome. Am J Obstet Gynecol. 2015; 215: 346. e1–7.Google Scholar
Murata, M, Ishii, K, Taguchi, T, Mabuchi, A, Kawaguchi, H, Yamamoto, R, Hayashi, S, Mitsuda, N. The prevalence and clinical features of twin-twin transfusion syndrome with onset during the third trimester. J Perinat Med. 2014; 42: 93–8.Google Scholar
Mari, G, Roberts, A, Detti, L, Kovanci, E, Stefos, T, Bahado-Singh, RO, Deter, RL, Fisk, NM. Perinatal morbidity and mortality rates in severe twin-twin transfusion syndrome: results of the International Amnioreduction Registry. Am J Obstet Gynecol. 2001; 185: 708–15.Google Scholar
Fichera, A, Ambrosi, C, Taddei, F, Gasparotti, R, Frusca, T. Severe brain damage from twin-twin transfusion syndrome treated with serial amnioreductions after 26 weeks: a case to reconsider the gestational age limits of laser therapy. Fetal Diagn Ther. 2009; 25: 203–5.Google Scholar

References

Casanova, J, Paiva, C, Carvalho, C, Cunha, AC. Twin anemia polycythemia sequence: a report of three cases. J Reprod Med. 2014; 59: 596–8.Google Scholar
Lopriore, E, Deprest, J, Slaghekke, F, Oepkes, D, Middeldorp, JM, Vandenbussche, FP, et al. Placental characteristics in monochorionic twins with and without twin anemia-polycythemia sequence. Obstet Gynecol. 2008; 112: 753–8.Google Scholar
Gucciardo, L, Lewi, L, Vaast, P, Debska, M, De Catte, L, Van Mieghem, T, et al. Twin anemia polycythemia sequence from a prenatal perspective. Prenat Diagn. 2010; 30: 438–42.Google Scholar
Habli, M, Bombrys, A, Lewis, D, Lim, FY, Polzin, W, Maxwell, R, et al. Incidence of complications in twin-twin transfusion syndrome after selective fetoscopic laser photocoagulation: a single-center experience. Am J Obstet Gynecol. 2009; 201: 417. e1–7.Google Scholar
Lopriore, E, Slaghekke, F, Middeldorp, JM, Klumper, FJ, Oepkes, D, Vandenbussche, FP. Residual anastomoses in twin-to-twin transfusion syndrome treated with selective fetoscopic laser surgery: localization, size, and consequences. Am J Obstet Gynecol. 2009; 201: 66. e1–4.Google Scholar
Lopriore, E, Middeldorp, JM, Oepkes, D, Kanhai, HH, Walther, FJ, Vandenbussche, FP. Twin anemia-polycythemia sequence in two monochorionic twin pairs without oligo-polyhydramnios sequence. Placenta. 2007; 28: 4751.Google Scholar
Lopriore, E, van den Wijngaard, JP, Middeldorp, JM, Oepkes, D, Walther, FJ, van Gemert, MJ, et al. Assessment of feto-fetal transfusion flow through placental arterio-venous anastomoses in a unique case of twin-to-twin transfusion syndrome. Placenta. 2007; 28: 209–11.Google Scholar
van den Wijngaard, JP, Lewi, L, Lopriore, E, Robyr, R, Middeldorp, JM, Vandenbussche, FP, et al. Modeling severely discordant hematocrits and normal amniotic fluids after incomplete laser therapy in twin-to-twin transfusion syndrome. Placenta. 2007; 28: 611–15.Google Scholar
Zhao, DP, de Villiers, SF, Slaghekke, F, Walther, FJ, Middeldorp, JM, Oepkes, D, et al. Prevalence, size, number and localization of vascular anastomoses in monochorionic placentas. Placenta. 2013; 34: 589–93.Google Scholar
Suzuki, S. Twin anemia-polycythemia sequence with placental arterio-arterial anastomoses. Placenta. 2010; 31: 652.Google Scholar
de Villiers, SF, Slaghekke, F, Middeldorp, JM, Walther, FJ, Oepkes, D, Lopriore, E. Placental characteristics in monochorionic twins with spontaneous versus post-laser twin anemia-polycythemia sequence. Placenta. 2013; 34: 456–9.Google Scholar
de Villiers, SF, Slaghekke, F, Middeldorp, JM, Walther, FJ, Oepkes, D, Lopriore, E. Arterio-arterial vascular anastomoses in monochorionic placentas with and without twin-twin transfusion syndrome. Placenta. 2012; 33: 652–4.Google Scholar
Zhao, DP, Cambiaso, O, Otano, L, Lewi, L, Deprest, J, Sun, LM, et al. Veno-venous anastomoses in twin-twin transfusion syndrome: a multicenter study. Placenta. 2015; 36: 911–14.Google Scholar
de Villiers, SF, Zhao, DP, Cohen, D, van Zwet, EW, Duan, T, Oepkes, D, et al. Correlation between veno-venous anastomoses, TTTS and perinatal mortality in monochorionic twin pregnancies. Placenta. 2015; 36: 603–6.Google Scholar
Mahieu-Caputo, D, Dommergues, M, Delezoide, AL, Lacoste, M, Cai, Y, Narcy, F, et al. Twin-to-twin transfusion syndrome. Role of the fetal renin-angiotensin system. Am J Pathol. 2000; 156: 629–36.Google Scholar
Oepkes, D, Seaward, PG, Vandenbussche, FP, Windrim, R, Kingdom, J, Beyene, J, et al. Doppler ultrasonography versus amniocentesis to predict fetal anemia. N Engl J Med. 2006; 355: 156–64.Google Scholar
Robyr, R, Lewi, L, Salomon, LJ, Yamamoto, M, Bernard, JP, Deprest, J, et al. Prevalence and management of late fetal complications following successful selective laser coagulation of chorionic plate anastomoses in twin-to-twin transfusion syndrome. Am J Obstet Gynecol. 2006; 194: 796803.Google Scholar
Slaghekke, F, Pasman, S, Veujoz, M, Middeldorp, JM, Lewi, L, Devlieger, R, et al. Middle cerebral artery peak systolic velocity to predict fetal hemoglobin levels in twin anemia-polycythemia sequence. Ultrasound Obstet Gynecol. 2015; 46: 432–6.Google Scholar
Slaghekke, F, Kist, WJ, Oepkes, D, Pasman, SA, Middeldorp, JM, Klumper, FJ, et al. Twin anemia-polycythemia sequence: diagnostic criteria, classification, perinatal management and outcome. Fetal Diagn Ther. 2010; 27: 181–90.Google Scholar
Fishel-Bartal, M, Weisz, B, Mazaki-Tovi, S, Ashwal, E, Chayen, B, Lipitz, S, et al. Can middle cerebral artery peak systolic velocity predict polycythemia in monochorionic-diamniotic twins? Evidence from a prospective cohort study. Ultrasound Obstet Gynecol. 2016; 48: 470–5.Google Scholar
Tollenaar, LS, Lopriore, E, Middeldorp, JM, Haak, MC, Klumper, FJ, Oepkes, D, et al. Improved antenatal prediction of twin anemia polycythemia sequence by delta middle cerebral artery peak systolic velocity: new antenatal classification system. Ultrasound Obstet Gynecol. 2019; 53: 788–93.Google Scholar
Soundararajan, LP, Howe, DT. Starry sky liver in twin anemia-polycythemia sequence. Ultrasound Obstet Gynecol. 2014; 43: 597–9.Google Scholar
Movva, VC, Rijhsinghani, A. Discrepancy in placental echogenicity: a sign of twin anemia polycythemia sequence. Prenat Diagn. 2014; 34: 809–11.Google Scholar
Stritzke, A, Thomas, S, Somerset, D. Placental dichotomy: a hint in twin anemia polycythemia sequence. J Obstet Gynaecol Can. 2014; 36: 1097–100.Google Scholar
Lopriore, E, Slaghekke, F, Oepkes, D, Middeldorp, JM, Vandenbussche, FP, Walther, FJ. Hematological characteristics in neonates with twin anemia-polycythemia sequence (TAPS). Prenat Diagn. 2010; 30: 251–5.Google Scholar
Lopriore, E, Holtkamp, N, Sueters, M, Middeldorp, JM, Walther, FJ, Oepkes, D. Acute peripartum twin-twin transfusion syndrome: incidence, risk factors, placental characteristics and neonatal outcome. J Obstet Gynaecol Res. 2014; 40: 1824.Google Scholar
Lopriore, E, Slaghekke, F, Middeldorp, JM, Klumper, FJ, van Lith, JM, Walther, FJ, et al. Accurate and simple evaluation of vascular anastomoses in monochorionic placenta using colored dye. J Vis Exp. 2011; 55: e3208.Google Scholar
Tollenaar, LS, Zhao, DP, Middeldorp, JM, Slaghekke, F, Oepkes, D, Lopriore, E. Color difference in placentas with twin anemia-polycythemia sequence: an additional diagnostic criterion? Fetal Diagn Ther. 2016; 40: 123–7.Google Scholar
De Paepe, ME, Gundogan, F, Mao, Q, Chu, S, Shapiro, S. Redness discordance in monochorionic twin placentas: correlation with clinical and placental findings. Placenta. 2017; 60: 5460.Google Scholar
Tollenaar, LSA, Zhao, DP, Middeldorp, JM, Oepkes, D, Slaghekke, F, Lopriore, E. Can color difference on the maternal side of the placenta distinguish between acute peripartum twin-twin transfusion syndrome and twin anemia-polycythemia sequence? Placenta. 2017; 57: 189–93.Google Scholar
Lopriore, E, Slaghekke, F, Kersbergen, KJ, de Vries, LS, Drogtrop, AP, Middeldorp, JM, et al. Severe cerebral injury in a recipient with twin anemia-polycythemia sequence. Ultrasound Obstet Gynecol. 2013; 41: 702–6.Google Scholar
Luminoso, D, Figueira, CO, Marins, M, Peralta, CF. Fetal brain lesion associated with spontaneous twin anemia-polycythemia sequence. Ultrasound Obstet Gynecol. 2013; 42: 721–2.Google Scholar
Tollenaar, LS, Slaghekke, F, Middeldorp, JM, Klumper, FJ, Haak, MC, Oepkes, D, et al. Twin anemia polycythemia sequence: current views on pathogenesis, diagnostic criteria, perinatal management, and outcome. Twin Res Hum Genet. 2016; 19: 222–33.Google Scholar
Lopriore, E, Hecher, K, Vandenbussche, FP, van den Wijngaard, JP, Klumper, FJ, Oepkes, D. Fetoscopic laser treatment of twin-to-twin transfusion syndrome followed by severe twin anemia-polycythemia sequence with spontaneous resolution. Am J Obstet Gynecol. 2008; 198: e4–7.Google Scholar
Herway, C, Johnson, A, Moise, K, Moise, KJ Jr. Fetal intraperitoneal transfusion for iatrogenic twin anemia-polycythemia sequence after laser therapy. Ultrasound Obstet Gynecol. 2009; 33: 592–4.Google Scholar
Slaghekke, F, van den Wijngaard, JP, Akkermans, J, van Gemert, MJ, Middeldorp, JM, Klumper, FJ, et al. Intrauterine transfusion combined with partial exchange transfusion for twin anemia polycythemia sequence: modeling a novel technique. Placenta. 2015; 36: 599602.Google Scholar
Bahtiyar, MO, Ekmekci, E, Demirel, E, Irani, RA, Copel, JA. In utero partial exchange transfusion combined with in utero blood transfusion for prenatal management of twin anemia-polycythemia sequence. Fetal Diagn Ther. 2018; 45: 2835.Google Scholar
Genova, L, Slaghekke, F, Klumper, FJ, Middeldorp, JM, Steggerda, SJ, Oepkes, D, et al. Management of twin anemia-polycythemia sequence using intrauterine blood transfusion for the donor and partial exchange transfusion for the recipient. Fetal Diagn Ther. 2013; 34: 121–6.Google Scholar
Slaghekke, F, Lopriore, E, Lewi, L, Middeldorp, JM, van Zwet, EW, Weingertner, AS, et al. Fetoscopic laser coagulation of the vascular equator versus selective coagulation for twin-to-twin transfusion syndrome: an open-label randomised controlled trial. Lancet. 2014; 383: 2144–51.Google Scholar
Senat, MV, Deprest, J, Boulvain, M, Paupe, A, Winer, N, Ville, Y. Endoscopic laser surgery versus serial amnioreduction for severe twin-to-twin transfusion syndrome. N Engl J Med. 2004; 351: 136–44.Google Scholar
Slaghekke, F, Favre, R, Peeters, SH, Middeldorp, JM, Weingertner, AS, van Zwet, EW, et al. Laser surgery as a management option for twin anemia-polycythemia sequence. Ultrasound Obstet Gynecol. 2014; 44: 304–10.Google Scholar
Sananes, N, Veujoz, M, Severac, F, Barthoulot, M, Meyer, N, Weingertner, AS, et al. Evaluation of the utility of in utero treatment of twin anemia-polycythemia sequence. Fetal Diagn Ther. 2015; 38: 170–8.Google Scholar
Slaghekke, F, van Klink, JM, Koopman, HM, Middeldorp, JM, Oepkes, D, Lopriore, E. Neurodevelopmental outcome in twin anemia-polycythemia sequence after laser surgery for twin-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2014; 44: 316–21.Google Scholar
Verbeek, L, Slaghekke, F, Hulzebos, CV, Oepkes, D, Walther, FJ, Lopriore, E. Hypoalbuminemia in donors with twin anemia-polycythemia sequence: a matched case-control study. Fetal Diagn Ther. 2013; 33: 241–5.Google Scholar
Verbeek, L, Slaghekke, F, Favre, R, Vieujoz, M, Cavigioli, F, Lista, G, et al. Short-term postnatal renal function in twin anemia-polycythemia sequence. Fetal Diagn Ther. 2016; 39: 192–7.Google Scholar
Stranak, Z, Korcek, P, Hympanova, L, Kyncl, M, Krofta, L. Prenatally acquired multiple limb ischemia in a very low birth weight monochorionic twin. Fetal Diagn Ther. 2017; 41: 237–8.Google Scholar
Taniguchi, K, Sumie, M, Sugibayashi, R, Wada, S, Matsuoka, K, Sago, H. Twin anemia-polycythemia sequence after laser surgery for twin-twin transfusion syndrome and maternal morbidity. Fetal Diagn Ther. 2015; 37: 148–53.Google Scholar

References

Deprest, J, Jani, J, Lewi, L, et al. Fetoscopic surgery: encouraged by clinical experience and boosted by instrument innovation. Semin Fetal Neonatal Med. 2006; 11: 398412.Google Scholar
Rossi, AC, D’Addario, V. Laser therapy and serial amnioreduction as treatment for twin-twin transfusion syndrome: a meta-analysis and review of literature. Am J Obstet Gynecol. 2008; 198: 147–52.Google Scholar
van Klink, JMM, Koopman, HM, van Zwet, EW, et al. Cerebral injury and neurodevelopmental impairment after amnioreduction versus laser surgery in twin-twin transfusion syndrome: a systematic review and meta-analysis. Fetal Diagn Ther. 2013; 33: 81–9.Google Scholar
Senat, MV, Deprest, J, Boulvain, M, et al. Endoscopic laser surgery versus serial amnioreduction for severe twin-to-twin transfusion syndrome. New Engl J Med. 2004; 351: 136–44.Google Scholar
Slaghekke, F, Lopriore, E, Lewi, L, et al. Fetoscopic laser coagulation of the vascular equator versus selective coagulation for twin-to-twin transfusion syndrome: an open-label randomised controlled trial. Lancet. 2014; 383: 2144–51.Google Scholar
van Klink, JM, Koopman, HM, Rijken, M, et al. Long-term neurodevelopmental outcome in survivors of twin-to-twin transfusion syndrome. Twin Res Hum Genet. 2016; 19: 255–61.Google Scholar
Stirnemann, J, Chalouhi, G, Essaoui, M, et al. Fetal brain imaging following laser surgery in twin-to-twin surgery. BJOG. 2016; 125: 1186–91.Google Scholar
Lopriore, E, van Wezel-Meijler, G, Middeldorp, JM, et al. Incidence, origin, and character of cerebral injury in twin-to-twin transfusion syndrome treated with fetoscopic laser surgery. Am J Obstet Gynecol. 2006; 194: 1215–20.Google Scholar
Salomon, LJ, Ortqvist, L, Aegerter, P, 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: 444. e1–7.Google Scholar
Tosello, B, Blanc, J, Haumonté, JB, D'Ercole, C, Gire, C. Short and medium-term outcomes of live-born twins after fetoscopic laser therapy for twin-twin transfusion syndrome. J Perinat Med. 2014; 42: 99105.Google Scholar
Mullers, SM, McAuliffe, FM, Kent, E, et al. Outcome following selective fetoscopic laser ablation for twin to twin transfusion syndrome: an 8 year national collaborative experience. Eur J Obstet Gynecol Reprod Biol. 2015; 191: 125–9.Google Scholar
Campos, D, Arias, AV, Campos-Zanelli, TM, et al. Twin-twin transfusion syndrome: neurodevelopment of infants treated with laser surgery. Arq Neuropsiquiatr. 2016; 74: 307–13.Google Scholar
Lopriore, E, Ortibus, E, Acosta-Rojas, R, et al. Risk factors for neurodevelopment impairment in twin-twin transfusion syndrome treated with fetoscopic laser surgery. Obstet Gynecol. 2009; 113: 361–6.Google Scholar
van Klink, JM, Slaghekke, F, Balestriero, MA, et al. Neurodevelopmental outcome at 2 years in twin-twin transfusion syndrome survivors randomized for the Solomon trial. Am J Obstet Gynecol. 2015; 214: 113. e1–7.Google Scholar
Slaghekke, F, van Klink, JM, Koopman, HM, et al. Neurodevelopmental outcome in twin anemia-polycythemia sequence after laser surgery for twin-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2014; 44: 316–21.Google Scholar
Taniguchi, K, Sumie, M, Sugibayashi, R, et al. Twin anemia-polycythemia sequence after laser surgery for twin-twin transfusion syndrome and maternal morbidity. Fetal Diagn Ther. 2015; 37: 148–53.Google Scholar
Ashwal, E, Yinon, Y, Fishel-Bartal, M, et al. Twin Anemia-Polycythemia Sequence: perinatal Management and Outcome. Fetal Diagn Ther. 2016; 40: 2834.Google Scholar
Tollenaar, LS, Slaghekke, F, Middeldorp, JM, et al. Twin anemia polycythemia sequence: current views on pathogenesis, diagnostic criteria, perinatal management, and outcome. Twin Res Hum Genet. 2016; 19: 222–33.Google Scholar
Lopriore, E, Sluimers, C, Pasman, SA, et al. Neonatal morbidity in growth-discordant monochorionic twins: comparison between the larger and the smaller twin. Twin Res Hum Genet. 2012; 15: 541–6.Google Scholar
Inklaar, MJ, van Klink, JM, Stolk, TT, et al. Cerebral injury in monochorionic twins with selective intrauterine growth restriction: a systematic review. Prenat Diagn. 2014; 34: 205–13.Google Scholar
Swamy, RS, McConachie, H, Ng, J, et al. Cognitive outcome in childhood of birth weight discordant monochorionic twins: the long-term effects of fetal growth restriction. Arch Dis Child Fetal Neonatal Ed. 2018; 103: F512–16.Google Scholar
Edmonds, CJ, Isaacs, EB, Cole, TJ, et al. The effect of intrauterine growth on verbal IQ scores in childhood: a study of monozygotic twins. Pediatrics. 2010; 126: e1095–101.Google Scholar
Barker, DJ. Adult consequences of fetal growth restriction. Clin Obstet Gynecol. 2006; 49: 270–83.Google Scholar
Gluckman, PD, Hanson, MA, Cooper, C, Thornburg, KL. Effect of in utero and early-life conditions on adult health and disease. New Engl J Med. 2008; 359: 6173.Google Scholar
Heijmans, BT, Tobi, EW, Stein, AD, et al. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci U S A. 2008; 105: 17046–9.Google Scholar
Rossi, AC, D’Addario, V. Umbilical cord occlusion for selective feticide in complicated monochorionic twins:a systematic review of the literature. Am J Obstet Gynecol. 2009; 200: 123–9.Google Scholar
van den Bos, EM, van Klink, JM, Middeldorp, JM et al. Perinatal outcome after selective feticide in monochorionic twin pregnancies. Ultrasound Obstet Gynecol. 2013; 41: 653–8.Google Scholar
Robyr, R, Yamamoto, M, Ville, Y. Selective feticide in complicated monochorionic twin pregnancies using ultrasound-guided bipolar cord coagulation. BJOG. 2005; 112: 1344–8.Google Scholar
Lewi, L, Gratacós, E, Ortibus, E, et al. Pregnancy and infant outcome of 80 consecutive cord coagulations in complicated monochorionic multiple pregnancies. Am J Obstet Gynecol. 2006; 194: 782–9.Google Scholar
Moise, KJ Jr., Johnson, A, Moise, KY, Nickeleit, V. Radiofrequency ablation for selective reduction in the complicated monochorionic gestation. Am J Obstet Gynecol. 2008; 198: 198. e1–198. e5.Google Scholar
Delabaere, A, Favre, N, Velemir, L, et al. Pediatric outcome after selective feticide for 30 complicated monochorionic twin pregnancies. Gynecol Obstet Fertil. 2013; 41: 85–9.Google Scholar
van Klink, JM, Koopman, HM, Middeldorp, JM, et al. Long-term neurodevelopmental outcome after selective feticide in monochorionic pregnancies. BJOG. 2015; 122: 1517–24.Google Scholar
Wang, HM, Li, HY, Wang, XT, et al. Radiofrequency ablation for selective reduction in complex monochorionic multiple pregnancies: A case series. Taiwan J Obstet Gynecol. 2017; 56: 740–4.Google Scholar
Hillman, SC, Morris, RK, Kilby, MD. Co-twin prognosis after single fetal death. A systematic review and meta-analysis. Obstet Gynecol. 2011; 118: 928–40.Google Scholar
van Klink, JM, van Steenis, A, Steggerda, SJ, et al. Single fetal demise in monochorionic pregnancies: incidence and patterns of cerebral injury. Ultrasound Obstet Gynecol. 2015; 45: 294300.Google Scholar
Conte, G, Righini, A, Griffiths, PD, et al. Brain-injured survivors of monochorionic twin pregnancies complicated by single intrauterine death: MR findings in a multicenter study. Radiology. 2018; 288: 582–90.Google Scholar
O’Donoghue, K, Barigye, O, Pasquini, L, et al. Interstitial laser therapy for fetal reduction in monochorionic multiple pregnancy: loss rate and association with aplasia cutis congenita. Prenat. Diagn. 2008; 28: 535–43.Google Scholar
Mackie, FL, Morris, RK, Kilby, MD. Fetal brain injury in survivors of twin pregnancies complicated by demise of one twin: a review. Twin Res Hum Genet. 2016; 19: 262–7.Google Scholar
Aylward, GP, Hatcher, RP, Stripp, B, Gustafson, NF, Leavitt, LA. Who goes and who stays: subject loss in a multicenter, longitudinal follow-up study. J Dev Behav Pediatr. 1985; 6: 38.Google Scholar
Wolke, D, Söhne, B, Ohrt, B, Riegel, K. Follow-up of preterm children: important to document dropouts. Lancet. 1995; 345: 447.Google Scholar
Reisner, DP, Mahony, BS, Petty, CN, et al. Stuck twin syndrome: outcome in thirty-seven consecutive cases. Am J Obstet Gynecol. 1993; 169: 991–5.Google Scholar
Mari, G, Detti, L, Oz, U, Abuhamad, AZ. Long-term outcome in twin-twin transfusion syndrome treated with serial aggressive amnioreduction. Am J Obstet Gynecol. 2000; 183: 211–17.Google Scholar
Cincotta, RB, Gray, PH, Phythian, G, Rogers, YM, Chan, FY. Long term outcome of twin-twin transfusion syndrome. Arch Dis Child Fetal Neonatal Ed. 2000; 83: F171–6.Google Scholar
Haverkamp, F, Lex, C, Hanisch, C, Fahnenstich, H, Zerres, K. Neurodevelopmental risks in twin-to-twin transfusion syndrome: preliminary findings. Eur J Paediatr Neurol. 2001; 5: 21–7.Google Scholar
Frusca, T, Soregaroli, M, Fichera, A, et al. Pregnancies complicated by twin-twin transfusion syndrome: outcome and long-term neurological follow-up. Eur J Obstet Gynecol Reprod Biol. 2003; 107: 145–50.Google Scholar
Lopriore, E, Middeldorp, JM, Sueters, M, et al. Long-term neurodevelopmental outcome in twin-to-twin transfusion syndrome treated with fetoscopic laser surgery. Am J Obstet Gynecol. 2007; 196: 231. e1–4.Google Scholar
Dickinson, JE, Duncombe, GJ, Evans, SF, French, NP, Hagan, R. The long term neurologic outcome of children from pregnancies complicated by twin-to-twin transfusion syndrome. BJOG. 2005; 112: 63–8.Google Scholar
Lenclen, R, Ciarlo, G, Paupe, A, Bussieres, L, Ville, Y. Neurodevelopmental outcome at 2 years in children born preterm treated by amnioreduction or fetoscopic laser surgery for twin-to-twin transfusion syndrome: comparison with dichorionic twins. Am J Obstet Gynecol. 2009; 201: 291. e1–5.Google Scholar
Li, X, Morokuma, S, Fukushima, K, et al. Prognosis and long-term neurodevelopmental outcome in conservatively treated twin-to-twin transfusion syndrome. BMC Pregnancy Childbirth. 2011; 11: 32.Google Scholar
De Lia, JE, Kuhlmann, RS, Lopez, KP. Treating previable twin-twin transfusion syndrome with fetoscopic laser surgery: outcomes following the learning curve. J Perinat Med. 1999; 27: 61–7.Google Scholar
Sutcliffe, AG, Sebire, NJ, Pigott, AJ, et al. Outcome for children born after in utero laser ablation therapy for severe twin-to-twin transfusion syndrome. BJOG. 2001; 108: 1246–50.Google Scholar
Banek, CS, Hecher, K, Hackeloer, BJ, Bartmann, P. Long-term neurodevelopmental outcome after intrauterine laser treatment for severe twin-twin transfusion syndrome. Am J Obstet Gynecol. 2003; 188: 876–80.Google Scholar
Graef, C, Ellenrieder, B, Hecher, K, et al. Long-term neurodevelopmental outcome of 167 children after intrauterine laser treatment for severe twin-twin transfusion syndrome. Am J Obstet Gynecol. 2006; 194: 303–8.Google Scholar
Gray, PH, Poulsen, L, Gilshenan, K, et al. Neurodevelopmental outcome and risk factors for disability for twin-twin transfusion syndrome treated with laser surgery. Am J Obstet Gynecol. 2011; 204: 159. e1–6.Google Scholar
Chang, YL, Chao, AS, Chang, SD, et al. The neurological outcomes of surviving twins in severe twin-twin transfusion syndrome treated by fetoscopic laser photocoagulation at a newly established center. Prenat Diagn. 2012; 32: 893–6.Google Scholar
Graeve, P, Banek, C, Stegmann-Woessner, G, et al. Neurodevelopmental outcome at 6 years of age after intrauterine laser therapy for twin-twin transfusion syndrome. Acta Paediatr. 2012; 101: 1200–5.Google Scholar
McIntosh, J, Meriki, N, Joshi, A, et al. Long term developmental outcomes of pre-school age children following laser surgery for twin-to-twin transfusion syndrome. Early Hum Dev. 2014; 90: 837–42.Google Scholar
Vanderbilt, DL, Schrager, SM, Llanes, A, et al. Predictors of 2-year cognitive performance after laser surgery for twin-twin transfusion syndrome. Am J Obstet Gynecol. 2014; 211: 388. e1–7.Google Scholar
Sananes, N, Gabriele, V, Weingertner, AS, et al. Evaluation of long-term neurodevelopment in twin-twin transfusion syndrome after laser therapy. Prenat Diagn. 2016; 36: 1139–45.Google Scholar
van Klink, JM, Koopman, HM, van Zwet, EW, et al. Improvement in neurodevelopmental outcome in survivors of twin-twin transfusion syndrome treated with laser surgery. Am J Obstet Gynecol. 2014; 210: 540. e1–7.Google Scholar
Adegbite, AL, Castille, S, Ward, S, Bajoria, R. Neuromorbidity in preterm twins in relation to chorionicity and discordant birth weight. Am J Obstet Gynecol. 2004; 190: 156–63.Google Scholar
Hack, KE, Koopman-Esseboomet, C, Derks, JB, et al. Long-term neurodevelopmental outcome of monochorionic and matched dichorionic twins. PLoS ONE. 2009; 4: e6815.Google Scholar
Halling, C, Malone, FD, Breathnach, FM, et al. Neuro-developmental outcome of a large cohort of growth discordant twins. Eur J Pediatr. 2016; 175: 381–9.Google Scholar
Vedel, C, Oldenburg, A, Worda, K, et al. Short- and long-term perinatal outcome in twin pregnancies affected by weight discordance. Acta Obstet Gynecol Scand. 2017; 96: 233–42.Google Scholar
Rustico, MA, Consonni, D, Lanna, M, et al. Selective intrauterine growth restriction in monochorionic twins: changing patterns in umbilical artery Doppler flow and outcomes. Ultrasound Obstet Gynecol. 2017; 49: 387–93.Google Scholar
Lanna, MM, Rustico, MA, Dell’Avanzo, M, et al. Bipolar cord coagulation for selective feticide in complicated monochorionic twin pregnancies: 118 consecutive cases at a single center. Ultrasound Obstet Gynecol. 2012; 39: 407–13.Google Scholar

References

Coutinho Nunes, F, Domingues, AP, Vide Tavares, M, Belo, A, Ferreira, C, Fonseca, E, et al. Monochorionic versus dichorionic twins: are obstetric outcomes always different? J Obstet Gynaecol (Lahore). 2016; 36: 598601.Google Scholar
Glinianaia, SV, Obeysekera, MA, Sturgiss, S, Bell, R. Stillbirth and neonatal mortality in monochorionic and dichorionic twins: a population-based study. Hum Reprod. 2011; 26: 2549–57.Google Scholar
Khalil, A, Rodgers, M, Baschat, A, Bhide, A, Gratacos, E, Hecher, K, et al. ISUOG practice guidelines: role of ultrasound in twin pregnancy. Ultrasound Obstet Gynecol. 2016; 47: 247–63.Google Scholar
National Institute for Health and Care Excellence. (2011). Multiple pregnancy: antenatal care for twin and triplet pregnancies. Clinical Guideline CG129. www.nice.org.uk/guidance/cg129Google Scholar
Morin, L, Lim, K, Bly, S, Butt, K, Cargill, YM, Davies, G, et al. Ultrasound in twin pregnancies. J Obstet Gynaecol Canada. 2011; 33: 643–56.Google Scholar
Committee on Practice Bulletins—Obstetrics, Society for Maternal–Fetal Medicine. Practice bulletin 169: multifetal gestations: twin, triplet and higher-order multifetal pregnancies. Obstet Gynecol. 2016; 128: e131–46.Google Scholar
Chervenak, FA, Skupski, DW, Romero, R, Myers, MK, Smith-Levitin, M, Rosenwaks, Z, et al. How accurate is fetal biometry in the assessment of fetal age? Am J Obstet Gynecol. 1998; 178: 678–87.Google Scholar
Khalil, A, D’Antonio, F, Dias, T, Cooper, D, Thilaganathan, B, Southwest Thames Obstetric Research Collaborative (STORK). Ultrasound estimation of birth weight in twin pregnancy: comparison of biometry algorithms in the STORK multiple pregnancy cohort. Ultrasound Obstet Gynecol. 2014; 44: 210–20.Google Scholar
Kalafat, E, Sebghati, M, Thilaganathan, B, Khalil, A, Southwest Thames Obstetric Research Collaborative (STORK). Predictive accuracy of the Southwest Thames Obstetric Research Collaborative (STORK) chorionicity-specific twin growth charts for stillbirth: a validation study. Ultrasound Obstet Gynecol. 2019; 53: 193–9.Google Scholar
Stirrup, OT, Khalil, A, D’Antonio, F, Thilaganathan, B, Southwest Thames Obstetric Research Collaborative (STORK). Fetal growth reference ranges in twin pregnancy: analysis of the Southwest Thames Obstetric Research Collaborative (STORK) multiple pregnancy cohort. Ultrasound Obstet Gynecol. 2015; 45: 301–7.Google Scholar
Gielen, M, Lindsey, PJ, Derom, C, Loos, RJF, Souren, NY, Paulussen, ADC, et al. Twin-specific intrauterine ‘growth’ charts based on cross-sectional birthweight data. Twin Res Hum Genet. 2008; 11: 224–35.Google Scholar
Odibo, AO, Cahill, AG, Goetzinger, KR, Harper, LM, Tuuli, MG, Macones, GA. Customized growth charts for twin gestations to optimize identification of small-for-gestational age fetuses at risk of intrauterine fetal death. Ultrasound Obs Gynecol. 2013; 41: 637–42.Google Scholar
Khalil, AA, Khan, N, Bowe, S, Familiari, A, Papageorghiou, A, Bhide, A, et al. Discordance in fetal biometry and Doppler are independent predictors of the risk of perinatal loss in twin pregnancies. Am J Obstet Gynecol. 2015; 213: 222. e1–222. e10.Google Scholar
Harper, LM, Weis, MA, Odibo, AO, Roehl, KA, Macones, GA, Cahill, AG. Significance of growth discordance in appropriately grown twins. Am J Obstet Gynecol. 2013; 208: 15.Google Scholar
Chauhan, SP, Shields, D, Parker, D, Sanderson, M, Scardo, JA, Magann, EF. Detecting fetal growth restriction or discordant growth in twin gestations stratified by placental chorionicity. J Reprod Med. 2004; 49: 279–84.Google Scholar
Lewi, L, Jani, J, Blickstein, I, Huber, A, Gucciardo, L, Van Mieghem, T, et al. The outcome of monochorionic diamniotic twin gestations in the era of invasive fetal therapy: a prospective cohort study. Am J Obstet Gynecol. 2008; 199: 514. e1–8.Google Scholar
Gratacós, E, Ortiz, JU, Martinez, JM. A systematic approach to the differential diagnosis and management of the complications of monochorionic twin pregnancies. Fetal Diagn Ther. 2012; 32: 145–55.Google Scholar
Lewi, L, Lewi, P, Diemert, A, Jani, J, Gucciardo, L, Van Mieghem, T, et al. The role of ultrasound examination in the first trimester and at 16 weeks’ gestation to predict fetal complications in monochorionic diamniotic twin pregnancies. Am J Obstet Gynecol. 2008; 199: 493. e1–493. e7.Google Scholar
Mackie, FL, Hall, MJ, Morris, RK, Kilby, MD. Early prognostic factors of outcomes in monochorionic twin pregnancy: systematic review and meta-analysis. Am J Obstet Gynecol. 2018; 219: 436–46.Google Scholar
Mackie, FL, Morris, RK, Kilby, MD. The prediction, diagnosis and management of complications in monochorionic twin pregnancies: the OMMIT (Optimal Management of Monochorionic Twins) study. BMC Pregnancy Childbirth. 2017; 17: 153.Google Scholar
Cambiaso, O, Zhao, D-P, Abasolo, JI, Aiello, HA, Oepkes, D, Lopriore, E, et al. Discordance of cord insertions as a predictor of discordant fetal growth in monochorionic twins. Placenta. 2016; 47: 81–5.Google Scholar
Memmo, A, Dias, T, Mahsud-Dornan, S, Papageorghiou, AT, Bhide, A, Thilaganathan, B. Prediction of selective fetal growth restriction and twin-to-twin transfusion syndrome in monochorionic twins. BJOG. 2012; 119: 417–21.Google Scholar
D’Antonio, F, Khalil, A, Pagani, G, Papageorghiou, AT, Bhide, A, Thilaganathan, B. Crown-rump length discordance and adverse perinatal outcome in twin pregnancies: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2014; 44: 138–46.Google Scholar
D’Antonio, F, Khalil, A, Mantovani, E, Thilaganathan, B, Hamid, R, Gandhi, H, et al. Embryonic growth discordance and early fetal loss: the STORK multiple pregnancy cohort and systematic review. Hum Reprod. 2013; 28: 2621–7.Google Scholar
Kagan, KO, Gazzoni, A, Sepulveda-Gonzalez, G, Sotiriadis, A, Nicolaides, KH. Discordance in nuchal translucency thickness in the prediction of severe twin-to-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2007; 29: 527–32.Google Scholar
Queirós, A, Blickstein, I, Valdoleiros, S, Felix, N, Cohen, A, Simões, T. Prediction of birth weight discordance from fetal weight estimations at 21–24 weeks’ scans in monochorionic and dichorionic twins. J Matern Neonatal Med. 2017; 30: 1944–7.Google Scholar
Breathnach, FM, McAuliffe, FM, Geary, M, Daly, S, Higgins, JR, Dornan, J, et al. Definition of intertwin birth weight discordance. Obstet Gynecol. 2011; 118: 94103.Google Scholar
Bekhit, MT, Greenwood, PA, Warren, R, Aarons, E, Jauniaux, E. In utero treatment of severe fetal anaemia due to parvovirus B19 in one fetus in a twin pregnancy—a case report and literature review. Fetal Diagn Ther. 2009; 25: 153–7.Google Scholar
De la Calle, M, Baquero, F, Rodriguez, R, González, M, Fernández, A, Omeñaca, F, et al. Successful treatment of intrauterine cytomegalovirus infection with an intraventricular cyst in a dichorionic diamniotic twin gestation using cytomegalovirus immunoglobulin. J Matern Neonatal Med. 2017; 6: 14.Google Scholar
Yinon, Y, Yagel, S, Tepperberg-Dikawa, M, Feldman, B, Schiff, E, Lipitz, S. Prenatal diagnosis and outcome of congenital cytomegalovirus infection in twin pregnancies. BJOG. 2006; 113: 295300.Google Scholar
Linden, VV, Linden, HV Jr., Leal, MC, Rolim, ELF, Linden, AV, Aragão, MFVV, et al. Discordant clinical outcomes of congenital Zika virus infection in twin pregnancies. Arq Neuropsiquiatr. 2017; 75: 381–6.Google Scholar
Hall, JG. Twinning. Lancet. 2003; 362: 735–43.Google Scholar
McFadden, P, Smithson, S, Massaro, R, Huang, J, Prado, GT, Shertz, W. Monozygotic Twins Discordant for Trisomy 13. Pediatr Dev Pathol. 2017; 20: 340–7.Google Scholar
Lu, J, Cheng, YKY, Ting, YH, Law, KM, Leung, TY. Pitfalls in assessing chorioamnionicity: novel observations and literature review. Am J Obstet Gynecol. 2018; 219: 242–54.Google Scholar
Hillman, SC, Morris, RK, Kilby, MD. Co-twin prognosis after single fetal death: a systematic review and meta-analysis. Obstet Gynecol. 2011; 118: 928–40.Google Scholar
Gaerty, K, Greer, RM, Kumar, S. Systematic review and meta-analysis of perinatal outcomes after radiofrequency ablation and bipolar cord occlusion in monochorionic pregnancies. Am J Obstet Gynecol. 2015; 213: 637–43.Google Scholar
Lewi, L, Deprest, J, Hecher, K, Sebire, NJ, Snijders, RJ, Hughes, K, et al. The vascular anastomoses in monochorionic twin pregnancies and their clinical consequences. Am J Obstet Gynecol. 2013; 208: 1930.Google Scholar
Kalafat, E, Thilaganathan, B, Papageorghiou, A, Bhide, A, Khalil, A. The significance of placental cord insertion site in twin pregnancy. Ultrasound Obstet Gynecol. 2018; 52: 378–84.Google Scholar
Van Winden, KR, Quintero, RA, Kontopoulos, EV, Korst, LM, Llanes, A, Chmait, RH. Decreased total placental mass found in twin-twin transfusion syndrome gestations with selective growth restriction. Fetal Diagn Ther. 2016; 40: 116–22.Google Scholar
Fox, N, Rebarber, A, Klauser, C, Roman, A, Saltzman, D. Intrauterine growth restriction in twin pregnancies: incidence and associated risk factors. Am J Perinatol. 2011; 28: 267–72.Google Scholar
Khalil, A, Beune, I, Hecher, K, Wynia, K, Ganzevoort, W, Reed, K, et al. Consensus definition and essential reporting parameters of selective fetal growth restriction in twin pregnancy: a Delphi procedure. Ultrasound Obstet Gynecol. 2019; 53: 4754.Google Scholar
Inklaar, MJ, van Klink, JMM, Stolk, TT, van Zwet, EW, Oepkes, D, Lopriore, E. Cerebral injury in monochorionic twins with selective intrauterine growth restriction: a systematic review. Prenat Diagn. 2014; 34: 205–13.Google Scholar
Buca, D, Pagani, G, Rizzo, G, Familiari, A, Flacco, ME, Manzoli, L, et al. Outcome in monochorionic twin pregnancies with selective intrauterine growth restriction according to the umbilical artery Doppler pattern of the smaller twin: a systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2017; 50: 559–68.Google Scholar
Gratacós, E, Lewi, L, Muñoz, B, Acosta-Rojas, R, Hernandez-Andrade, E, Martinez, JM, et al. A classification system for selective intrauterine growth restriction in monochorionic pregnancies according to umbilical artery Doppler flow in the smaller twin. Ultrasound Obstet Gynecol. 2007; 30: 2834.Google Scholar
Rustico, MA, Consonni, D, Lanna, M, Faiola, S, Schena, V, Scelsa, B, et al. Selective intrauterine growth restriction in monochorionic twins: changing patterns in umbilical artery Doppler flow and outcomes. Ultrasound Obstet Gynecol. 2017; 49: 387–93.Google Scholar
Ishii, K, Murakoshi, T, Takahashi, Y, Shinno, T, Matsushita, M, Naruse, H, et al. Perinatal outcome of monochorionic twins with selective intrauterine growth restriction and different types of umbilical artery doppler under expectant management. Fetal Diagn Ther. 2009; 26: 157–61.Google Scholar
Gratacós, E, Antolin, E, Lewi, L, Martínez, JM, Hernandez-Andrade, E, Acosta-Rojas, R, et al. Monochorionic twins with selective intrauterine growth restriction and intermittent absent or reversed end-diastolic flow (Type III): feasibility and perinatal outcome of fetoscopic placental laser coagulation. Ultrasound Obstet Gynecol. 2008; 31: 669–75.Google Scholar
Ishii, K, Murakoshi, T, Hayashi, S, Saito, M, Sago, H, Takahashi, Y, et al. Ultrasound predictors of mortality in monochorionic twins with selective intrauterine growth restriction. Ultrasound Obstet Gynecol. 2011; 37: 22–6.Google Scholar

References

Gratacós, E, Carreras, E, Becker, J, et al. Prevalence of neurological damage in monochorionic twins with selective intrauterine growth restriction and intermittent absent or reversed end-diastolic umbilical artery flow. Ultrasound Obstet Gynecol. 2004; 24: 159–63.Google Scholar
Lewi, L, Gucciardo, L, Huber, A, et al. Clinical outcome and placental characteristics of monochorionic diamniotic twin pairs with early- and late-onset discordant growth. Am J Obstet Gynecol. 2008; 199: 511. e1–7.Google Scholar
Valsky, DV, Eixarch, E, Martinez, JM, et al. Selective intrauterine growth restriction in monochorionic twins: pathophysiology, diagnostic approach and management dilemmas. Semin Fetal Neonatal Med. 2010; 15: 342–8.Google Scholar
Valsky, DV, Eixarch, E, Martinez, JM, et al. Selective intrauterine growth restriction in monochorionic diamniotic twin pregnancies. Prenat Diagn. 2010; 30: 719–26.Google Scholar
Sebire, NJ, Snijders, RJ, Hughes, K, et al. The hidden mortality of monochorionic twin pregnancies. BJOG. 1997; 104: 1203–7.Google Scholar
Ishii, K, Murakoshi, T, Takahashi, Y, et al. Perinatal outcome of monochorionic twins with selective intrauterine growth restriction and different types of umbilical artery Doppler under expectant management. Fetal Diagn Ther. 2009; 26: 157–61.Google Scholar
Victoria, A, Mora, G, Arias, F. Perinatal outcome, placental pathology, and severity of discordance in monochorionic and dichorionic twins. Obstet Gynecol. 2001; 97: 310–15.Google Scholar
Gratacós, E, Lewi, L, Munoz, B, et al. A classification system for selective intrauterine growth restriction in monochorionic pregnancies according to umbilical artery Doppler flow in the smaller twin. Ultrasound Obstet Gynecol. 2007; 30: 2834.Google Scholar
Ortibus, E, Lopriore, E, Deprest, J, et al. The pregnancy and long-term neurodevelopmental outcome of monochorionic diamniotic twin gestations: a multicenter prospective cohort study from the first trimester onward. Am J Obstet Gynecol. 2009; 200: 494. e1–8.Google Scholar
Chang, YL, Chang, SD, Chao, AS, et al. Clinical outcome and placental territory ratio of monochorionic twin pregnancies and selective intrauterine growth restriction with different types of umbilical artery Doppler. Prenat Diagn. 2009; 29: 253–6.Google Scholar
Fick, AL, Feldstein, VA, Norton, ME, et al. Unequal placental sharing and birth weight discordance in monochorionic diamniotic twins. Am J Obstet Gynecol. 2006; 195: 178–83.Google Scholar
Lewi, L, Cannie, M, Blickstein, I, et al. Placental sharing, birthweight discordance, and vascular anastomoses in monochorionic diamniotic twin placentas. Am J Obstet Gynecol. 2007; 197: 587. e1–8.Google Scholar
Denbow, ML, Cox, P, Taylor, M, et al. Placental angioarchitecture in monochorionic twin pregnancies: relationship to fetal growth, fetofetal transfusion syndrome, and pregnancy outcome. Am J Obstet Gynecol. 2000; 182: 417–26.Google Scholar
Machin, GA.Velamentous cord insertion in monochorionic twin gestation. An added risk factor. J Reprod Med. 1997; 42: 785–8.Google Scholar
Hack, KE, Nikkels, PG, Koopman-Esseboom, C, et al. Placental characteristics of monochorionic diamniotic twin pregnancies in relation to perinatal outcome. Placenta. 2008: 29: 976–81.Google Scholar
Gratacós, E, Lewi, L, Carreras, E, et al. Incidence and characteristics of umbilical artery intermittent absent and/or reversed end-diastolic flow in complicated and uncomplicated monochorionic twin pregnancies. Ultrasound Obstet Gynecol. 2004; 23: 456–60.Google Scholar
Vanderheyden, TM, Fichera, A, Pasquini, L, et al. Increased latency of absent end diastolic flow in the umbilical artery of monochorionic twin fetuses. Ultrasound Obstet Gynecol. 2005; 26: 44–9.Google Scholar
Khalil, A, Rodgers, M, Baschat, A, Bhide, A, Gratacós, E, Hecher, K, et al. ISUOG Practice Guidelines: role of ultrasound in twin pregnancy. Ultrasound Obstet Gynecol. 2016; 47: 247–63.Google Scholar
Bejar, R, Vigliocco, G, Gramajo, H, et al. Antenatal origin of neurologic damage in newborn infants. II. Multiple gestations. Am J Obstet Gynecol. 1990; 162: 1230–6.Google Scholar
Bennasar, M, Eixarch, E, Martínez, JM et al. Selective intrauterine growth restriction in monochorionic diamniotic twin pregnancies. Semin Fetal Neonatal Med. 2017; 22: 376–82.Google Scholar
Buca, D, Pagani, G, Rizzo, G, et al. Outcome in monochorionic twin pregnancies with selective intrauterine growth restriction according to the umbilical artery Doppler pattern of the smaller twin: a systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2017; 50: 559–68.Google Scholar
Gratacós, E, Van Schoubroeck, D, Carreras, E, et al. Impact of laser coagulation in severe twin-twin transfusion syndrome on fetal Doppler indices and venous blood flow volume. Ultrasound Obstet Gynecol. 2002; 20: 125–30.Google Scholar
Hecher, K, Jauniaux, E, Campbell, S, et al. Artery-to-artery anastomosis in monochorionic twins. Am J Obstet Gynecol. 1994; 171: 570–2.Google Scholar
Wee, LY, Taylor, MJ, Vanderheyden, T, et al. Transmitted arterio-arterial anastomosis waveforms causing cyclically intermittent absent/reversed end-diastolic umbilical artery flow in monochorionic twins. Placenta. 2003; 24:772–8.Google Scholar
Gaziano, E, Gaziano, C, Brandt, D. Doppler velocimetry determined redistribution of fetal blood flow: correlation with growth restriction in diamniotic monochorionic and dizygotic twins. Am J Obstet Gynecol. 1998; 178: 1359–67.Google Scholar
Bajoria, R, Wee, LY, Anwar, S, et al. Outcome of twin pregnancies complicated by single intrauterine death in relation to vascular anatomy of the monochorionic placenta. Hum Reprod. 1999; 14: 2124–30.Google Scholar
Ullberg, U, Sandstedt, B, Lingman, G. Hyrtl’s anastomosis, the only connection between the two umbilical arteries. A study in full term placentas from AGA infants with normal umbilical artery blood flow. Acta Obstet Gynecol Scand. 2001; 80: 16.Google Scholar
Taylor, MJ, Denbow, ML, Tanawattanacharoen, S, et al. Doppler detection of arterio-arterial anastomoses in monochorionic twins: feasibility and clinical application. Hum Reprod. 2000; 15: 1632–6.Google Scholar
Inklaar, MJvan Klink, JMStolk, TT, et al. Cerebral injury in monochorionic twins with selective intrauterine growth restriction: a systematic review. Prenat Diagn. 2014; 34: 205–13.Google Scholar
Ishii, K, Murakoshi, T, Hayashi, S, et al. Ultrasound predictors of mortality in monochorionic twins with selective intrauterine growth restriction. Ultrasound Obstet Gynecol. 2011; 37: 22–6.Google Scholar
Chauhan, SP, Shields, D, Parker, D, et al. Detecting fetal growth restriction or discordant growth in twin gestations stratified by placental chorionicity. J Reprod Med. 2004; 49: 279–84.Google Scholar
Muñoz-Abellana, BHernandez-Andrade, EFigueroa-Diesel, Het al. Hypertrophic cardiomyopathy-like changes in monochorionic twin pregnancies with selective intrauterine growth restriction and intermittent absent/reversed end-diastolic flow in the umbilical artery. Ultrasound Obstet Gynecol. 2007; 30: 977–82.Google Scholar
Gardiner, HM, Matsui, H, Roughton, M, et al. Cardiac function in 10-year-old twins following different fetal therapies for twin-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2014; 43: 652–7.Google Scholar
Rossi, AC, D’Addario, V. Umbilical cord occlusion for selective feticide in complicated monochorionic twins: a systematic review of literature. Am J Obstet Gynecol. 2009; 200: 123–9.Google Scholar
Peeva, GBower, SOrosz, Let al. Endoscopic Placental Laser Coagulation in Monochorionic Diamniotic Twins with Type II Selective Fetal Growth Restriction. Fetal Diagn Ther. 2015; 38: 8693.Google Scholar
Quintero, RA, Bornick, PW, Morales, WJ, et al. Selective photocoagulation of communicating vessels in the treatment of monochorionic twins with selective growth retardation. Am J Obstet Gynecol. 2001; 185: 689–96.Google Scholar
Parra-Cordero, MBennasar, M, Martínez, JM, et al. Cord occlusion in monochorionic twins with early selective intrauterine growth restriction and abnormal umbilical artery Doppler: a consecutive series of 90 cases. Fetal Diagn Ther. 2016; 39: 186–91.Google Scholar
Gratacós, E, Antolín, E, Lewi, L, et al. Monochorionic twins with selective intrauterine growth restriction and intermittent absent or reversed end-diastolic flow (Type III): feasibility and perinatal outcome of fetoscopic placental laser coagulation. Ultrasound Obstet Gynecol. 2008; 31: 669–75.Google Scholar
Quintero, RA, Bornick, PW, Morales, WJ, et al. Selective photocoagulation of communicating vessels in the treatment of monochorionic twins with selective growth retardation. Am J Obstet Gynecol. 2001; 185: 689–96.Google Scholar
Chalouhi, GE, Marangoni, MA, Quibel, T, et al. Active management of selective intrauterine growth restriction with abnormal Doppler in monochorionic diamniotic twin pregnancies diagnosed in the second trimester of pregnancy. Prenat Diagn. 2013; 33: 109–15.Google Scholar
Yinon, Y, Ashwal, E, Weis, B, et al. Selective reduction in complicated monochorionic twins: prediction of obstetric outcome and comparison of techniques. Ultrasound Obstetric Gynecol. 2015; 46: 670–7.Google Scholar
Roman, A, Papanna, R, Johnson, A, et al. Selective reduction in complicated monochorionic pregnancies: radiofrequency ablation vs. bipolar cord coagulation. Ultrasound Obstet Gynecol. 2010; 36: 3741.Google Scholar

References

van Gemert, MJ, van den Wijngaard, JP, Vandenbussche, FP. Twin reversed arterial perfusion sequence is more common than generally accepted. Birth Defects Res A Clin Mol Teratol. 2015; 103: 641–3.Google Scholar
Van Allen, MI, Smith, DW, Shepard, TH. Twin reversed arterial perfusion (TRAP) sequence: a study of 14 twin pregnancies with acardius. Semin Perinatol. 1983; 7: 285–93.Google Scholar
Peyvandi, S, Feldstein, VA, Hirose, S, et al. Twin-reversed arterial perfusion sequence associated with decreased fetal cerebral vascular impedance. Ultrasound Obstet Gynecol. 2015; 45: 447–51.Google Scholar
Sherer, DM, Dalloul, M, Garza, M, et al. Prenatal sonographic diagnosis of acardiac twin embedded within the placenta. Ultrasound Obstet Gynecol. 2017; 52 : 120–1.Google Scholar
López-Pérez, R, Lorente, M, Martínez-Uriarte, J, et al. Twin-reversed arterial perfusion sequence in a triple monochorionic pregnancy with two direct pump fetuses results in significant cyclic Doppler waveform. Fetal Diagn Ther. 2015; 37: 157–60.Google Scholar
Lewi, L, Valencia, C, Gonzalez, E, et al. The outcome of twin reversed arterial perfusion sequence diagnosed in the first trimester. Am J Obstet Gynecol. 2010; 203: 213. e1–4.Google Scholar
Moore, TR, Gale, S, Benirschke, K. Perinatal outcome of forty-nine pregnancies complicated by acardiac twinning. Am J Obstet Gynecol. 1990; 163: 907–12.Google Scholar
Jelin, E, Hirose, S, Rand, L, et al. Perinatal outcome of conservative management versus fetal intervention for twin reversed arterial perfusion sequence with a small acardiac twin. Fetal Diagn Ther. 2010; 27: 138–41.Google Scholar
Brassard, M, Fouron, JC, Leduc, L, et al. Prognostic markers in twin pregnancies with an acardiac fetus. Obstet Gynecol. 1999; 94: 409–14.Google Scholar
Wong, AE, Sepulveda, W. Acardiac anomaly: current issues in prenatal assessment and treatment. Prenat Diagn. 2005; 25: 796806.Google Scholar
Dashe, JS, Fernandez, CO, Twickler, DM. Utility of Doppler velocimetry in predicting outcome in twin reversed-arterial perfusion sequence. Am J Obstet Gynecol. 2001; 185: 135–9.Google Scholar
Lee, H, Bebbington, M, Crombleholme, TM, North American Fetal Therapy Network. The North American Fetal Therapy Network Registry data on outcomes of radiofrequency ablation for twin-reversed arterial perfusion sequence. Fetal Diagn Ther. 2013; 33: 224–9.Google Scholar
Ichizuka, K, Hasegawa, J, Nakamura, M, et al. High-intensity focused ultrasound treatment for twin reversed arterial perfusion sequence. Ultrasound Obstet Gynecol. 2012; 40: 476–8.Google Scholar
Chaveeva, P, Poon, LC, Sotiriadis, A, et al. Optimal method and timing of intrauterine intervention in twin reversed arterial perfusion sequence: case study and meta-analysis. Fetal Diagn Ther. 2014; 35: 267–79.Google Scholar
Pagani, G, D’Antonio, F, Khalil, A, et al. Intrafetal laser treatment for twin reversed arterial perfusion sequence: cohort study and meta-analysis. Ultrasound Obstet Gynecol. 2013; 42: 614.Google Scholar
Sugibayashi, R, Ozawa, K, Sumie, M, et al. Forty cases of twin reversed arterial perfusion sequence treated with radio frequency ablation using the multistep coagulation method: a single-center experience. Prenat Diagn. 2016; 36: 437–43.Google Scholar
O’Donoghue, K, Barigye, O, Pasquini, L, et al. Interstitial laser therapy for fetal reduction in monochorionic multiple pregnancy: loss rate and association with aplasia cutis congenita. Prenat Diagn. 2008; 28: 535–43.Google Scholar
Stephenson, CD, Temming, LA, Pollack, R, et al. Microwave ablation for twin-reversed arterial perfusion sequence: a novel application of technology. Fetal Diagn Ther. 2015; 38: 3540.Google Scholar
Hecher, K, Lewi, L, Gratacós, E, et al. Twin reversed arterial perfusion: fetoscopic laser coagulation of placental anastomoses or the umbilical cord. Ultrasound Obstet Gynecol. 2006; 28: 688–91.Google Scholar

References

Evans, MI, Fletcher, JC, Zador, IE, Newton, BW, Struyk, CK, Quigg, MH. Selective first trimester termination in octuplet and quadruplet pregnancies: clinical and ethical issues. Obstet Gynecol. 1988; 71: 289–96.Google Scholar
Cohen, AB, Hanft, RS. Technology in American Health Care: Policy Direction for Effective Evaluation and Management. Ann Arbor: University of Michigan Press, 2004.Google Scholar
Evans, MI, Hanft, RS. The introduction of new technologies. ACOG Clinical Seminars. 1997; 2: 13.Google Scholar
Society for Assisted Reproductive Technology (2019). SART National Summary Report 2017. www.sartcorsonline.com/rptCSR_PublicMultYear.aspx?reportingYear=2017.Google Scholar
Martin, JA, Hamilton, BE, Osterman, MJK: Births in the United States, 2018. NCHS Data Brief #346, July 2019. https://www.cdc.gov/nchs/products/databriefs/db346.htm.Google Scholar
Centers for Disease Control and Prevention (2015). Assisted Reproductive Technology (ART). www.cdc.gov/art/reports/Google Scholar
Evans, MI, Britt, DW. Medical, ethical, and legal aspects of fetal reduction. In Schenker, JL, ed., Ethical and Legal Aspects of ART. Berlin & New York: Walter De Gruyter GmbH & Co, 2011, pp. 121130.Google Scholar
Martin, JA, Hamilton, BE, Osterman, MJK, Driscoll, AK, Drake, P. Births: Final Data for 2016. National Vital Statistics Report 67#1. Washington, DC: US Department Health and Human Services, CDC, NCHS, 2018.Google Scholar
Evans, MI, Andriole, SA, Britt, DW. Fetal Reduction – 25 years’ experience. Fetal Diagn Ther. 2014; 35: 6982.Google Scholar
Lawlor, DA, Nelson, SM. Effect of age on decisions about the number of embryos to transfer in assisted conception: a prospective study. Lancet. 2012; 379: 521–7.Google Scholar
Task Force of American College of Obstetricians and Gynecologists. Neonatal Encephalopathy and Cerebral Palsy: Defining the Pathogenesis and Pathophysiology. Washington, DC: ACOG, 2003.Google Scholar
Task Force of American College of Obstetricians and Gynecologists. Neonatal Encephalopathy and Neurologic Outcome. Washington, DC: ACOG, 2014.Google Scholar
Petterson, B, Nelson, K, Watson, L, Stanley, F. Twins, triplets, and cerebral palsy in births in Western Australia in the 1980s. BMJ. 1993; 307: 1239–43.Google Scholar
Pharoah, PO, Cooke, T. Cerebral Palsy and Multiple Births. Arch Dis Child Fetal Neonatal Ed. 1996; 75: F174–7.Google Scholar
Dimitiiou, G, Pharoah, PO, Nicolaides, KH, Greenough, A. Cerebral palsy in triplet pregnancies with and without iatrogenic reduction. Eur J Pediatr. 2004; 163: 449–51.Google Scholar
Van Naarden Braun, K, Doernberg, N, Schieve, L, Christensen, D, Goodman, A, Yeargin-Allsopp, M. Birth prevalence of cerebral palsy: a population-based study. Pediatrics. 2016; 137: e2015872.Google Scholar
Christensen, D, Van Naarden Braun, K, Doernberg, NS, Maenner, MJ, Arneson, CL, Durkin, MS, et al. Prevalence of cerebral palsy, co-occurring autism spectrum disorders, and motor functioning – Autism and Developmental Disabilities Monitoring Network, USA, 2008. Devel Med Child Neurol. 2014; 56: 5965.Google Scholar
Dahsling, MO, Anderson, GL, Irgens, L, Skranes, J, Vik, T. Risk of cerebral palsy in term-born singletons according to growth status at birth. Devel Med Child Neurol. 2014; 56: 5358.Google Scholar
Boyle, CA, Boulet, S, Schieve, LA, Cohen, RA, Blumberg, SJ, Yeargin-Allsopp, M, Visser, S, Kogan, MD. Trends in the prevalence of developmental disabilities in US children, 1997–2008. Pediatrics. 2011; 127: 1034–42.Google Scholar
St. John, EB, Nelson, KG, Oliver, SP, Bishno, RR, Goldenberg, RL. Cost of neonatal care according to gestational age at birth and survival status. Am J Obstet Gynecol. 2000; 182: 170–5.Google Scholar
Kirby, RS. Contribution of cost of preterm infants to the total cost of infant health care in the United States. Pediatrics. 2017; 140: e20172240.Google Scholar
March of Dimes (2013). Long-term health effects of premature birth. www.marchofdimes.org/complications/long-term-health-effects-of-premature-birth.aspxGoogle Scholar
Grosse, SD, Waitzman, NJ, Yang, N, Abe, K, Barfield, WD. Employer sponsored plan expenditures for infants born preterm. Pediatrics. 2017; 140: e20171078.Google Scholar
Cuevas, KD, Silver, DR, Brooten, D, Youngblut, JM, Bobo, CM. The cost of prematurity: Hospital charges at birth and frequency of rehospitalizations and acute care visits over the first year of life: a comparison by gestational age and birth weight. Am J Nurs. 2005; 105: 5664.Google Scholar
Marlow, N, Wolke, D, Bracewell, MA, Samara, M, EPICure Study Group. Neurologic and developmental disability at six years of age after extremely preterm birth. N Engl J Med. 2005; 352: 919.Google Scholar
Rosenbaum, P, Paneth, N, Leviton, A, Goldstein, M, Bax, M, Damiano, D, Dan, B, Jacobsson, B. A report: The definition and classification of cerebral palsy April 2006. Dev Med Child Neurol. 2007; 49: 814. [Corrected in Rosenbaum et al. Dev Med Child Neurol. 2007; Suppl. 109: 8–14]Google Scholar
Hack, M, Taylor, HG, Drotar, D, Schluchter, M, Cartar, L, Andreias, L, Wilson-Costello, D, Klein, N. Chronic conditions, functional limitations, and special health care needs of school-aged children born with extremely low birth weights in the 1990s. JAMA. 2008; 94: 318–25.Google Scholar
Stoll, BJ, Hansen, NI, Bell, EF, Shankaran, S, Laptook, AR, Walsh, MC, et al. Neonatal outcomes of extremely preterm infants from the NICHD Neonatal Research Network. Pediatrics. 2010; 126: 443–56.Google Scholar
Yogev, Y, Melamed, N, Bardin, R, Tenenbaum-Gavish, K, Ben-Shitrit, G, Ben-Haroush, AB. Pregnancy outcome at extremely advanced maternal age. Am J Obstet Gynecol. 2010; 203: 558. e1–7.Google Scholar
Dumez, Y, Oury, JF. Method for first trimester selective abortion in multiple pregnancy. Contrib Gynecol Obstet. 1986; 15: 50.Google Scholar
Berkowitz, RL, Lynch, L, Chitkara, U, Wilkins, IA, Mehalek, KE, Alvarez, E. Selective reduction of multiple pregnancies in the first trimester. N Engl J Med. 1988; 318: 1043–7.Google Scholar
Wapner, RJ, Davis, GH, Johnson, A. Selective reduction of multifetal pregnancies. Lancet 1990; 335: 90–3.Google Scholar
Timor-Tritsch, IE, Peisner, DB, Monteagudo, A, Lerner, JP, Sharma, S. Multifetal pregnancy reduction by transvaginal puncture: evaluation of the technique used in 134 cases. Am J Obstet Gynecol. 1993; 168: 799804.Google Scholar
Li, R, Yang, R, Chen, X, Yang, S, Ma, C, Liu, P, Qiao, J. Intracranial KCl injection – an alternative method for multifetal pregnancy reduction in the early second trimester. Fetal Diag Ther. 2013; 34: 2630.Google Scholar
Evans, MI, Dommergues, M, Wapner, RJ, Lynch, L, Dumez, Y, Goldberg, JD, et al. Efficacy of transabdominal multifetal pregnancy reduction: collaborative experience among the world’s largest centers. Obstet Gynecol. 1993; 82: 61–7.Google Scholar
Evans, MI, Drugan, A, Fletcher, JC, Platt, LD, Rodeck, CA, Hansmann, M, Bottoms, SF. Attitudes on the ethics of abortion, sex selection & selective termination among health care professionals, ethicists & clergy likely to encounter such situations. Am J Obstet Gynecol. 1991; 164: 1092–9.Google Scholar
Yaron, Y, Bryant-Greenwood, PK, Dave, N, Moldenhauer, JS, Kramer, RL, Johnson, MP, Evans, MI. Multifetal pregnancy reduction (MFPR) of triplets to twins: Comparison with non-reduced triplets and twins. Am J Obstet Gynecol. 1999; 180: 1268–71.Google Scholar
Antsaklis, A, Souka, AP, Daskalakis, G, Papantoniou, N, Koutra, P, Kavalakis, Y, Mesogitis, S. Embryo reduction versus expectant management in triplet pregnancies. J Matern Fetal Neonatal Med. 2004; 16: 219–22.Google Scholar
Luke, B, Brown, MD, Nugent, C, Gonzalez-Quintero, VH, Witter, FR, Newman, RB. Risk factors for adverse outcomes in spontaneous versus assisted conception in twin pregnancies. Fertil Steril. 2004; 81: 315–19.Google Scholar
Kozinsky, Z, Zadori, J, Orvos, H, Katona, M, Pál, A, Kovács, L. Obstetric and neonatal risk of pregnancies after assisted reproductive technology: a matched control study. Acta Obstet Gynecol Scand. 2003; 82: 850–6.Google Scholar
McDonald, S, Murphy, K, Beyene, J, Ohlsson, A. Perinatal outcomes of in vitro fertilization twins: a systematic review and meta-analysis. Am J Obstet Gynecol. 2005; 193: 141–52.Google Scholar
Lipitz, S, Shulman, A, Achiron, R, Zalel, Y, Seidman, DS. A comparative study of multifetal pregnancy reduction from triplets to twins in the first versus early second trimesters after detailed fetal screening. Ultrasound Obstet Gynecol. 2001; 18: 35–8.CrossRefGoogle ScholarPubMed
Sepulveda, W, Munoz, H, Alcalde, JL. Conjoined twins in a triplet pregnancy: early prenatal diagnosis with three-dimensional ultrasound and review of the literature. Ultrasound Obstet Gynecol. 2003; 22: 199204.Google Scholar
Evans, MI, Berkowitz, R, Wapner, R, Carpenter, R, Goldberg, J, Ayoub, MA, et al. Multifetal pregnancy reduction (MFPR): improved outcomes with increased experience. Am J Obstet Gynecol. 2001; 184: 97103.Google Scholar
Blickstein, I. How and why are triplets disadvantaged compared to twins. Best Pract Res Clin Obstet Gynecol. 2004; 18: 631–44.Google Scholar
Rosner, M, Pergament, E, Andriole, S, Gebb, J, Dar, P, Evans, MI. Detection of genetic abnormalities using CVS and FISH prior to fetal reduction in sonographically normal appearing fetuses. Prenat Diagn. 2013; 33: 940–4.Google Scholar
Chaveeva, P, Kosinski, P, Puglia, D, Poon, LC, Nicolaides, KH. Trichorionic and dichorionic triplet pregnancies at 10–14 weeks: outcome after embryo reduction compared to expectant management. Fetal Diag Ther. 2013; 34: 199205.Google Scholar
Evans, MI, Kaufman, MI, Urban, AJ, Krivchenia, EL, Britt, DW, Wapner, RJ. Fetal reduction from twins to a singleton: a reasonable consideration. Obstet Gynecol. 2004; 104: 102–9.Google Scholar
Templeton, A. The multiple gestation epidemic: the role of the assisted reproductive technologies. Am J Obstet Gynecol. 2004; 190: 894–8.Google Scholar
Kalra, SK, Milad, MP, Klock, SC, Grobman, WA. Infertility patients and their partners: differences in the desire for twin gestations. Obstet Gynecol. 2003; 102: 152–5.Google Scholar
Evans, MI, Britt, DW. Selective Reduction in Multifetal Pregnancies. In Paul, M, Grimes, D, Stubblefield, P, Borgatta, L, Lichfield, S, Creinin, M, eds., Management of Unintended and Abnormal Pregnancy. London: Blackwell-Wiley Publishing Co, 2009, pp. 312–18.Google Scholar
Evans, MI, Britt, DW. Fetal reduction: ethical and societal issues. Sem Reprod Med. 2010; 28: 295302.CrossRefGoogle Scholar
Britt, DW, Von-Voris Schoenborn, S, Jamil, S, Gebb, J, Rosner, M, Evans, MI. The impact of area conservatism on deviations from best practice: women choosing to undergo selective reduction. Intl J Health Well Soc. 2017; 7: 115–40.Google Scholar
Balasch, J, Gratacós, E. Delayed childbearing: effects on fertility and the outcome of pregnancy. Curr Opin Obstet Gynecol. 2012; 24: 187–93.Google Scholar
Balasch, J, Gratacós, E. Delayed childbearing: effects on fertility and the outcome of pregnancy. Fetal Diagn Ther. 2011; 29: 263–73.Google Scholar
McLean, LK, Evans, MI, Carpenter, RJ, Johnson, MP, Goldberg, JD. Genetic amniocentesis (AMN) following multifetal pregnancy reduction (MFPR) does not increase the risk of pregnancy loss. Prenat Diagn. 1998; 18: 186–8.Google Scholar
Wapner, RJ, Johnson, A, Davis, G, Urban, A, Morgan, P, Jackson, L. Prenatal diagnosis in twin gestations: a comparison between second-trimester amniocentesis and first-trimester chorionic villus sampling. Obstet Gynecol. 1993; 82: 4956.Google Scholar
Brambati, B, Tului, L, Baldi, M, Guercilena, S. Genetic analysis prior to selective fetal reduction in multiple pregnancy: technical aspects and clinical outcome. Hum Reprod. 1995; 10: 818–25.CrossRefGoogle ScholarPubMed
Tabor, A, Alfirevic, Z. Update on procedure-related risks for prenatal diagnosis techniques. Fetal Diagn Ther. 2010; 27: 17.CrossRefGoogle ScholarPubMed
Hern, WM. Selective termination for fetal anomaly/genetic disorder in twin pregnancy at 32+ menstrual weeks. Report of four cases. Fetal Diagn Ther. 2004; 19: 292–5.CrossRefGoogle ScholarPubMed
Evans, MI, Goldberg, J, Horenstein, J, Wapner, R, Ayoub, MA, Stone, J, et al. Selective termination (ST) for structural (STR), chromosomal (CHR), and Mendelian (MEN) anomalies: International experience. Am J Obstet Gynecol. 1999; 181: 893–7.Google Scholar
Eddleman, KA, Stone, JL, Lynch, L, Berkowitz, RL. Selective termination of anomalous fetuses in multiple pregnancies: two hundred cases at a single center. Am J Obstet Gynecol. 2002; 187: 1168–72.Google Scholar
Lu, J, Ting, YH, Law, KM, Lau, TK, Leung, TY. Radiofrequency ablation for selective reduction in complicated monochorionic multiple pregnancies. Fetal Diagn Ther. 2013; 34: 211–16.Google Scholar
Pergament, E, Schulman, JD, Copeland, K, Fine, B, Black, SH, Ginsberg, NA, Frederiksen, MC, Carpenter, RJ. The risk and efficacy of chorionic villus sampling in multiple gestations. Prenat Diagn. 1992; 12: 377–84.CrossRefGoogle ScholarPubMed
Nicolini, U, Lalatta, F, Natacci, F, Curcio, C, Bui, TH. The introduction of QF-PCR in prenatal diagnosis of fetal aneuploidies: time for reconsideration. Hum Reprod Update. 2004; 10: 541–8.Google Scholar
Wapner, RJ, Martin, CL, Levy, B, Ballif, BC, Eng, CM, Zachary, JM, et al. Chromosomal microarray versus karyotyping for prenatal diagnosis. N Engl J Med. 2012; 367: 2175–84.Google Scholar
Evans, MI, Wapner, RJ, Berkowitz, RL. Noninvasive prenatal testing or advanced diagnostic testing: caveat emptor. Am J Obstet Gynecol. 2016; 215: 298305.Google Scholar
Evans, MI, Evans, SM, Bennett, TA, Wapner, RJ. The price of abandoning testing for cell-free fetal DNA screening. Prenat Diagn. 2018; 38: 243–5.Google Scholar
Evans, MI, Andriole, S, Curtis, J, Evans, SM, Kessler, AA, Rubenstein, AF. The epidemic of abnormal copy number variants missed because of reliance upon noninvasive prenatal screening. Prenat Diagn. 2018; 38: 730–4.CrossRefGoogle ScholarPubMed
Wapner, RJ, Babiarz, JE, Levy, B, Stosic, M, Zimmermann, B, Sigurjonsoon, S, et al. Expanding the scope of noninvasive prenatal testing: detection of fetal microdeletion syndromes. Am J Obstet Gynecol. 2015: 212: 322. e1–9.Google Scholar
Dreesen, J, Destouni, A, Kourlaba, G, Degn, B, Mette, WC, Carvalho, F, et al. Evaluation of PCR-based preimplantation genetic diagnosis applied to monogenic disease: a collaborative ESHRE PGD consortium study. Eur J Hum Genet. 2013; 22: 1012–18.Google ScholarPubMed
Yang, Z, Liu, J, Collins, GS, Salem, SA, Liu, X, Lyle, SS, et al. Selection of single blactocysts for fresh transfer via standard morphology assessment alone and with array CGH for good prognosis IVF patients: results from a randomized pilot study. Mol Cytogenet. 2012; 5: 2432.Google Scholar
Dondorp, W, de Wert, G, Bombard, Y, Bianchi, DW, Bergmann, C, Borry, P, et al. Non-invasive prenatal testing for aneuploidy and beyond: challenges of responsible innovation in prenatal screening. Eur J Hum Genet. 2015; 57: 18.Google Scholar
Pantos, K, Kokkali, G, Petroutsou, K, Lekka, K, Malligiannis, P, Koratzis, A. Monochorionic triplet and monoamniotic twins gestation after intracytoplasmic sperm injection andlaser-assisted hatching. Fetal Diagn Ther. 2009; 25: 144–7.CrossRefGoogle ScholarPubMed
Peeters, SH, Evans, MI, Slaghekke, F, Klumper, FJ, Middeldorp, JM, Lopriore, E, Oepkes, D. Pregnancy complications for di-chorionic, tri-amniotic triplets: markedly increased over trichorionic and reduced cases. Am J Obstet Gynecol. 2014; 210: S288.Google Scholar
Chaveeva, P, Peeva, G, Pugliese, SG, Shterev, A, Nicolaides, KH. Intrafetal laser ablation for embryo reduction from dichorionic triplets to dichorionic twins. Ultrasound Obstet Gynecol. 2017; 50; 632–4.CrossRefGoogle ScholarPubMed
Evans, MI, Rosner, M, Andriole, S, Alkalay, A, Gebb, J, Britt, DW. Evolution of gender preferences in multiple pregnancies. Prenat Diagn. 2013; 33: 935–9.Google Scholar
Evans, MI, Andriole, S, Pergament, E, Curtis, J, Britt, DW. Paternity balancing. Fetal Diagn Ther. 2013; 33: 935–9.Google Scholar
Evans, MI, Goldberg, JD, Horenstein, J, Wapner, RJ, Ayoub, MA, Stone, J, et al. Selective termination (ST) for structural (STR), chromosomal (CHR), and Mendelian (MEN) anomalies: International experience. Am J Obstet Gynecol. 1999; 181: 893–7.Google Scholar
Hack, KE, Derks, JB, Elias, SG, Franx, A, Roos, EJ, Voerman, SK, et al. Increased perinatal mortality and morbidity in monochorionic versus dichorionic twin pregnancies: clinical implications of a large Dutch cohort study. BJOG. 2008; 115: 5867.Google Scholar
Evans, MI, Lau, TK. Making decisions when no good options exist: Delivery of the survivor after intrauterine death of the co-twin in monochorionic twin pregnancies. Fetal Diagn Ther. 2010; 28: 191–5.Google Scholar
Quintero, RA, Reich, H, Puder, KS, Bardicef, M, Evans, MI, Cotton, DB, Romero, R. Brief report: umbilical cord ligation of an acardiac twin by fetoscopy at 19 weeks of gestation. N Engl J Med. 1994; 330: 469–71.Google Scholar
Gebb, J, Rosner, M, Dar, P, Evans, MI. Long term neurologic outcomes after fetal interventions: meta-analysis Am J Obstet Gynecol. 2014; 210: S115.Google Scholar
Beauchamp, TL, Childress, JC. Principles of Biomedical Ethics, 5th edn. New York: Oxford University Press, 2001, pp. 358–9.Google Scholar
Dziadosz, M, Evans, MI. Re-thinking single embryo transfer: increased risks of monozygotic twinning, a systematic review. Fetal Diagn Ther. 2017; 42: 8191.Google Scholar
Benjamin, M. Splitting the Difference: Compromise and Integrity in Ethics and Politics. Lawrence: University Press of Kansas, 1990, pp. 72–4.Google Scholar
Britt, DW, Evans, MI. Sometimes doing the right thing sucks: Frame combinations and multifetal pregnancy reduction decision difficulty. Soc Sci Med. 2007; 65: 2342–56.Google Scholar
Britt, DW, Evans, MI: Information sharing among couples considering multifetal pregnancy reduction. Fertil Steril. 2007; 87: 490–5.CrossRefGoogle ScholarPubMed
Tavrow, P. Promote or discourage: how providers can influence service use. In Malarcher, S, ed., Social Determinants of Sexual and Reproductive Health: Informing Future Research and Programme Implementation. Geneva: World Health Organization, 2010, pp. 1736.Google Scholar
Britt, DW, Norton, J, Hubanks, A, Arias-Navidad, SG, Perkins, RJ, Lowery, C. A two period assessment of changes in specialist contact in a high risk pregnancy telemedical program. Telemed J E Health. 2006; 12: 17.CrossRefGoogle Scholar
Britt, DW, Bronstein, J, Norton, JA. Absorbing and transferring risk: a logistic regression analysis of a statewide high-risk-pregnancy telemedical program on VLBW maternal transports. BMC Pregnancy Childbirth. 2006; 6: 11.Google Scholar
Yeung, D. Social media as a catalyst for policy action and social change for health and well-being – a viewpoint. J Med Internet Res. 2018; 20: 3 e94.Google Scholar
Britt, DW, Eden, RD, Evans, MI. Matching risk and resources in high-risk pregnancies. J Matern Fetal Neonatal Med. 2006; 19: 645–50.Google Scholar

References

Khalil, A, Rodgers, M, Baschat, A, et al. ISUOG Practice Guidelines: role of ultrasound in twin pregnancy. Ultrasound Obstet Gynecol. 2016; 47: 247–63.Google Scholar
Wimalasundera, RC. Selective reduction and termination of multiple pregnancies. Semin Fetal Neonatal Med. 2010; 15: 327–35.Google Scholar
Evans, MI, Goldberg, JD, Dommergues, M, et al. Efficacy of second-trimester selective termination for fetal abnormalities: international collaborative experience among the world’s largest centers. Am J Obstet Gynecol. 1994; 171: 90–4.Google Scholar
Moore, TR, Gale, S, Benirschke, K. Perinatal outcome of forty-nine pregnancies complicated by acardiac twinning. Am J Obstet Gynecol. 1990; 163: 907–12.CrossRefGoogle ScholarPubMed
Royal College of Obstetricians & Gynaecologists. Management of Monochorionic Twin Pregnancy: Green-top Guideline No. 51. BJOG. 2017; 124: e145.Google Scholar
Healey, MG. Acardia: predictive risk factors for the co-twin’s survival. Teratology. 1994; 50: 205–13.Google Scholar
Tan, TY, Sepulveda, W. Acardiac twin: a systematic review of minimally invasive treatment modalities. Ultrasound Obstet Gynecol. 2003; 22: 409–19.Google Scholar
Wong, AE, Sepulveda, W. Acardiac anomaly: current issues in prenatal assessment and treatment. Prenatal Diagn. 2005; 25: 796806.Google Scholar
Pagani, G, D’Antonio, F, Khalil, A, et al. Intrafetal laser treatment for twin reversed arterial perfusion sequence: cohort study and meta-analysis. Ultrasound Obstet Gynecol. 2013; 42: 614.Google Scholar
Chaveeva, P, Poon, LC, Sotiriadis, A, et al. Optimal method and timing of intrauterine intervention in twin reversed arterial perfusion sequence: case study and meta-analysis. Fetal Diagn Ther. 2014; 35: 267–79.Google Scholar
Cabassa, P, Fichera, A, Prefumo, F, et al. The use of radiofrequency in the treatment of twin reversed arterial perfusion sequence: a case series and review of the literature. Eur J Obstet Gynecol Reprod Biol. 2013; 166: 127–32.Google Scholar
Emery, SP, Bahtiyar, MO, Moise, KJ. The North American Fetal Therapy Network Consensus Statement: Management of Complicated Monochorionic Gestations. Obstet Gynecol. 2015; 126: 575–84.Google Scholar
Buca, D, Pagani, G, Rizzo, G, et al. Outcome of monochorionic twin pregnancy with selective intrauterine growth restriction according to umbilical artery Doppler flow pattern of smaller twin: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2017; 50: 559–68.Google Scholar
Khalil, A, Beune, I, Hecher, K, et al. Consensus definition and essential reporting parameters of selective fetal growth restriction in twin pregnancy: a Delphi procedure. Ultrasound Obstet Gynecol. 2019; 53: 4754.Google Scholar
Gratacós, E, Lewi, L, Munoz, B, et al. A classification system for selective intrauterine growth restriction in monochorionic pregnancies according to umbilical artery Doppler flow in the smaller twin. Ultrasound Obstet Gynecol. 2007; 30: 2834.Google Scholar
Bennasar, M, Eixarch, E, Martinez, JM, et al. Selective intrauterine growth restriction in monochorionic diamniotic twin pregnancies. Semin Fetal Neonatal Med. 2017; 22: 376–82.Google Scholar
Roberts, D, Neilson, JP, Kilby, MD, et al. Interventions for the treatment of twin-twin transfusion syndrome. Cochrane Database Syst Rev. 2014; 1: CD002073.Google Scholar
Lopriore, E, Middeldorp, JM, Oepkes, D, et al. Twin anemia-polycythemia sequence in two monochorionic twin pairs without oligo-polyhydramnios sequence. Placenta. 2007; 28: 4751.Google Scholar
Slaghekke, F, Favre, R, Peeters, SH, et al. Laser surgery as a management option for twin anemia-polycythemia sequence. Ultrasound Obstet Gynecol. 2014; 44: 304–10.Google Scholar
Vojtech, J, Haslik, L, Pock, R, et al. Selective feticide in monochorionic twin pregnancies with discordant fetal anomalies: management and outcome. Ceska Gynekol. 2017; 82: 345–50.Google ScholarPubMed
Glinianaia, SV, Rankin, J, Khalil, A, et al. Prevalence, antenatal management and perinatal outcomes of monochorionic monoamniotic twin pregnancies: a collaborative multicentre study in England, 2000–2013. Ultrasound Obstet Gynecol. 2019; 53: 184–92.Google Scholar
Middeldorp, JM, Klumper, FJ, Oepkes, D, et al. Selective feticide in monoamniotic twin pregnancies by umbilical cord occlusion and transection. Fetal Diagn Ther. 2008; 23: 121–5.Google Scholar
Rossi, AC, Prefumo, F. Impact of cord entanglement on perinatal outcome of monoamniotic twins: a systematic review of the literature. Ultrasound Obstet Gynecol. 2013; 41: 131–5.Google Scholar
Geipel, A, Berg, C, Katalinic, A, et al. Prenatal diagnosis and obstetric outcomes in triplet pregnancies in relation to chorionicity. BJOG. 2005; 112: 554–8.Google Scholar
Chaveeva, P, Kosinski, P, Birdir, C, et al. Embryo reduction in dichorionic triplets to dichorionic twins by intrafetal laser. Fetal Diagn Ther. 2014; 35: 83–6.Google Scholar
Chaveeva, P, Peeva, G, Pugliese, SG, et al. Intrafetal laser ablation for embryo reduction from dichorionic triplets to dichorionic twins. Ultrasound Obstet Gynecol. 2017; 50: 632–4.CrossRefGoogle ScholarPubMed
Abel, JS, Flock, A, Berg, C, et al. Expectant management versus multifetal pregnancy reduction in higher order multiple pregnancies containing a monochorionic pair and a review of the literature. Arch Gynecol Obstet. 2016; 294: 1167–73.Google Scholar
Morlando, M, Ferrara, L, D’Antonio, F, et al. Dichorionic triplet pregnancies: risk of miscarriage and severe preterm delivery with fetal reduction versus expectant management. Outcomes of a cohort study and systematic review. BJOG. 2015; 122: 1053–60.Google Scholar
Yinon, Y, Ashwal, E, Weisz, B, et al. Selective reduction in complicated monochorionic twins: prediction of obstetric outcome and comparison of techniques. Ultrasound Obstet Gynecol. 2015; 46: 670–7.Google Scholar
Ichizuka, K, Hasegawa, J, Nakamura, M, et al. High-intensity focused ultrasound treatment for twin reversed arterial perfusion sequence. Ultrasound Obstet Gynecol. 2012; 40: 476–8.CrossRefGoogle ScholarPubMed
Okai, T, Ichizuka, K, Hasegawa, J, et al. First successful case of non-invasive in-utero treatment of twin reversed arterial perfusion sequence by high-intensity focused ultrasound. Ultrasound Obstet Gynecol. 2013; 42: 112–14.Google Scholar
Deprest, JA, Audibert, F, Van Schoubroeck, D, et al. Bipolar coagulation of the umbilical cord in complicated monochorionic twin pregnancy. Am J Obstet Gynecol. 2000; 182: 340–5.Google Scholar
Lanna, MM, Rustico, MA, Dell’Avanzo, M, et al. Bipolar cord coagulation for selective feticide in complicated monochorionic twin pregnancies: 118 consecutive cases at a single center. Ultrasound Obstet Gynecol. 2012; 39: 407–13.Google Scholar
Bebbington, MW, Danzer, E, Moldenhauer, J, et al. Radiofrequency ablation vs bipolar umbilical cord coagulation in the management of complicated monochorionic pregnancies. Ultrasound Obstet Gynecol. 2012; 40: 319–24.Google Scholar
Tsao, K, Feldstein, VA, Albanese, CT, et al. Selective reduction of acardiac twin by radiofrequency ablation. Am J Obstet Gynecol. 2002; 187: 635–40.Google Scholar
Paramasivam, G, Wimalasundera, R, Wiechec, M, et al. Radiofrequency ablation for selective reduction in complex monochorionic pregnancies. BJOG. 2010. 117: 1294–8.Google Scholar
Roman, A, Papanna, R, Johnson, A, et al. Selective reduction in complicated monochorionic pregnancies: radiofrequency ablation vs. bipolar cord coagulation. Ultrasound Obstet Gynecol. 2010; 36: 3741.Google Scholar
Bebbington, M. Selective reduction in multiple gestations. Best Pract Res Clin Obstet Gynaecol. 2014; 28: 239–47.Google Scholar
Bebbington, M. Selective reduction in complex monochorionic gestations. Am J Perinatol. 2014; 31 (Suppl. 1): S51–8.Google Scholar
Lu, J, Ting, YH, Law, KM, et al. Radiofrequency ablation for selective reduction in complicated monochorionic multiple pregnancies. Fetal Diagn Ther. 2013; 34: 211–16.Google Scholar
Klaritsch, P, Albert, K, Van Mieghem, T, et al. Instrumental requirements for minimal invasive fetal surgery. BJOG. 2009; 116: 188–97.Google Scholar
O’Donoghue, K, Barigye, O, Pasquini, L, et al. Interstitial laser therapy for fetal reduction in monochorionic multiple pregnancy: loss rate and association with aplasia cutis congenita. Prenat Diagn. 2008; 28: 535–43.Google ScholarPubMed
Prefumo, F, Cabassa, P, Fichera, A, et al. Preliminary experience with microwave ablation for selective feticide in monochorionic twin pregnancies. Ultrasound Obstet Gynecol. 2013; 41: 470–1.Google Scholar
Prefumo, F, Cabassa, P, Fichera, A, et al. Microwave ablation in complicated monochorionic twin pregnancies. Fetal Diagn Ther. 2015; 38: 159.Google Scholar
Stephenson, CD, Temming, LA, Pollack, R, et al. Microwave ablation for twin-reversed arterial perfusion sequence: a novel application of technology. Fetal Diagn Ther. 2015; 38: 3540.Google Scholar
Hillman, SC, Morris, RK, Kilby, MD. Co-twin prognosis after single fetal death: a systematic review and meta-analysis. Obstet Gynecol. 2011; 118: 928–40.Google Scholar
Ong, SS, Zamora, J, Khan, KS, et al. Prognosis for the co-twin following single-twin death: a systematic review. BJOG. 2006; 113: 992–8.Google Scholar
van Klink, JM, Koopman, HM, Oepkes, D, et al. Long-term neurodevelopmental outcome in monochorionic twins after fetal therapy. Early Hum Dev. 2011; 87: 601–6.Google Scholar
Panciatici, M, Tosello, B, Blanc, J, et al. Newborn outcomes after radiofrequency ablation for selective reduction in the complicated monochorionic pregnancies. Gynecol Obstet Fertil Senol. 2017; 45: 197201.Google Scholar
Lewi, L, Gratacós, E, Ortibus, E, et al. Pregnancy and infant outcome of 80 consecutive cord coagulations in complicated monochorionic multiple pregnancies. Am J Obstet Gynecol. 2006; 194: 782–9.Google Scholar
Kumar, S, Paramasivam, G, Zhang, E, et al. Perinatal- and procedure-related outcomes following radiofrequency ablation in monochorionic pregnancy. Am J Obstet Gynecol. 2014; 210: 454. e1–6.Google Scholar
Lee, H, Bebbington, M, Crombleholme, TM. The North American Fetal Therapy Network Registry data on outcomes of radiofrequency ablation for twin-reversed arterial perfusion sequence. Fetal Diagn Ther. 2013; 33: 224–9.Google Scholar
Schou, KV, Jensen, LN, Jørgensen, C, et al. Ultrasound-guided bipolar umbilical cord occlusion in complicated monochorionic pregnancies: is there a learning curve. Fetal Diagn Ther. 2018; 44: 6571.Google Scholar
Gaerty, K, Greer, RM, Kumar, S. Systematic review and meta-analysis of perinatal outcomes after radiofrequency ablation and bipolar cord occlusion in monochorionic pregnancies. Am J Obstet Gynecol. 2015; 213: 637–43.Google Scholar
Peeters, SH, Devlieger, R, Middeldorp, JM, DeKoninck, P, Deprest, J, Lopriore, E, et al. Fetal surgery in complicated monoamniotic pregnancies: case series and systematic review of the literature. Prenat Diagn. 2014; 34: 586–91.Google Scholar

References

Grandjean, H, Larroque, D, Levi, S. The performance of routine ultrasonographic screening of pregnancies in the Eurofetus Study. Am J Obstet Gynecol. 1999; 181: 446–54.Google Scholar
Lewis, MA. Demography of renal disease in childhood. Semin Fetal Neonatal Med. 2008; 13: 118–24.Google Scholar
Hamilton, AJ, Braddon, F, Casula, A, Lewis, M, Mallett, T, Marks, SD, Shenoy, M, Sinha, MD, Tse, Y, Maxwell, H. UK Renal Registry 19th Annual Report: Chapter 4 Demography of the UK Paediatric Renal Replacement Therapy Population in 2015. Nephron. 2017; 137: 103–16.Google Scholar
Freedman, AL, Johnson, MP, Gonzalez, R. Fetal therapy for obstructive uropathy: past, present, future? Pediatr Nephrol. 2000; 14: 167–76.Google Scholar
Parkhouse, HF, Barratt, TM, Dillon, MJ, Duffy, PG, Fay, J, Ransley, PG, Woodhouse, CR, Williams, DI. Long-term outcome of boys with posterior urethral valves. Br J Urol. 1988; 62: 5962.Google Scholar
Anumba, DO, Scott, JE, Plant, ND, Robson, SC. Diagnosis and outcome of fetal lower urinary tract obstruction in the northern region of England. Prenat Diagn. 2005; 25: 713.Google Scholar
Malin, G, Tonks, AM, Morris, RK, Gardosi, J, Kilby, MD. Congenital lower urinary tract obstruction: a population-based epidemiological study. BJOG. 2012; 119: 1455–64.Google Scholar
Taghavi, K, Sharpe, C, Stringer, MD. Fetal megacystis: a systematic review. J Pediatr Urol. 2017; 13: 715.Google Scholar
Liao, AW, Sebire, NJ, Geerts, L, Cicero, S, Nicolaides, KH. Megacystis at 10–14 weeks of gestation: chromosomal defects and outcome according to bladder length. Ultrasound Obstet Gynecol. 2003; 21: 338–41.Google Scholar
Boghossian, NS, Sicko, RJ, Kay, DM, Rigler, SL, Caggana, M, Tsai, MY, et al. Rare copy number variants implicated in posterior urethral valves. Am J Med Genet A. 2016; 170: 622–33.Google Scholar
Richer, J, Milewicz, DM, Gow, R, de Nanassy, J, Maharajh, G, Miller, E, Oppenheimer, L, Weiler, G, O’Connor, M. R179H mutation in ACTA2 expanding the phenotype to include prune-belly sequence and skin manifestations. Am J Med Genet A. 2012; 158A: 664–8.Google Scholar
Boghossian, NS, Sicko, RJ, Giannakou, A, Dimopoulos, A, Caggana, M, Tsai, MY, et al. Rare copy number variants identified in prune belly syndrome. Eur J Med Genet. 2018; 61: 145–51.Google Scholar
De Sousa, J, Upadhyay, V, Stone, P. Megacystis microcolon intestinal hypoperistalsis syndrome: case reports and discussion of the literature. Fetal Diagn Ther. 2016; 39: 152–7.Google Scholar
Weber, S, Thiele, H, Mir, S, Toliat, MR, Sozeri, B, Reutter, H, et al. Muscarinic acetylcholine receptor M3 mutation causes urinary bladder disease and a prune-belly-like syndrome. Am J Hum Genet. 2011; 89: 668–74.Google Scholar
Williams, O, Hutchings, G, Hubinont, C, Debauche, C, Greenough, A. Pulmonary effects of prolonged oligohydramnios following mid-trimester rupture of the membranes – antenatal and postnatal management. Neonatology. 2012; 101: 8390.Google Scholar
Harrison, MR, Ross, N, Noall, R, de Lorimier, AA. Correction of congenital hydronephrosis in utero. I. The model: fetal urethral obstruction produces hydronephrosis and pulmonary hypoplasia in fetal lambs. J Pediatr Surg. 1983; 18: 247–56.Google Scholar
Harrison, MR, Nakayama, DK, Noall, R, de Lorimier, AA. Correction of congenital hydronephrosis in utero. II. Decompression reverses the effects of obstruction on the fetal lung and urinary tract. J Pediatr Surg. 1982; 17: 965–74.Google Scholar
Pringle, KC, Kitagawa, H, Seki, Y, Koike, J, Zuccollo, J. Development of an animal model to study congenital urinary obstruction. Pediatr Surg Int. 2013; 29: 1083–9.Google Scholar
McHugo, J, Whittle, M. Enlarged fetal bladders: aetiology, management and outcome. Prenat Diagn. 2001; 21: 958–63.Google Scholar
Kagan, KO, Staboulidou, I, Syngelaki, A, Cruz, J, Nicolaides, KH. The 11–13-week scan: diagnosis and outcome of holoprosencephaly, exomphalos and megacystis. Ultrasound Obstet Gynecol. 2010; 36: 1014.Google Scholar
Fontanella, F, Duin, L, Adama van Scheltema, PN, Cohen-Overbeek, TE, Pajkrt, E, Bekker, M, Willekes, C, Bax, CJ, Bilardo, CM. Fetal megacystis: prediction of spontaneous resolution and outcome. Ultrasound Obstet Gynecol. 2017; 50: 458–63.Google Scholar
Newman, J, Antonakopoulos, GN. The fine structure of the human fetal urinary bladder. Development and maturation. A light, transmission and scanning electron microscopic study. J Anat. 1989; 166: 135–50.Google Scholar
Robyr, R, Benachi, A, Daikha-Dahmane, F, Martinovich, J, Dumez, Y, Ville, Y. Correlation between ultrasound and anatomical findings in fetuses with lower urinary tract obstruction in the first half of pregnancy. Ultrasound Obstet Gynecol. 2005; 25: 478–82.Google Scholar
Fievet, L, Faure, A, Coze, S, Harper, L, Panait, N, Braunstein, D, Carson, J, Gorincour, G, Chaumoitre, K, Guys, JM, Alessandrini, P, D’Ercole, C, Merrot, T. Fetal megacystis: etiologies, management, and outcome according to the trimester. Urology. 2014; 84: 185–90.Google Scholar
Montemarano, H, Bulas, DI, Rushton, HG, Selby, D. Bladder distention and pyelectasis in the male fetus: causes, comparisons, and contrasts. J Ultrasound Med. 1998; 17: 743–9.Google Scholar
Chitrit, Y, Bourdon, M, Korb, D, Grapin-Dagorno, C, Joinau-Zoulovits, F, Vuillard, E, et al. Posterior urethral valves and vesicoureteral reflux: can prenatal ultrasonography distinguish between these two conditions in male fetuses? Prenat Diagn. 2016; 36: 831–7.Google Scholar
Bernardes, LS, Aksnes, G, Saada, J, Masse, V, Elie, C, Dumez, Y, Lortat-Jacob, SL, Benachi, A. Keyhole sign: how specific is it for the diagnosis of posterior urethral valves? Ultrasound Obstet Gynecol. 2009; 34: 419–23.Google Scholar
Fontanella, F, Duin, LK, Adama van Scheltema, PN, Cohen-Overbeek, TE, Pajkrt, E, Bekker, M, Willekes, C, Bax, CJ, Gracchi, V, Oepkes, D, Bilardo, CM. Prenatal diagnosis of LUTO: how to improve diagnostic accuracy. Ultrasound Obstet Gynecol. 2017; 52: 739–43.Google Scholar
Abdelazim, IA, Abdelrazak, KM, Ramy, AR, Mounib, AM. Complementary roles of prenatal sonography and magnetic resonance imaging in diagnosis of fetal renal anomalies. Aust N Z J Obstet Gynaecol. 2010; 50: 237–41.Google Scholar
Capito, C, Belarbi, N, Paye Jaouen, A, Leger, J, Carel, JC, Oury, JF, Sebag, G, El-Ghoneimi, A. Prenatal pelvic MRI: additional clues for assessment of urogenital obstructive anomalies. J Pediatr Urol. 2014; 10: 162–6.Google Scholar
Millischer, AE, Grevent, D, Rousseau, V, O’Gorman, N, Sonigo, P, Bessieres, B, Ville, Y, Boddaert, N, Salomon, LJ. Fetal MRI compared with ultrasound for the diagnosis of obstructive genital malformations. Prenat Diagn. 2017; 37: 1138–45.Google Scholar
Werner, H, Lopes, J, Ribeiro, G, Jésus, NR, Santos, GR, Alexandria, HAF, Ruano, R, Araujo Júnior, E. Three-dimensional virtual cystoscopy: Noninvasive approach for the assessment of urinary tract in fetuses with lower urinary tract obstruction. Prenat Diagn. 2017; 37: 1350–2.Google Scholar
Morris, RK, Malin, GL, Khan, KS, Kilby, MD. Antenatal ultrasound to predict postnatal renal function in congenital lower urinary tract obstruction: systematic review of test accuracy. BJOG. 2009; 116: 1290–9.Google Scholar
Harper, L, Waubant, A, Vignes, J, Amat, S, Dobremez, E, Lefevre, Y, Ferdynus, C. Can quantity of amniotic fluid reliably predict postnatal renal function in boys with posterior urethral valves: a decision curve analysis. Prenat Diagn. 2017; 37: 931–4Google Scholar
Johnson, MP, Danzer, E, Koh, J, Polzin, W, Harman, C, O’Shaughnessy, R, Brown, R, Zaretsky, MV, North American Fetal Therapy Network (NAFTNet). Natural history of fetal lower urinary tract obstruction with normal amniotic fluid volume at initial diagnosis. Fetal Diagn Ther. 2017; 44: 1017.Google Scholar
Ruano, R, Safdar, A, Au, J, Koh, CJ, Gargollo, P, Shamshirsaz, AA, Espinoza, J, Cass, DL, Olutoye, OO, Olutoye, OA, Welty, S, Roth, DR, Belfort, MA, Braun, MC. Defining and predicting ‘intrauterine fetal renal failure’ in congenital lower urinary tract obstruction. Pediatr Nephrol. 2016; 31: 605–12.Google Scholar
Johnson, MP, Corsi, P, Bradfield, W, Hume, RF, Smith, C, Flake, AW, Qureshi, F, Evans, MI. Sequential urinalysis improves evaluation of fetal renal function in obstructive uropathy. Am J Obstet Gynecol. 1995; 173: 5965.Google Scholar
Walsh, DS, Johnson, MP. Fetal interventions for obstructive uropathy. Semin Perinatol. 1999; 23: 484–95.Google Scholar
Morris, RK, Quinlan-Jones, E, Kilby, MD, Khan, KS. Systematic review of accuracy of fetal urine analysis to predict poor postnatal renal function in cases of congenital urinary tract obstruction. Prenat Diagn. 2007; 27: 900–11.Google Scholar
Nassr, AA, Koh, CK, Shamshirsaz, AA, Espinoza, J, Sangi-Haghpeykar, H, Sharhan, D, Welty, S, Angelo, J, Roth, D, Belfort, MA, Braun, M, Ruano, R. Are ultrasound renal aspects associated with urinary biochemistry in fetuses with lower urinary tract obstruction? Prenat Diagn. 2016; 36: 1206–10.Google Scholar
Ruano, R, Sananes, N, Wilson, C, Au, J, Koh, CJ, Gargollo, P, et al. Fetal lower urinary tract obstruction: proposal for standardized multidisciplinary prenatal management based on disease severity. Ultrasound Obstet Gynecol. 2016; 48: 476–82.Google Scholar
Ruano, R, Dunn, T, Braun, MC, Angelo, JR, Safdar, A. Lower urinary tract obstruction: fetal intervention based on prenatal staging. Pediatr Nephrol. 2017; 32: 1871–8.Google Scholar
Farrugia, MK, Braun, MC, Peters, CA, Ruano, R, Herndon, CD. Report on the society for fetal urology panel discussion on the selection criteria and intervention for fetal bladder outlet obstruction. J Pediatr Urol. 2017; 13: 345–51.Google Scholar
Morris, RK, Malin, GL, Quinlan-Jones, E, Middleton, LJ, Hemming, K, Burke, D, Daniels, JP, Khan, KS, Deeks, J, Kilby, MD, Percutaneous vesicoamniotic shunting in Lower Urinary Tract Obstruction (PLUTO) Collaborative Group. Percutaneous vesicoamniotic shunting versus conservative management for fetal lower urinary tract obstruction (PLUTO): a randomised trial. Lancet. 2013; 382: 1496–506.Google Scholar
Nassr, AA, Shazly, SAM, Abdelmagied, AM, Araujo Júnior, E, Tonni, G, Kilby, MD, Ruano, R. Effectiveness of vesicoamniotic shunt in fetuses with congenital lower urinary tract obstruction: an updated systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2017; 49: 696703.Google Scholar
Freedman, AL, Johnson, MP, Smith, CA, Gonzalez, R, Evans, MI. Long-term outcome in children after antenatal intervention for obstructive uropathies.Lancet. 1999; 354: 374–7.Google Scholar
Morris, RK, Ruano, R, Kilby, MD. Effectiveness of fetal cystoscopy as a diagnostic and therapeutic intervention for lower urinary tract obstruction: a systematic review. Ultrasound Obstet Gynecol. 2011; 37: 629–37.Google Scholar
Sananes, N, Cruz-Martinez, R, Favre, R, Ordorica-Flores, R, Moog, R, Zaloszy, A, Giron, AM, Ruano, R. Two-year outcomes after diagnostic and therapeutic fetal cystoscopy for lower urinary tract obstruction. Prenat Diagn. 2016; 36: 297303.Google Scholar
Sananes, N, Favre, R, Koh, CJ, Zaloszyc, A, Braun, MC, Roth, DR, Moog, R, Becmeur, F, Belfort, MA, Ruano, R. Urological fistulas after fetal cystoscopic laser ablation of posterior urethral valves: surgical technical aspects. Ultrasound Obstet Gynecol. 2015; 45: 183–9.Google Scholar
Ruano, R, Sananes, N, Sangi-Haghpeykar, H, Hernandez-Ruano, S, Moog, R, Becmeur, F, Zaloszyc, A, Giron, AM, Morin, B, Favre, R. Fetal intervention for severe lower urinary tract obstruction: a multicenter case-control study comparing fetal cystoscopy with vesicoamniotic shunting. Ultrasound Obstet Gynecol. 2015; 45: 452–8.Google Scholar
Kurtz, MP, Koh, CJ, Jamail, GA, Sangi-Haghpeykar, H, Shamshirsaz, AA, Espinoza, J, Cass, DL, Olutoye, OO, Olutoye, OA, Braun, MC, Roth, DR, Belfort, MA, Ruano, R. Factors associated with fetal shunt dislodgement in lower urinary tract obstruction. Prenat Diagn. 2016; 36: 720–5.Google Scholar
Diehl, W, Diemert, A, Grasso, D, Sehner, S, Wegscheider, K, Hecher, K. Fetoscopic laser coagulation in 1020 pregnancies with twin-twin transfusion syndrome demonstrates improvement in double-twin survival rate. Ultrasound Obstet Gynecol. 2017; 50: 728–35.Google Scholar
Haeri, S, Simon, DH, Pillutla, K. Serial amnioinfusions for fetal pulmonary palliation in fetuses with renal failure. J Matern Fetal Neonatal Med. 2017; 30: 174–6.Google Scholar
Gimpel, C, Avni, FE, Bergmann, C, Cetiner, M, Habbig, S, Haffner, D, et al. Perinatal diagnosis, management, and follow-up of cystic renal diseases: a clinical practice recommendation with systematic literature reviews. JAMA Pediatr. 2018; 172: 7486.Google Scholar
Sugarman, J, Anderson, J, Baschat, AA, Herrera Beutler, J, Bienstock, JL, Bunchman, TE, et al. Ethical considerations concerning amnioinfusions for treating fetal bilateral renal agenesis. Obstet Gynecol. 2018; 131: 130–4.Google Scholar

References

Longaker, MT, Laberge, JM, Dansereau, J, et al. Primary fetal hydrothorax: natural history and management. J Pediatr Surg. 1989; 24: 573–6.Google Scholar
Rustico, MA, Lanna, M, Coviello, D, Smoleniec, J, Nicolini, U. Fetal pleural effusion. Prenat Diagn. 2007; 27: 793–9.Google Scholar
Yinon, Y, Kelly, E, Ryan, G. Fetal pleural effusions. Best Pract Res Clin Obstet Gynaecol. 2008; 22: 7796.Google Scholar
Castillo, RA, Devoe, LD, Falls, G, et al. Pleural effusions and pulmonary hypoplasia. Am J Obstet Gynecol. 1987; 157: 1252–5.Google Scholar
Yinon, Y, Grisaru-Granovsky, S, Chaddha, V, et al. Perinatal outcome following fetal chest shunt insertion for pleural effusion. Ultrasound Obstet Gynecol. 2010; 36: 5864.Google Scholar
Nicolaides, K, Azar, GB. Thoraco-amniotic shunting. Fetal Diagn Ther. 1990; 5: 153–64.Google Scholar
Waller, K, Chaithongwongwatthana, S, Yamasmit, W, et al. Chromosomal abnormalities among 246 fetuses with pleural effusions detected on prenatal ultrasound examination: factors associated with an increased risk of aneuploidy. Genet Med. 2005; 7: 417.Google Scholar
Shaffer, LG, Rosenfeld, JA, Dabell, MP, et al. Detection rates of clinically significant genomic alterations by microarray analysis for specific anomalies detected by ultrasound. Prenat Diagn. 2012; 32: 986–95.Google Scholar
Aubard, Y, Derouineau, I, Aubard, V, Chalifour, V, Preux, PM. Primary fetal hydrothorax: a literature review and proposed antenatal clinical strategy. Fetal Diagn Ther. 1998; 13: 325–33.Google Scholar
Weber, AM, Philipson, EH. Fetal pleural effusion: a review and meta-analysis for prognostic indicators. Obstet Gynecol. 1992; 79: 281–6.Google Scholar
Rodeck, CH, Fisk, NM, Fraser, DI, Nicolini, U. Long-term in utero drainage of fetal hydrothorax. N Engl J Med. 1988; 319: 1135–8.Google Scholar
Smith, R, Illanes, S, Denbow, ML, Soothill, PW. Outcome of fetal pleural effusions treated by thoracoamniotic shunting. Ultrasound Obstet Gynecol. 2005; 26: 63–6.Google Scholar
Picone, O, Benachi, A, Mandelbrot, L, et al. Thoracoamniotic shunting for fetal pleural effusions with hydrops. Am J Obstet Gynecol. 2004; 191: 2047–50.Google Scholar
Pettersen, H, et al. Pleural effusions. In Fisk, NM, ed., Pleural Effusions. Cambridge, UK: Cambridge University Press, 1997, pp. 261–72.Google Scholar
Pellegrinelli, J, Kohler, A, Kohler, M, et al. Prenatal management and thoracoamniotic shunting in primary fetal pleural effusions: a single centre experience. Prenat Diagn. 2012; 32: 467–71.Google Scholar
Jeong, BD, Won, HS, Lee, MY, et al. Perinatal outcomes of fetal pleural effusion following thoracoamniotic shunting. Prenat Diagn. 2015; 35: 1365–70Google Scholar
Peranteau, WH, Adzick, NS, Boelig, MM, et al. Thoracoamniotic shunts for the management of fetal lung lesions and pleural effusions: a single-institution review and predictors of survival in 75 cases. J Pediatr Surg. 2015; 50: 301–5.Google Scholar
Wada, S, Jwa, SC, Yumoto, Y, et al. The prognostic factors and outcomes of primary fetal hydrothorax with the effects of fetal intervention. Prenat Diagn. 2017; 37: 184–92.Google Scholar
Witlox, RSGM, Lopriore, E, Rijken, M, et al. Long-term neurodevelopmental and respiratory outcome after intrauterine therapy for fetal thoracic abnormalities. Fetal Diagn Ther. 2018; 7: 16.Google Scholar
Okawa, T, Takano, Y, Fujimori, K, Yanagida, K, Sato, A. A new fetal therapy for chylothorax: pleurodesis with OK‐432. Ultrasound Obstet Gynecol. 2001; 18: 376–7.Google Scholar
Chen, M, Shih, JC, Wang, BT, Chen, CP, Yu, CL. Fetal OK‐432 pleurodesis: complete or incomplete? Ultrasound Obstet Gynecol. 2005; 26: 791–3.Google Scholar
Nygaard, U, Sundberg, K, Nielsen, HS, Hertel, S, Jørgensen, C. New treatment of early fetal chylothorax. Obstet Gynecol. 2007; 109: 1088–92.Google Scholar
O’Brien, B, Kesby, G, Ogle, R, Rieger, I, Hyett, JA. Treatment of primary fetal hydrothorax with OK-432 (Picibanil): outcome in 14 fetuses and a review of the literature. Fetal Diagn Ther. 2015; 37: 259–66.Google Scholar
Davenport, M, Warne, SA, Cacciaguerra, S, Patel, S, Greenough, A, Nicolaides, K. Current outcome of antenally diagnosed cystic lung disease. J Pediatr Surg. 2004; 39: 549–56.Google Scholar
Cass, DL, Crombleholme, TM, Howell, LJ, Stafford, PW, Ruchelli, ED, Adzick, NS. Cystic lung lesions with systemic arterial blood supply: a hybrid of congenital cystic adenomatoid malformation and bronchopulmonary sequestration. J Pediatr Surg. 1997; 32: 986–90.Google Scholar
Stocker, JT, Madewell, JE, Drake, RM. Congenital cystic adenomatoid malformation of the lung: classification and morphologic spectrum. Human Pathol. 1977; 8: 155–71.Google Scholar
Adzick, NS, Harrison, MR, Glick, PL, et al. Fetal cystic adenomatoid malformation: prenatal diagnosis and natural history. J Pediatr Surg. 1985; 20: 483–8.Google Scholar
Cavoretto, P, Molina, F, Poggi, S, Davenport, M, Nicolaides, KH. Prenatal diagnosis and outcome of echogenic fetal lung lesions. Ultrasound Obstet Gynecol. 2008; 32: 769–83.Google Scholar
Thorpe‐Beeston, J, Nicolaides, KH. Cystic adenomatoid malformation of the lung: prenatal diagnosis and outcome. Prenat Diagn. 1994; 14: 677–88.Google Scholar
Ehrenberg-Buchner, S, Stapf, AM, Berman, DR, et al. Fetal lung lesions: can we start to breathe easier? Am J Obstet Gynecol. 2013; 208: 151. e1–7.Google Scholar
Carter, R. Pulmonary sequestration. Ann Thorac Surg. 1969; 7: 6888.Google Scholar
DeParedes, CG, Pierce, WS, Johnson, DG, Waldhausen, JA. Pulmonary sequestration in infants and children: a 20-year experience and review of the literature. J Pediatr Surg. 1970; 5: 136–47.Google Scholar
Dhingsa, R, Coakley, FV, Albanese, CT, Filly, RA, Goldstein, R. Prenatal sonography and MR imaging of pulmonary sequestration. Am J Roentgenol. 2003; 180: 433–7.Google Scholar
Adzick, NS, Harrison, MR, Crombleholme, TM, Flake, AW, Howell, LJ. Fetal lung lesions: management and outcome. Am J Obstet Gynecol. 1998; 179: 884–9.Google Scholar
Grethel, EJ, Wagner, AJ, Clifton, MS, et al. Fetal intervention for mass lesions and hydrops improves outcome: a 15-year experience. J Pediatr Surg. 2007; 42: 117–23.Google Scholar
Crombleholme, TM, Coleman, B, Hedrick, H, et al. Cystic adenomatoid malformation volume ratio predicts outcome in prenatally diagnosed cystic adenomatoid malformation of the lung. J Pediatr Surg. 2002; 37: 331–8.Google Scholar
Cass, DL, Olutoye, OO, Cassady, CI, et al. Prenatal diagnosis and outcome of fetal lung masses. J Pediatr Surg. 2011; 46: 292–8.Google Scholar
Schrey, S, Kelly, EN, Langer, JC, et al. Fetal thoracoamniotic shunting for large macrocystic congenital cystic adenomatoid malformations of the lung. Ultrasound Obstet Gynecol. 2012; 39: 515–20.Google Scholar
Litwińska, M, Litwińska, E, Janiak, K, et al. Thoracoamniotic shunts in macrocystic lung lesions: case series and review of the literature. Fetal Diagn Ther. 2017; 41: 179–83.Google Scholar
Knox, EM, Kilby, MD, Martin, WL, Khan, KS. In‐utero pulmonary drainage in the management of primary hydrothorax and congenital cystic lung lesion: a systematic review. Ultrasound Obstet Gynecol. 2006; 28: 726–34.Google Scholar
Adzick, NS. Management of fetal lung lesions. Clin Perinatol. 2009; 36: 363–76, x.Google Scholar
Al-Refai, A, Ryan, G, Van Mieghem, T. Maternal risks of fetal therapy. Curr Opin Obstet Gynecol. 2017; 29: 80–4.Google Scholar
Curran, PF, Jelin, EB, Rand, L, et al. Prenatal steroids for microcystic congenital cystic adenomatoid malformations. J Pediatr Surg. 2010; 45: 145–50.Google Scholar
Peranteau, WH, Boelig, MM, Khalek, N, et al. Effect of single and multiple courses of maternal betamethasone on prenatal congenital lung lesion growth and fetal survival. J Pediatr Surg. 2016; 51: 2832.Google Scholar
Baud, D, Windrim, R, Kachura, JR, et al. Minimally invasive fetal therapy for hydropic lung masses: three different approaches and review of the literature. Ultrasound Obstet Gynecol. 2013; 42: 440–8.Google Scholar
Ruano, R, da Silva, MM, Salustiano, EM, et al. Percutaneous laser ablation under ultrasound guidance for fetal hyperechogenic microcystic lung lesions with hydrops: a single center cohort and a literature review. Prenat Diagn. 2012; 32: 1127–32.Google Scholar
Vu, L, Tsao, K, Lee, H, et al. Characteristics of congenital cystic adenomatoid malformations associated with nonimmune hydrops and outcome. J Pediatr Surg. 2007; 42: 1351–6.Google Scholar
Bermudez, C, Pérez-Wulff, J, Bufalino, G, et al. Percutaneous ultrasound‐guided sclerotherapy for complicated fetal intralobar bronchopulmonary sequestration. Ultrasound Obstet Gynecol. 2007; 29: 586–9.Google Scholar
Bermúdez, C, Pérez-Wulff, J, Arcadipane, M, et al. Percutaneous fetal sclerotherapy for congenital cystic adenomatoid malformation of the lung. Fetal Diagn Ther. 2008; 24: 237–40.Google Scholar
Downard, CD, Calkins, CM, Williams, RF, et al. Treatment of congenital pulmonary airway malformations: a systematic review from the APSA outcomes and evidence based practice committee. Pediatr Surg Int. 2017; 33: 939–53.Google Scholar
Hedrick, MH, Ferro, MM, Filly, RA, et al. Congenital high airway obstruction syndrome (CHAOS): a potential for perinatal intervention. J Pediatr Surg. 1994; 29: 271–4.Google Scholar
Tessier, A, Sarreau, M, Pelluard, F, et al. Fraser syndrome: features suggestive of prenatal diagnosis in a review of 38 cases. Prenat Diagn. 2016; 36: 1270–5.Google Scholar
Roybal, JL, Liechty, KW, Hedrick, HL, et al. Predicting the severity of congenital high airway obstruction syndrome. J Pediatr Surg. 2010; 45: 1633–9.Google Scholar
Martinez, JM, Castañón, M, Gómez, O, et al. Evaluation of fetal vocal cords to select candidates for successful fetoscopic treatment of congenital high airway obstruction syndrome: preliminary case series. Fetal Diagn Ther. 2013; 34: 7784.Google Scholar
Martínez, JM, Prat, J, Gómez, O, et al. Decompression through tracheobronchial endoscopy of bronchial atresia presenting as massive pulmonary tumor: a new indication for fetoscopic surgery. Fetal Diagn Ther. 2013; 33: 6974.Google Scholar
Ryan, G, Somme, S, Crombleholme, TM. Airway compromise in the fetus and neonate: Prenatal assessment and perinatal management. Semin Fetal Neonatal Med; 2016; 21: 230–9.Google Scholar
Abbasi, N, Morency, AM, Langer, JC, et al. Fetal Sclerotherapy for Hydropic Congenital Cystic Adenomatoid Malformations of the Lung Refractory to Steroids: A Case Report and Review of the Literature. https://www.ncbi.nlm.nih.gov/pubmed/31112955. Fetal Diagn Ther. 2019; May 21:1–10. doi: 10.1159/000497143. [Epub ahead of print]Google Scholar

References

Botto, LD, Moore, CA, Khoury, MJ, et al. Neural-tube defects. N Engl J Med. 1999; 341: 1509–19.Google Scholar
Michie, CA. Neural tube defects in 18th century. Lancet. 1991; 337: 504.Google Scholar
Padmanabhan, R. Etiology, pathogenesis and prevention of neural tube defects. Congenit Anom (Kyoto). 2006; 46: 5567.Google Scholar
Deak, KL, Siegel, DG, George, TM, et al. Further evidence for a maternal genetic effect and a sex-influenced effect contributing to risk for human neural tube defects. Birth Defects Res A Clin Mol Teratol 2008; 82: 662–9.Google Scholar
Wilde, JJ, Petersen, JR, Niswander, L. Genetic, epigenetic, and environmental contributions to neural tube closure. Annu Rev Genet. 2014; 48: 583611.Google Scholar
Au, KS, Ashley-Koch, A, Northrup, H. Epidemiologic and genetic aspects of spina bifida and other neural tube defects. Dev Disabil Res Rev. 2010; 16: 615.Google Scholar
Copp, AJ, Adzick, NS, Chitty, LS, et al. Spina bifida. Nat Rev Dis Primers. 2015; 1: 15007.Google Scholar
Viswanathan, M, Treiman, KA, Doto, JK, et al. Folic Acid Supplementation: An Evidence Review for the U.S. Preventive Services Task Force: U.S. Preventive Services Task Force Evidence Syntheses. Rockville: Agency for Healthcare Research and Quality, 2017.Google Scholar
van der Put, NM, Steegers-Theunissen, RP, Frosst, P, et al. Mutated methylenetetrahydrofolate reductase as a risk factor for spina bifida. Lancet. 1995; 346: 1070–1.Google Scholar
Besser, LM, Williams, LJ, Cragan, JD. Interpreting changes in the epidemiology of anencephaly and spina bifida following folic acid fortification of the U.S. grain supply in the setting of long-term trends, Atlanta, Georgia, 1968–2003. Birth Defects Res A Clin Mol Teratol. 2007; 79: 730–6.Google Scholar
Becerra, JE, Khoury, MJ, Cordero, JF, et al. Diabetes mellitus during pregnancy and the risks for specific birth defects: a population-based case-control study. Pediatrics. 1990; 85: 19.Google Scholar
Mitchell, LE, Adzick, NS, Melchionne, J, et al. Spina bifida. Lancet. 2004; 364: 1885–95.Google Scholar
Groenen, PM, Peer, PG, Wevers, RA, et al. Maternal myo-inositol, glucose, and zinc status is associated with the risk of offspring with spina bifida. Am J Obstet Gynecol. 2003; 189: 1713–19.Google Scholar
Carmichael, SL, Rasmussen, SA, Shaw, GM. Prepregnancy obesity: a complex risk factor for selected birth defects. Birth Defects Res A Clin Mol Teratol. 2010; 88: 804–10.Google Scholar
Parker, SE, Yazdy, MM, Tinker, SC, et al. The impact of folic acid intake on the association among diabetes mellitus, obesity, and spina bifida. Am J Obstet Gynecol. 2013; 209: 239. e1–8.Google Scholar
Desrosiers, TA, Siega-Riz, AM, Mosley, BS, et al. Low carbohydrate diets may increase risk of neural tube defects. Birth Defects Res. 2018; 110: 901–9.Google Scholar
Vujkovic, M, Steegers, EA, Looman, CW, et al. The maternal Mediterranean dietary pattern is associated with a reduced risk of spina bifida in the offspring. BJOG. 2009; 116: 408–15.Google Scholar
Suarez, L, Cardarelli, K, Hendricks, K. Maternal stress, social support, and risk of neural tube defects among Mexican Americans. Epidemiology. 2003; 14: 612–16.Google Scholar
Moretti, ME, Bar-Oz, B, Fried, S, et al. Maternal hyperthermia and the risk for neural tube defects in offspring: systematic review and meta-analysis. Epidemiology. 2005; 16: 216–19.Google Scholar
Molloy, AM, Pangilinan, F, Brody, LC. Genetic risk factors for folate-responsive neural tube defects. Annu Rev Nutr. 2017; 37: 269–91.Google Scholar
Smithells, RW, Sheppard, S, Schorah, CJ, et al. Apparent prevention of neural tube defects by periconceptional vitamin supplementation. Arch Dis Child. 1981; 56: 911–18.Google Scholar
MRC Vitamin Study Research Group. Prevention of neural tube defects: results of the Medical Research Council Vitamin Study. MRC Vitamin Study Research Group. Lancet. 1991; 338: 131–7.Google Scholar
Czeizel, AE, Dudas, I. Prevention of the first occurrence of neural-tube defects by periconceptional vitamin supplementation. N Engl J Med. 1992; 327: 1832–5.Google Scholar
Giardini, V, Russo, FM, Ornaghi, S, et al. Seasonal impact in the frequency of isolated spina bifida. Prenat Diagn. 2013; 33: 1007–9.CrossRefGoogle ScholarPubMed
Ray, JG, Wyatt, PR, Thompson, MD, et al. Vitamin B12 and the risk of neural tube defects in a folic-acid-fortified population. Epidemiology. 2007; 18: 362–6.Google Scholar
Steegers-Theunissen, RP, Boers, GH, Trijbels, FJ, et al. Maternal hyperhomocysteinemia: a risk factor for neural-tube defects? Metabolism. 1994; 43: 1475–80.Google Scholar
Steegers-Theunissen, RP, Boers, GH, Trijbels, FJ, et al. Neural-tube defects and derangement of homocysteine metabolism. N Engl J Med. 1991; 324: 199200.Google Scholar
Yang, M, Li, W, Wan, Z, et al. Elevated homocysteine levels in mothers with neural tube defects: a systematic review and meta-analysis. J Matern Fetal Neonatal Med. 2017; 30: 2051–7.Google Scholar
Shaw, GM, Finnell, RH, Blom, HJ, et al. Choline and risk of neural tube defects in a folate-fortified population. Epidemiology. 2009; 20: 714–19.Google Scholar
Greene, ND, Leung, KY, Copp, AJ. Inositol, neural tube closure and the prevention of neural tube defects. Birth Defects Res. 2017; 109: 6880.Google Scholar
Grewal, J, Carmichael, SL, Ma, C, et al. Maternal periconceptional smoking and alcohol consumption and risk for select congenital anomalies. Birth Defects Res A Clin Mol Teratol. 2008; 82: 519–26.Google Scholar
Schmidt, RJ, Romitti, PA, Burns, TL, et al. Maternal caffeine consumption and risk of neural tube defects. Birth Defects Res A Clin Mol Teratol. 2009; 85: 879–89.Google Scholar
Vajda, FJ, Eadie, MJ. Maternal valproate dosage and foetal malformations. Acta Neurol Scand. 2005; 112: 137–43.Google Scholar
Padula, AM, Tager, IB, Carmichael, SL, et al. The association of ambient air pollution and traffic exposures with selected congenital anomalies in the San Joaquin Valley of California. Am J Epidemiol. 2013; 177: 1074–85.Google Scholar
Lupo, PJ, Symanski, E, Waller, DK, et al. Maternal exposure to ambient levels of benzene and neural tube defects among offspring: Texas, 1999-2004. Environ Health Perspect. 2011; 119: 397402.Google Scholar
Li, Z, Zhang, L, Ye, R, et al. Indoor air pollution from coal combustion and the risk of neural tube defects in a rural population in Shanxi Province, China. Am J Epidemiol. 2011; 174: 451–8.Google Scholar
Righi, E, Bechtold, P, Tortorici, D, et al. Trihalomethanes, chlorite, chlorate in drinking water and risk of congenital anomalies: a population-based case-control study in Northern Italy. Environ Res. 2012; 116: 6673.Google Scholar
Brender, JD, Felkner, M, Suarez, L, et al. Maternal pesticide exposure and neural tube defects in Mexican Americans. Ann Epidemiol. 2010; 20: 1622.Google Scholar
Honein, MA, Paulozzi, LJ, Mathews, TJ, et al. Impact of folic acid fortification of the US food supply on the occurrence of neural tube defects. JAMA. 2001; 285: 2981–6.Google Scholar
Williams, J, Mai, CT, Mulinare, J, et al. Updated estimates of neural tube defects prevented by mandatory folic acid fortification – United States, 1995–2011. MMWR Morb Mortal Wkly Rep. 2015; 64: 15.Google Scholar
De Wals, P, Tairou, F, Van Allen, MI, et al. Reduction in neural-tube defects after folic acid fortification in Canada. N Engl J Med. 2007; 357: 135–42.Google Scholar
Bentley, TG, Weinstein, MC, Willett, WC, et al. A cost-effectiveness analysis of folic acid fortification policy in the United States. Public Health Nutr. 2009; 12: 455–67.Google Scholar
Atta, CA, Fiest, KM, Frolkis, AD, et al. Global birth prevalence of spina bifida by folic acid fortification status: a systematic review and meta-analysis. Am J Public Health. 2016; 106: e24–34.Google Scholar
Mills, JL, Von Kohorn, I, Conley, MR, et al. Low vitamin B-12 concentrations in patients without anemia: the effect of folic acid fortification of grain. Am J Clin Nutr. 2003; 77: 1474–7.Google Scholar
Vollset, SE, Clarke, R, Lewington, S, et al. Effects of folic acid supplementation on overall and site-specific cancer incidence during the randomised trials: meta-analyses of data on 50,000 individuals. Lancet. 2013; 381: 1029–36.Google Scholar
Tinker, SC, Hamner, HC, Qi, YP, et al. U.S. women of childbearing age who are at possible increased risk of a neural tube defect-affected pregnancy due to suboptimal red blood cell folate concentrations, National Health and Nutrition Examination Survey 2007 to 2012. Birth Defects Res A Clin Mol Teratol. 2015; 103: 517–26.Google Scholar
Centers for Disease Control and Prevention. Racial/ethnic differences in the birth prevalence of spina bifida – United States, 1995–2005. MMWR Morb Mortal Wkly Rep. 2009; 57: 1409–13.Google Scholar
Greene, ND, Leung, KY, Gay, V, et al. Inositol for prevention of neural tube defects: a pilot randomised controlled trial – CORRIGENDUM. Br J Nutr. 2016; 115: 1697.Google Scholar
Khoshnood, B, Loane, M, de Walle, H, et al. Long term trends in prevalence of neural tube defects in Europe: population based study. BMJ. 2015; 351: h5949.Google Scholar
Busby, A, Abramsky, L, Dolk, H, et al. Preventing neural tube defects in Europe: population based study. BMJ. 2005; 330: 574–5.CrossRefGoogle ScholarPubMed

References

Bowman, RM, McLone, DG, Grant, JA, et al. Spina bifida outcome: a 25-year prospective. Pediatr Neurosurg. 2001; 34: 114–20.Google Scholar
Shin, M, Kucik, JE, Siffel, C, et al. Improved survival among children with spina bifida in the United States. J Pediatrics. 2012; 161: 1132–7. e3.Google Scholar
Rintoul, NE, Sutton, LN, Hubbard, AM, et al. A new look at myelomeningoceles: Functional level, vertebral level, shunting and the implications for fetal intervention. Pediatrics. 2002; 109: 409–13.Google Scholar
Caldarelli, M, Di Rocco, C, La Marca, F. Shunt complications in the first postoperative year in children with meningomyelocele. Childs Nerv Syst. 1996; 12: 748–54.Google Scholar
McLone, DG, Dias, MS. The Chiari II malformation: cause and impact. Childs Nerv Sys. 2003; 19: 540–50.Google Scholar
Just, M, Schwarz, M, Ludwig, B, et al. Cerebral and spinal MR-findings in patients with postrepair myelomeningocele. Pediatr Radiol. 1990; 20: 262–6.Google Scholar
Mitchell, LE, Adzick, NS, Melchionne, J, et al. Spina bifida. Lancet. 2004; 364: 1885–95.Google Scholar
Cochrane, DD, Wilson, RD, Steinbok, P, et al. Prenatal spinal evaluation and functional outcome of patients born with myelomeningocele: information for improved prenatal counselling and outcome prediction. Fetal Diagn Ther. 1996; 11: 159–68.Google Scholar
Cass, AS, Luxenberg, M, Johnson, CF, et al. Incidence of urinary tract complications with myelomeningocele. Urology. 1985; 25: 374–8.Google Scholar
Drennan, JC. Foot deformities in myelomeningocele. In Thilos, HS, ed., Instruction Course Lectures. Park Ridge, IL: American Academy of Orthopedic Surgeons, 1991.Google Scholar
Oakeshott, P, Hunt, GM, Poulton, A, et al. Expectation of life and unexpected death in open spina bifida: a 40-year complete, nonselective, longitudinal cohort study. Dev Med Child Neurol. 2010; 52: 749–53.Google Scholar
Oakeshott, P, Hunt, GM. Long-term outcome in spina bifida. Br J Gen Pract. 2003; 53: 632–6.Google Scholar
Parker, SE, Mai, CT, Canfield, MA, et al. Updated national birth prevalence estimates for selected birth defects in the United States, 2004–2006. Birth Defects Res A Clin Mol Teratol. 2010; 88: 1008–16.Google Scholar
Christianson, A, Modell, B, Howson, C. March of Dimes Global Report on Birth Defects: The Hidden Toll of Dying and Disabled Children. White Plains, NY: March of Dimes, 2006.Google Scholar
Centers for Disease Control and Prevention. Hospital stays, hospital charges, and in-hospital deaths among infants with selected birth defects – United States, 2003. MMWR Morb Mortal Wkly Rep. 2007; 56: 25–9.Google Scholar
Radcliff, E, Cassell, CHTanner, JP, et al. Hospital use, associated costs, and payer status for infants born with spina bifida. Birth Defects Res A Clin Mol Teratol. 2012; 94: 1044–53.Google Scholar
Ouyang, L, Grosse, SD, Armour, BS, et al. Health care expenditures of children and adults with spina bifida in a privately insured U.S. population. Birth Defects Res A Clin Mol Teratol. 2007; 79: 552–8.Google Scholar
Werner, EF, Han, CS, Burd, I, et al. Evaluating the cost-effectiveness of prenatal surgery for myelomeningocele: a decision analysis. Ultrasound Obstet Gynecol. 2012; 40: 158–64.Google Scholar
Michejda, M. Intrauterine treatment of spina bifida: primate model. Z Kinderchir. 1984; 39: 259–61.Google Scholar
Heffez, DS, Aryanpur, J, Rotellini, NA, et al. Intrauterine repair of experimental surgically created dysraphism. Neurosurgery. 1993; 32: 1005–10.Google Scholar
Heffez, DS, Aryanpur, J, Hutchins, GM, et al. The paralysis associated with myelomeningocele: clinical and experimental data implicating a preventable spinal cord injury. Neurosurgery 1990; 26: 987–92.Google Scholar
Meuli, M, Meuli-Simmen, C, Yingling, CD, et al. Creation of myelomeningocele in utero: a model of functional damage from spinal cord exposure in fetal sheep. J Pediatr Surg. 1995; 30: 1028–32.Google Scholar
Meuli, M, Meuli-Simmen, C, Hutchins, GM, et al. In utero surgery rescues neurological function at birth in sheep with spina bifida. Nat Med. 1995; 1: 342–7.Google Scholar
Paek, BW, Farmer, DL, Wilkinson, CC, et al. Hindbrain herniation develops in surgically created myelomeningocele but is absent after repair in fetal lambs. Am J Obstet Gynecol. 2000; 183: 1119–23.Google Scholar
Bouchard, S, Davey, MG, Rintoul, NE, et al. Correction of hindbrain herniation and anatomy of the vermis after in utero repair of myelomeningocele in sheep. J Pediatr Surg. 2003; 38: 451–8.Google Scholar
Bruner, JP, Tulipan, NB, Richards, WO. Endoscopic coverage of fetal open myelomeningocele in utero. Am J Obstet Gynecol. 1997; 176: 256–7.Google Scholar
Adzick, NS, Sutton, LN, Crombleholme, TM, et al. Successful fetal surgery for spina bifida. Lancet. 1998; 352: 1675–6.Google Scholar
Tulipan, N, Hernanz-Schulman, M, Bruner, JP. Reduced hindbrain herniation after intrauterine myelomeningocele repair: a report of four cases. Pediatr Neurosurg. 1998; 29: 274–8.Google Scholar
Sutton, LN, Adzick, NS, Bilaniuk, LT, et al. Improvement in hindbrain herniation demonstrated by serial fetal magnetic resonance imaging following fetal surgery for myelomeningocele. JAMA. 1999; 282: 1826–31.Google Scholar
Adzick, NS, Thom, EA, Spong, CY, et al. A randomized trial of prenatal versus postnatal repair of myelomeningocele. N Engl J Med. 2011; 364: 9931004. (Full study protocol available in the Supplementary Appendix at NEJM.org)Google Scholar
Tulipan, N, Wellons, JC III, Thom, EA, et al. Prenatal surgery for myelomeningocele and the need for cerebrospinal fluid shunt placement. J Neurosurg Pediatr. 2015; 16: 613–20.Google Scholar
Farmer, DL, Thom, EA, Brock, JW, et al. The Management of the Myelomeningocele Study: Full cohort 30 month pediatric outcomes. Am J Obstet Gyn. 2018; 218: 256. e1–13.Google Scholar
Brock, JW III, Carr, MC, Adzick, NS, et al. Bladder function after fetal surgery for myelomeningocele. Pediatrics. 2015; 136: e906–13.Google Scholar
Johnson, MP, Bennett, KA, Rand, L, et al. The Management of Myelomeningocele Study: obstetrical outcomes and risk factors for obstetrical complications following prenatal surgery. Am J Obstet Gyn. 2016; 215: 778–9.Google Scholar
Antiel, RM, Adzick, NS, Thom, EA, et al. Impact on family and parental stress of prenatal versus postnatal repair of myelomeningocele. Am J Obstet Gyn. 2016; 215: 522. e1–6.Google Scholar
Moldenhauer, JS, Soni, S, Rintoul, NE, et al. Fetal myelomeningocele repair: the post-MOMS experience at the Children’s Hospital of Philadelphia. Fetal Diagn Ther. 2015; 37: 235–40.Google Scholar
Soni, S, Moldenhauer, JS, Spinner, SS, et al. Chorioamniotic membrane separation and preterm premature rupture of membranes complicating in utero myelomeningocele repair. Am J Obstet Gynecol. 2016; 214: 647. e1–7.Google Scholar
Wilson, RD, Johnson, MP, Crombleholme, TM, et al. Chorioamniotic membrane separation following open fetal surgery: pregnancy outcome. Fetal Diagn Ther. 2003; 18: 314–20.Google Scholar
Heuer, GG, Adzick, NS, Sutton, LN. Fetal myelomeningocele closure: technical considerations. Fetal Diagn Ther. 2015; 37: 166–71.Google Scholar
Flanders, TM, Heuer, GG, Madsen, PJ, et al. Modified myofascial technique for open fetal myelomeningocele results in improved outcomes. Presented at the 88th meeting of the American Association of Neurological Surgery, New Orleans, LA, May 1, 2018.Google Scholar
American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 550: Maternal-fetal surgery for myelomeningocele. Obstet Gynecol. 2013; 121: 218–19.Google Scholar
Simpson, JL, Greene, MF. Fetal surgery for myelomeningocele? N Engl J Med. 2011; 364: 1076–7.Google Scholar
Cohen, AR, Couto, J, Cummings, JJ, et al. Position statement on fetal myelomeningocele repair. Am J Obstet Gynecol. 2014; 210: 107–11.Google Scholar
Bennett, KA, Carroll, MA, Shannon, CN, et al. Reducing perinatal complications and preterm delivery for patients undergoing in utero closure of fetal myelomeningocele: further modifications to the multidisciplinary surgical technique. J Neurosurg Pediatr. 2014; 14: 108–14.Google Scholar
Moise, KJ Jr, Moldenhauer, JS, Bennett, KA, et al. Current selection criteria and perioperative therapy used for fetal myelomeningocele surgery. Obstet Gynecol. 2016; 127: 593–7.Google Scholar
Thom, EA. Maternal reproductive outcomes after in-utero repair of myelomeningocele. Am J Obstet Gynecol. 2016; 214: S36.Google Scholar
Wilson, RD, Lemerand, K, Johnson, MP, et al. Reproductive outcomes in subsequent pregnancies after a pregnancy complicated by open maternal-fetal surgery (1996–2007). Am J Obstet Gynecol. 2010; 203: 209. e1–6.Google Scholar
Kohl, T. Percutaneous minimally invasive fetoscopic surgery for spina bifida aperta. Part I: surgical technique and perioperative outcome. Ultrasound Obstet Gynecol. 2014; 44: 515–24.Google Scholar
Degenhardt, J, Schürg, R, Winarno, A, et al. Percutaneous minimal-access fetoscopic surgery for spina bifida aperta. Part II: maternal management and outcome. Ultrasound Obstet Gynecol. 2014; 44: 525–31.Google Scholar
Graf, K, Kohl, T, Neubauer, BA, et al. Percutaneous minimally invasive fetoscopic surgery for spina bifida aperta – Part III: postnatal neurosurgical interventions in the first year of life. Ultrasound Obstet Gynecol. 2016; 47: 158–61.Google Scholar
Flake, AW. Percutaneous minimal-access fetoscopic surgery for myelomeningocele – not so minimal! Ultrasound Obstet Gynecol. 2014; 44: 499500.Google Scholar
Pedreira, DA, Zanon, N, Nishikuni, K, et al. Endoscopic surgery for the antenatal treatment of myelomeningocele: the CECAM trial. Am J Obstet Gynecol. 2016; 214: 111.e1111.e11.Google Scholar
Belfort, MA, Whitehead, WE, Shamshirsaz, AA, et al. Fetoscopic open neural tube defect repair: Development and refinement of a two-port, carbon dioxide insufflation technique. Obstet Gynecol. 2017; 129: 734–43.Google Scholar
Luks, FI, Deprest, J, Marcus, M, et al. Carbon dioxide pneumoamnios causes acidosis in fetal lamb. Fetal Diagn Ther. 1994; 9: 105–9.Google Scholar
Skinner, S, DeKoninck, P, Crossley, K, et al. Partial amniotic carbon dioxide insufflation for fetal surgery. Prenat Diagn. 2018; 38: 983–93.Google Scholar
Lawrence, KM, Rossidis, AC, Baumgarten, HD et al. Safety of prolonged intra-amniotic carbon dioxide insufflation in a fetal sheep model. Presented at the 49th meeting of the American Pediatric Surgical Association, Palm Desert, CA, May 4, 2018.Google Scholar
Joyeux, L, Engels, AC, Russo, FM, et al. Fetoscopic versus open repair for spina bifida aperta: a systematic review of outcomes. Fetal Diagn Ther. 2016; 39: 161–71.Google Scholar
Araujo Junior, E, Eggink, AJ, van den Dobbelsteen, J, et al. Procedure-related complications of open vs. endoscopic fetal surgery for treatment of spina bifida in an era of intrauterine myelomeningocele repair: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2016; 48: 151–60.Google Scholar
Kabagambe, SK, Jensen, GW, Chen, YJ, et al. Fetal surgery for myelomeningocele: a systematic review and meta-analysis of outcomes in fetoscopic versus open repair. Fetal Diagn Ther. 2018; 43: 161–74.Google Scholar
Watanabe, M, Kim, AG, Flake, AW. Tissue engineering strategies for fetal myelomeningocele repair in animal models. Fetal Diagn Ther. 2015; 37: 197205.Google Scholar
Brown, EG, Saadai, P, Pivetti, CD, et al. In utero repair of myelomeningocele with autologous amniotic membrane in the fetal lamb model. J Pediatr Surg. 2014; 49: 133–8.Google Scholar

References

Williams, J, Mai, CT, Mulinare, J, et al. Updated estimates of neural tube defects prevented by mandatory folic acid fortification – United States, 1995–2011. MMWR Morb Mortal Wkly Rep. 2015; 64: 15.Google Scholar
Bowman, RM, Boshnjaku, V, McLone, DG. The changing incidence of myelomeningocele and its impact on pediatric neurosurgery: a review from the Children’s Memorial Hospital. Childs Nerv Syst. 2009; 25: 801–6.Google Scholar
Meuli, M, Meuli-Simmen, C, Yingling, CD, et al. In utero repair of experimental myelomeningocele saves neurological function at birth. J Pediatr Surg. 1996; 31: 397402.Google Scholar
Paek, BW, Farmer, DL, Wilkinson, CC, et al. Hindbrain herniation develops in surgically created myelomeningocele but is absent after repair in fetal lambs. Am J Obstet Gynecol. 2000; 183: 1119–23.Google Scholar
Tulipan, N, Bruner, JP. Myelomeningocele repair in utero: a report of three cases. Pediatr Neurosurg. 1998;28: 177–80.Google Scholar
Adzick, NS, Thom, EA, Spong, CY, et al. A randomized trial of prenatal versus postnatal repair of myelomeningocele. N Engl J Med. 2011; 364: 9931004.Google Scholar
Farmer, DL, Thom, EA, Brock, JW 3rd, Burrows, PK, Johnson, MP, Howell, LJ, et al. The Management of Myelomeningocele Study: full cohort 30-month pediatric outcomes. Am J Obstet Gynecol. 2018; 218: 256. e1–256. e13.Google Scholar
Brock, JW 3rd, Carr, MC, Adzick, NS, Burrows, PK, Thomas, JC, Thom, EA, et al. Bladder function after fetal surgery for myelomeningocele. Pediatrics. 2015; 136: e906–13.Google Scholar
Bruner, JP, Tulipan, NE, Richards, WO. Endoscopic coverage of fetal open myelomeningocele in utero. Am J Obstet Gynecol. 1997; 176: 256–7.Google Scholar
Bruner, JP, Richards, WO, Tulipan, NB, Arney, TL. Endoscopic coverage of fetal myelomeningocele in utero. Am J Obstet Gynecol. 1999; 180: 153–8.Google Scholar
Farmer, DL, von Koch, CS, Peacock, WJ, et al. In utero repair of myelomeningocele: experimental pathophysiology, initial clinical experience, and outcomes. Arch Surg. 2003; 138: 872–8.Google Scholar
Bruner, JP, Tulipan, NB, Richards, WO, Walsh, WF, Boehm, FH, Vrabcak, EK. In utero repair of myelomeningocele: a comparison of endoscopy and hysterotomy. Fetal Diagn Ther. 2000; 15: 83–8.Google Scholar
Kohl, T, Hering, R, Heep, A, et al. Percutaneous fetoscopic patch coverage of spina bifida aperta in the human – early clinical experience and potential. Fetal Diagn Ther. 2006; 21: 185–93.Google Scholar
Kohl, T. Percutaneous minimally invasive fetoscopic surgery for spina bifida aperta. Part I: Surgical technique and perioperative outcome. Ultrasound Obstet Gynecol. 2014; 44: 515–24.Google Scholar
Degenhardt, J, Schurg, R, Winarno, A, et al. Percutaneous minimal-access fetoscopic surgery for spina bifida aperta. Part II: maternal management and outcome. Ultrasound Obstet Gynecol. 2014; 44: 525–31.Google Scholar
Graf, K, Kohl, T, Neubauer, BA, et al. Percutaneous minimally invasive fetoscopic surgery for spina bifida aperta. Part III: neurosurgical intervention in the first postnatal year. Ultrasound Obstet Gynecol. 2016; 47: 158–61.Google Scholar
Ziemann, M, Fimmers, R, Khaleeva, A, Schürg, R, Weigand, MA, Kohl, T. Partial amniotic carbon dioxide insufflation (PACI) during minimally invasive fetoscopic interventions on fetuses with spina bifida aperta. Surg Endosc. 2018; 32: 3138–48.Google Scholar
Pedreira, DA, Zanon, N, de Sa, RA, et al. Fetoscopic single-layer repair of open spina bifida using a cellulose patch: preliminary clinical experience. J Mat Fetal Neonat Med. 2014; 27: 1613–19.Google Scholar
Pedreira, DA, Zanon, N,Nishikuni, K, et al. Endoscopic surgery for the antenatal treatment of myelomeningocele: the CECAM trial. Am J Obstet Gynecol. 2016; 214: 111. e1–111. e11.Google Scholar
Lapa Pedreira, DA, Acacio, GL, Gonçalves, RT, , RAM, Brandt, RA, Chmait, R, et al. Percutaneous fetoscopic closure of large open spina bifida using a bilaminar skin substitute. Ultrasound Obstet Gynecol. 2018; 52: 458–66.Google Scholar
Belfort, M, Deprest, J, Hecher, K. Current controversies in prenatal diagnosis 1: in utero therapy for spina bifida is ready for endoscopic repair. Prenat Diagn. 2016; 36: 1161–6.Google Scholar
Peiro, JL, Fontecha, CG, Ruano, R, Esteves, M, Fonseca, C, Marotta, M, et al. Single-access fetal endoscopy (SAFE) for myelomeningocele in sheep model I: amniotic carbon dioxide gas approach. Surg Endosc. 2013; 27: 3835–40.Google Scholar
Belfort, MA, Whitehead, WE, Bednov, A, Shamshirsaz, AA. Low-fidelity simulator for the standardized training of fetoscopic meningomyelocele repair. Obstet Gynecol. 2018; 131: 125–9.Google Scholar
Belfort, MA, Whitehead, WE, Shamshirsaz, AA, et al. Fetoscopic repair of meningomyelocele. Obstet Gynecol. 2015; 126: 881–4.Google Scholar
Pham, P. (2018). Sophisticated tools lead to breakthroughs in prenatal surgery. Wired. www.wired.com/story/nicu-tools-fetal-surgeryGoogle Scholar
Belfort, MA, Whitehead, WE, Shamshirsaz, AA, et al. Fetoscopic open neural tube defect repair: development and refinement of a two-port, carbon dioxide insufflation technique. Obstet Gynecol. 2017; 129: 734–43.Google Scholar
Kohn, JR, Rao, V, Sellner, AA, Sharhan, D, Espinoza, J, Shamshirsaz, AA, et al. Management of labor and delivery after fetoscopic repair of an open neural tube defect. Obstet Gynecol. 2018; 131: 1062–8.Google Scholar
Sanz Cortes, M, Torres, O, Sharhan, D, Yepez, M, Espinoza, J, Shamshirsaz, AA, et al. Neurodevelopmental assessment in patients who underwent prenatal fetoscopic and open fetal neural tube defect repair. Am J Obstet Gynecol. 2018; 1 (Suppl.): S294–5.Google Scholar
Kohl, T, Reckers, J, Strümper, D, Große Hartlage, M, Gogarten, W, Gembruch, U, et al. Amniotic air insufflation during minimally invasive fetoscopic fetal cardiac interventions is safe for the fetal brain in sheep. J Thorac Cardiovasc Surg. 2004; 128: 467–71.Google Scholar
Moise, KJ Jr., Tsao, K, Papanna, RM, Bebbington, MW. Fetoscopic repair of meningomyelocele. Obstet Gynecol. 2015; 126: 674.Google Scholar
Moise, KJ Jr., Flake, A. Fetoscopic open neural tube defect repair: development and refinement of a two-port, carbon dioxide insufflation technique. Obstet Gynecol. 2017; 130: 648.Google Scholar
Luks, FI, Deprest, J, Marcus, M, Vandenberghe, K, Vertommen, JD, Lerut, T, et al. Carbon dioxide pneumoamnios causes acidosis in fetal lamb. Fetal Diagn Ther. 1994; 9: 105–9.Google Scholar
Gratacós, E, Wu, J, Devlieger, R, Van de Velde, M, Deprest, JA. Effects of amniodistension with carbon dioxide on fetal acid–base status during fetoscopic surgery in the sheep model. Surg Endosc. 2001; 15: 368–72.Google Scholar
Gardner, DS, Fletcher, AJ, Bloomfield, MR, Fowden, AL, Giussani, DA. Effects of prevailing hypoxaemia, acidaemia or hypoglycaemia upon the cardiovascular, endocrine and metabolic responses to acute hypoxaemia in the ovine fetus. J Physiol. 2002; 540: 351–66.Google Scholar
Sabik, JF, Assad, RS, Hanley, FL. Halothane as an anesthetic for fetal surgery. J Pediatr Surg. 1993; 28: 542–6.Google Scholar
Saiki, Y, Litwin, DE, Bigras, JL, Waddell, J, Konig, A, Baik, S, et al. Reducing the deleterious effects of intrauterine CO2 during fetoscopic surgery. J Surg Res. 1997; 69: 51–4.Google Scholar
Akbar, SA, Brown, PR. Human erythrocyte CAI and CAII isoenzymes in hypoxemic and anemic fetuses. Clin Biochem. 1996; 29: 5762.Google Scholar
Lönnerholm, G, Wistrand, PJ. Carbonic anhydrase in the human fetal kidney. Pediatr Res. 1983; 17: 390–7.Google Scholar
Supuran, CT. Structure and function of carbonic anhydrases. Biochem J. 2016; 473: 2023–32.Google Scholar
Sly, WS, Hu, PY. Human carbonic anhydrases and carbonic anhydrase deficiencies. Annu Rev Biochem. 1995; 64: 375401.Google Scholar
Schipain, E, Mangiavini, L, Merceron, C. HIF-1a and growth plate development: what we really know. Bonekey Rep. 2015; 4: 730.Google Scholar
Thakor, AS, Giussani, DA. Effects of acute acidemia on the fetal cardiovascular defense to acute hypoxemia. Am J Physiol Regul Integr Comp Physiol. 2009; 296: R90–9.Google Scholar
Kohl, T. Impact of partial amniotic carbon dioxide insufflation (PACI) on middle cerebral artery blood flow in mid-gestation human fetuses undergoing fetoscopic surgery for spina bifida aperta. Ultrasound Obstet Gynecol. 2016; 47: 521–2.Google Scholar
Kassir, E, Belfort, MA, Shamshirsaz, AA, et al. Doppler changes in umbilical artery and ductus venosus during fetoscopic prenatal surgical repair of myelomeningocele. Ultrasound Obstet Gynecol. 2019; 53: 335–9.Google Scholar
Mann, DG, Nassr, AA, Whitehead, WE, Espinoza, J, Belfort, MA, Shamshirsaz, AA. Fetal bradycardia associated with maternal hypothermia after fetoscopic repair of neural tube defect. Ultrasound Obstet Gynecol. 2018; 51: 411–12.Google Scholar
Dean, M, Ramsay, R, Heriot, A, Mackay, J, Hiscock, R, Lynch, AC. Warmed, humidified CO2 insufflation benefits intraoperative core temperature during laparoscopic surgery: a meta-analysis. Asian J Endosc Surg. 2017; 10: 128–36.Google Scholar
Baschat, AA, Ahn, ES, Murphy, J, Miller, JL. Fetal blood gas values during fetoscopic myelomeningocele repair performed under carbon dioxide insufflation. Ultrasound Obstet Gynecol. 2018; 52: 400–2.Google Scholar
Partridge, EA, Davey, MG, Hornick, MA, McGovern, PE, Mejaddam, AY, Vrecenak, JD, et al. An extra-uterine system to physiologically support the extreme premature lamb. Nat Commun. 2017; 8: 15112.Google Scholar
Davey, AK, Hayward, J, Marshall, JK, Woods, AE. The effects of insufflation conditions on rat mesothelium. Int J Inflam. 2013; 2013: 816283.Google Scholar
Peng, Y, Zheng, M, Ye, Q, Chen, X, Yu, B, Liu, B. Heated and humidified CO2 prevents hypothermia, peritoneal injury, and intra-abdominal adhesions during prolonged laparoscopic insufflations. J Surg Res. 2009; 151: 40–7.Google Scholar
Sanz Cortes, M, Castro, E, Sharhan, D, Torres, P, Yepez, M, Espinoza, J, Shamshirsaz, AA, Nassr, AA, Popek, E, Whitehead, W, Belfort, MA. Amniotic membrane and placental histopathological findings after open and fetoscopic prenatal neural tube defect repair. Prenat Diagn. 2019 Mar; 39(4): 269279.Google Scholar
Erikoglu, M, Yol, S, Avunduk, MC, Erdemli, E, Can, A. Electron-microscopic alterations of the peritoneum after both cold and heated carbon dioxide pneumoperitoneum. J Surg Res. 2005; 125: 73–7.Google Scholar
Papanna, R, Mann, LK, Moise, KJ Jr., Kyriakides, T, Johnson, A, Garcia, E, et al. Histologic changes of the fetal membranes after fetoscopic laser surgery for twin-twin transfusion syndrome. Pediatr Res. 2015; 78: 247–55.Google Scholar

References

Oka, K, Okane, M, Okuno, S, et al. Congenital cervical immature teratoma arising in the left lobe of the thyroid gland. APMIS. 2007; 115: 75–9.Google Scholar
Hodges, MM, Crombleholme, TM, Marwan, AI, et al. Massive facial teratoma managed with the ex utero intrapartum treatment (EXIT) procedure and use of 3-dimensional printed model for planning of staged debulking. J Pediatr Surg. 2017; 17: 1519.Google Scholar
Kelly, MF, Berenholz, L, Rizzo, KA, et al. Approach for oxygenation of the newborn with airway obstruction due to a cervical mass. Ann Otol Rhinol Laryngol. 1990; 99: 179–82.Google Scholar
Hubbard, AM, Crombleholme, TM, Adzick, NS. Prenatal MRI evaluation of giant neck masses in preparation for the fetal exit procedure. Am J Perinatol. 1998; 15: 253–7.Google Scholar
Thawani, JP, Randazzo, MJ, Singh, N, Pisapia, JM, Abdullah, KG, Storm, PB. Management of giant cervical teratoma with intracranial extension diagnosed in utero. J Neurol Surg Rep. 2016; 77: e118–120.Google Scholar
Trecet, JC, Claramunt, V, Larraz, J, et al. Prenatal ultrasound diagnosis of fetal teratoma of the neck. J Clin Ultrasound. 1984; 12: 509–11.Google Scholar
Ryan, G, Somme, S, Crombleholme, TM. Airway compromise in the fetus and neonate: Prenatal assessment and perinatal management. Semin Fetal Neonatal Med. 2016; 21: 230–9.Google Scholar
Marwan, A, Crombleholme, TM. The EXIT procedure: principles, pitfalls, and progress. Semin Pediatr Surg. 2006; 15: 107–15.Google Scholar
Tonni, G, De Felice, C, Centini, G, Ginanneschi, C. Cervical and oral teratoma in the fetus: a systematic review of etiology, pathology, diagnosis, treatment and prognosis. Arch Gynecol Obstet. 2010; 282: 355–61.Google Scholar
Omissakin, OO, Kache, SA, Ajike, SO. Congenital epulis: a report of twin cases and review of the literature. Int J Med Biomed. 2016; 5: 130–4.Google Scholar
Vazquez, E, Castellote, A, Mayolas, N, et al. Congenital tumours involving the head, neck and central nervous system. Pediatr Radiol. 2009; 39: 1158–72.Google Scholar
Holley, DG, Martin, GR, Brenner, JI, et al. Diagnosis and management of fetal cardiac tumors: a multicenter experience and review of published reports. J Am Coll Cardiol. 1995; 26: 516–20.Google Scholar
Groves, AM, Fagg, NL, Cook, AC, Allan, LD. Cardiac tumours in intrauterine life. Arch Dis Child. 1992; 67: 1189–92.Google Scholar
Roach, ES, Sparagana, SP. Diagnosis of tuberous sclerosis complex. J Child Neurol. 2004; 19: 643–9.Google Scholar
Green, KW, Bors-Koefoed, R, Pollack, P, Weinbaum, PJ. Antepartum diagnosis and management of multiple fetal cardiac tumors. J Ultrasound Med. 1991; 10: 697–9.Google Scholar
Mehta, AV. Rhabdomyoma and ventricular preexcitation syndrome: a report of two cases and review of literature. Am J Dis Child. 1993; 147: 669–71.Google Scholar
Barnes, BT, Procaccini, D, Crino, J, et al. Maternal sirolimus therapy for fetal rhabdomyomas. N Engl J Med. 2018; 378: 1844–5.Google Scholar
Azizkhan, RG, Crombleholme, TM. Congenital cystic lung disease: contemporary antenatal and postnatal management. Pediatr Surg Int. 2008; 24: 643–57.Google Scholar
Oepkes, D, Devlieger, R, Lopriore, E, Klumper, FJ. Successful ultrasound-guided laser treatment of fetal hydrops caused by pulmonary sequestration. Ultrasound Obstet Gynecol. 2007; 29: 457–9.Google Scholar
Coleman, AM, Merrow, AC, Crombleholme, TM, Jaekle, R, Lim, FY. Fetal MRI of torsed bronchopulmonary sequestration with tension hydrothorax and hydrops in a twin gestation. Fetal Diagn Ther. 2016; 40: 156–60.Google Scholar
Witlox, RS, Lopriore, E, Oepkes, D. Prenatal interventions for fetal lung lesions. Prenatal Diagn. 2011; 31: 628–36.Google Scholar
Ruano, R, da Silva, MM, Salustiano, EM, et al. Percutaneous laser ablation under ultrasound guidance for fetal hyperechogenic microcystic lung lesions with hydrops : a single center cohort and literature review. Prenatal Diagn. 2012; 32: 1127–32.Google Scholar
Stocker, JT, Madewell, JE, Drake, RM. Congenital cystic adenomatoid malformation of the lung. Classification and morphologic spectrum. Hum Pathol. 1977; 8: 155–71.Google Scholar
Diamond, IR, Wales, PW, Smith, SD, Fecteau, A. Survival after CCAM associated with ascites: a report of a case and review of the literature. J Pediatr Surg. 2003; 38: E1–3.Google Scholar
Crombleholme, TM, Coleman, B, Hedrick, H, et al. Cystic adenomatoid malformation volume ratio predicts outcome in prenatally diagnosed cystic adenomatoid malformation of the lung. J Pediatr Surg. 2002; 37: 331–8.Google Scholar
Makin, E, Davenport, M. Fetal and neonatal liver tumours. Early Hum Dev. 2010; 86: 637.Google Scholar
Isaacs, H Jr. Fetal and neonatal hepatic tumors. J Pediatr Surg. 2007; 42: 1797–803.Google Scholar
Kamata, S, Nose, K, Sawai, T, et al. Fetal mesenchymal hamartoma of the liver: report of a case. J Pediatr Surg. 2003; 38: 639–41.Google Scholar
Foucar, E, Williamson, RA, Yiu-Chiu, V, Varner, MW, Kay, BR. Mesenchymal hamartoma of the liver identified by fetal sonography. AJR Am J Roentgenol. 1983; 140: 970–2.Google Scholar
Jones, VS, Cohen, RC. Atypical congenital mesoblastic nephroma presenting in the perinatal period. Pediatr Surg Int. 2007; 23: 205–9.Google Scholar
Linam, LE, Yu, X, Calvo-Garcia, MA, et al. Contribution of magnetic resonance imaging to prenatal differential diagnosis of renal tumors: report of two cases and review of the literature. Fetal Diagn Ther. 2010; 28: 100–8.Google Scholar
Powis, M. Neonatal renal tumours. Early Hum Dev. 2010; 86: 607–12.Google Scholar
Leclair, M-D, El-Ghoneimi, A, Audry, G, et al. The outcome of prenatally diagnosed renal tumors. J Urol. 2005; 173: 186–9.Google Scholar
Grundy, RG, Pritchard, J, Baraitser, M, Risdon, A, Robards, M. Perlman and Wiedemann–Beckwith syndromes: two distinct conditions associated with Wilms’ tumour. Eur J Pediatr. 1992; 151: 895–8.Google Scholar
Nuchtern, JG. Perinatal neuroblastoma. Semin Pediatr Surg. 2006; 15: 1016.Google Scholar
Athanassiadou, F, Kourti, M, Papageorgiou, T, Drevelegas, A, Zaramboukas, T. Prenatally diagnosed cystic neuroblastoma. Pediatr Blood Cancer. 2005; 44: 290–1.Google Scholar
Jennings, RW, LaQuaglia, MP, Leong, K, Hendren, WH, Adzick, NS. Fetal neuroblastoma: prenatal diagnosis and natural history. J Pediatr Surg. 1993; 28: 1168–74.Google Scholar
Askin, J, Geschickter, C. Neuroblastoma of the adrenal in children. J Pediatr. 1935; 7: 157–8.Google Scholar
Forrester, MB, Merz, RD. Descriptive epidemiology of teratoma in infants, Hawaii, 1986–2001. Paediatr Perinat Epidemiol. 2006; 20: 54–8.Google Scholar
Altman, RP, Randolph, JG, Lilly, JR. Sacrococcygeal teratoma: American Academy of Pediatrics Surgical Section Survey – 1973. J Pediatr Surg. 1974; 9: 389–98.Google Scholar
Chervenak, FA, Isaacson, G, Touloukian, R, et al. Diagnosis and management of fetal teratomas. Obstet Gynecol. 1985; 66: 666–71.Google Scholar
Danzer, E, Hubbard, AM, Hedrick, HL, et al. Diagnosis and characterization of fetal sacrococcygeal teratoma with prenatal MRI. AJR Am J Roentgenol. 2006; 187: W350–6.Google Scholar
Flake, AW, Harrison, MR, Adzick, NS, Laberge, JM, Warsof, SL. Fetal sacrococcygeal teratoma. J Pediatr Surg. 1986; 21: 563–6.Google Scholar
Hedrick, HL, Flake, AW, Crombleholme, TM, et al. Sacrococcygeal teratoma: prenatal assessment, fetal intervention, and outcome. J Pediatr Surg. 2004; 39: 430–8.Google Scholar
Adzick, NS, Harrison, MR. The unborn surgical patient. Curr Probl Surg. 1994; 31: 168.Google Scholar
Bahlmann, F, Wellek, S, Reinhardt, I, et al. Reference values of fetal aortic flow velocity waveforms and associated intra-observer reliability in normal pregnancies. Ultrasound Obstet Gynecol. 2001; 17: 42–9.Google Scholar
el-Qarmalawi, MA, Saddik, M, el Abdel Hadi, F, Muwaffi, R, Nageeb, K. Diagnosis and management of fetal sacrococcygeal teratoma. Int J Gynaecol Obstet. 1990; 31: 275–81.Google Scholar
Adzick, NS, Crombleholme, TM, Morgan, MA, Quinn, TM. A rapidly growing fetal teratoma. Lancet. 1997; 349: 538.Google Scholar
Sananes, N, Javadian, P, Schwach Werneck Britto, I, et al. Technical aspects and effectiveness of percutaneous fetal therapies for large sacrococcygeal teratomas: cohort study and literature review. Ultrasound Obstet Gynecol. 2016; 47: 712–19.Google Scholar
Van Mieghem, T, Al-Ibrahim, A, Deprest, J, et al. Minimally invasive therapy for fetal sacrococcygeal teratoma: case series and systematic review of the literature. Ultrasound Obstet Gynecol. 2014; 43: 611–19.Google Scholar
Cowles, RA, Stolar, CJH, Kandel, JJ, et al. Preoperative angiography with embolization and radiofrequency ablation as novel adjuncts to safe surgical resection of a large, vascular sacrococcygeal teratoma. Pediatr Surg Int. 2006; 22: 554–6.Google Scholar
Misra, D, Pritchard, J, Drake, DP, Kiely, EM, Spitz, L. Markedly improved survival in malignant sacro-coccygeal teratomas – 16 years’ experience. Eur J Pediatr Surg. 1997; 7: 152–5.Google Scholar

References

Greer, JJ. Current concepts on the pathogenesis and etiology of congenital diaphragmatic hernia. Respir Physiol Neurobiol. 2013; 189: 232–40.Google Scholar
Keijzer, R, Puri, P. Congenital diaphragmatic hernia. Semin Pediatr Surg. 2010; 19: 180–5.Google Scholar
Numanoglu, A, Steiner, Z, Millar, A, Cywes, S. Delayed presentation of congenital diaphragmatic hernia. S Afr J Surg. 1997; 35: 74–6.Google Scholar
Tovar, JA. Congenital diaphragmatic hernia. Orphanet J Rare Dis. 2012; 7: 1.Google Scholar
Kitagawa, M, Hislop, A, Boyden, EA, Reid, L. Lung hypoplasia in congenital diaphragmatic hernia. A quantitative study of airway, artery, and alveolar development. Br J Surg. 1971; 58: 342–6.Google Scholar
Rottier, R, Tibboel, D. Fetal lung and diaphragm development in congenital diaphragmatic hernia. Semin Perinatol. 2005; 29: 8693.Google Scholar
Shehata, SM, Sharma, HS, van der Staak, FH, van de Kaa-Hulsbergen, C, Mooi, WJ, Tibboel, D. Remodeling of pulmonary arteries in human congenital diaphragmatic hernia with or without extracorporeal membrane oxygenation. J Pediatr Surg. 2000; 35: 208–15.Google Scholar
Kool, H, Mous, D, Tibboel, D, de Klein, A, Rottier, RJ. Pulmonary vascular development goes awry in congenital lung abnormalities. Birth Defects Res C Embryo Today. 2014; 102: 343–58.Google Scholar
Neonatal Inhaled Nitric Oxide Study Group. Inhaled nitric oxide in full-term and nearly full-term infants with hypoxic respiratory failure. N Engl J Med. 1997; 336: 597604.Google Scholar
Vijfhuize, S, Schaible, T, Kraemer, U, Cohen-Overbeek, TE, Tibboel, D, Reiss, I. Management of pulmonary hypertension in neonates with congenital diaphragmatic hernia. Eur J Pediatr Surg. 2012; 22: 374–83.Google Scholar
Bucher, BT, Guth, RM, Saito, JM, Najaf, T, Warner, BW. Impact of hospital volume on in-hospital mortality of infants undergoing repair of congenital diaphragmatic hernia. Ann Surg. 2010; 252: 635–42.Google Scholar
Downard, CD, Jaksic, T, Garza, JJ, Dzakovic, A, Nemes, L, Jennings, RW, et al. Analysis of an improved survival rate for congenital diaphragmatic hernia. J Pediatr Surg. 2003; 38: 729–32.Google Scholar
Brownlee, EM, Howatson, AG, Davis, CF, Sabharwal, AJ. The hidden mortality of congenital diaphragmatic hernia: a 20-year review. J Pediatr Surg. 2009; 44: 317–20.Google Scholar
Chiu, P, Hedrick, HL. Postnatal management and long-term outcome for survivors with congenital diaphragmatic hernia. Prenat Diagn. 2008; 28: 592603.Google Scholar
Jaillard, SM, Pierrat, V, Dubois, A, Truffert, P, Lequien, P, Wurtz, AJ, et al. Outcome at 2 years of infants with congenital diaphragmatic hernia: a population-based study. Ann Thorac Surg. 2003; 75: 250–6.Google Scholar
van den Hout, L, Schaible, T, Cohen-Overbeek, TE, Hop, W, Siemer, J, van de Ven, K, et al. Actual outcome in infants with congenital diaphragmatic hernia: the role of a standardized postnatal treatment protocol. Fetal Diagn Ther. 2011; 29: 5563.Google Scholar
Muratore, CS, Kharasch, V, Lund, DP, Sheils, C, Friedman, S, Brown, C, et al. Pulmonary morbidity in 100 survivors of congenital diaphragmatic hernia monitored in a multidisciplinary clinic. J Pediatr Surg. 2001; 36: 133–40.Google Scholar
Rocha, G, Azevedo, I, Pinto, JC, Guimaraes, H. Follow-up of the survivors of congenital diaphragmatic hernia. Early Hum Dev. 2012; 88: 255–8.Google Scholar
Vanamo, K, Peltonen, J, Rintala, R, Lindahl, H, Jaaskelainen, J, Louhimo, I. Chest wall and spinal deformities in adults with congenital diaphragmatic defects. J Pediatr Surg. 1996; 31: 851–4.Google Scholar
Masumoto, K, Nagata, K, Uesugi, T, Yamada, T, Kinjo, T, Hikino, S, et al. Risk of respiratory syncytial virus in survivors with severe congenital diaphragmatic hernia. Pediatr Int. 2008; 50: 459–63.Google Scholar
Dillon, PW, Cilley, RE, Mauger, D, Zachary, C, Meier, A. The relationship of pulmonary artery pressure and survival in congenital diaphragmatic hernia. J Pediatr Surg. 2004; 39: 307–12.Google Scholar
Bagolan, P, Morini, F. Long-term follow up of infants with congenital diaphragmatic hernia. Semin Pediatr Surg. 2007; 16: 134–44.Google Scholar
Caruso, AM, Di Pace, MR, Catalano, P, Farina, F, Casuccio, A, Cimador, M, et al. Gastroesophageal reflux in patients treated for congenital diaphragmatic hernia: short- and long-term evaluation with multichannel intraluminal impedance. Pediatr Surg Int. 2013; 29: 553–9.Google Scholar
Qi, B, Soto, C, Diez-Pardo, JA, Tovar, JA. An experimental study on the pathogenesis of gastroesophageal reflux after repair of diaphragmatic hernia. J Pediatr Surg. 1997; 32: 1310–13.Google Scholar
Leeuwen, L, Walker, K, Halliday, R, Karpelowsky, J, Fitzgerald, DA. Growth in children with congenital diaphragmatic hernia during the first year of life. J Pediatr Surg. 2014; 49: 1363–6.Google Scholar
Vanamo, K, Rintala, RJ, Lindahl, H, Louhimo, I. Long-term gastrointestinal morbidity in patients with congenital diaphragmatic defects. J Pediatr Surg. 1996; 31: 551–4.Google Scholar
Rasheed, A, Tindall, S, Cueny, DL, Klein, MD, Delaney-Black, V. Neurodevelopmental outcome after congenital diaphragmatic hernia: Extracorporeal membrane oxygenation before and after surgery. J Pediatr Surg. 2001; 36: 539–44.Google Scholar
Morini, F, Capolupo, I, Masi, R, Ronchetti, MP, Locatelli, M, Corchia, C, et al. Hearing impairment in congenital diaphragmatic hernia: the inaudible and noiseless foot of time. J Pediatr Surg. 2008; 43: 380–4.Google Scholar
Javidnia, H, Vaccani, JP. Progressive sensorineural hearing loss in children with congenital diaphragmatic hernias. J Otolaryngol Head Neck Surg. 2009; 38: 2931.Google Scholar
Gallot, D, Boda, C, Ughetto, S, Perthus, I, Robert-Gnansia, E, Francannet, C, et al. Prenatal detection and outcome of congenital diaphragmatic hernia: a French registry-based study. Ultrasound Obstet Gynecol. 2007; 29: 276–83.Google Scholar
Doné, E, Gucciardo, L, Van Mieghem, T, Devriendt, K, Allegaert, K, Brady, P, et al. Clinically relevant discordances identified after tertiary reassessment of fetuses with isolated congenital diaphragmatic hernia. Prenat Diagn. 2017; 37: 883–8.Google Scholar
Deprest, J, Jani, J, Van Schoubroeck, D, Cannie, M, Gallot, D, Dymarkowski, S, et al. Current consequences of prenatal diagnosis of congenital diaphragmatic hernia. J Pediatr Surg. 2006; 41: 423–30.Google Scholar
Doné, E, Gucciardo, L, Van Mieghem, T. Clinically relevant discordances identified after tertiary reassessment of fetuses with isolated congenital diaphragmatic hernia. Prenat Diagn. 2017; 37: 883–8.Google Scholar
Metkus, AP, Filly, RA, Stringer, MD, Harrison, MR, Adzick, NS. Sonographic predictors of survival in fetal diaphragmatic hernia. J Pediatr Surg. 1996; 31: 148–51; discussion 151–2.Google Scholar
Kitano, Y, Okuyama, H, Saito, M, Usui, N, Morikawa, N, Masumoto, K, et al. Re-evaluation of stomach position as a simple prognostic factor in fetal left congenital diaphragmatic hernia: a multicenter survey in Japan. Ultrasound Obstet Gynecol. 2011; 37: 277–82.Google Scholar
Mayer, S, Klaritsch, P, Petersen, S, Doné, E, Sandaite, I, Till, H, et al. The correlation between lung volume and liver herniation measurements by fetal MRI in isolated congenital diaphragmatic hernia: a systematic review and meta-analysis of observational studies. Prenat Diagn. 2011; 31: 1086–96.Google Scholar
Cannie, M, Jani, J, Chaffiotte, C, Vaast, P, Deruelle, P, Houfflin-Debarge, V, et al. Quantification of intrathoracic liver herniation by magnetic resonance imaging and prediction of postnatal survival in fetuses with congenital diaphragmatic hernia. Ultrasound Obstet Gynecol. 2008; 32: 627–32.Google Scholar
Doné, E, Gratacos, E, Nicolaides, K, Allegaert, K, Valencia, C, Castanon, M, et al. Predictors of neonatal morbidity in fetuses with severe isolated congenital diaphragmatic hernia undergoing fetoscopic tracheal occlusion. Ultrasound Obstet Gynecol. 2013 ; 42 : 7783.Google Scholar
Cordier, AG, Cannie, MM, Guilbaud, L, De Laveaucoupet, J, Martinovic, J, Nowakowska, D, et al. Stomach position versus liver-to-thoracic volume ratio in left-sided congenital diaphragmatic hernia. J Matern Fetal Neonatal Med. 2015; 28: 190–5.Google Scholar
DeKoninck, P, Gomez, O, Sandaite, I, Richter, J, Nawapun, K, Eerdekens, A, et al. Right-sided congenital diaphragmatic hernia in a decade of fetal surgery. BJOG. 2015; 122: 940–6.Google Scholar
Lin, AE, Pober, BR, Adatia, I. Congenital diaphragmatic hernia and associated cardiovascular malformations: type, frequency, and impact on management. Am J Med Genet C Semin Med Genet. 2007; 145C: 201–16.Google Scholar
Losty, PD, Vanamo, K, Rintala, RJ, Donahoe, PK, Schnitzer, JJ, Lloyd, DA. Congenital diaphragmatic hernia–does the side of the defect influence the incidence of associated malformations? J Pediatr Surg. 1998; 33: 507–10.Google Scholar
Dillon, E, Renwick, M, Wright, C. Congenital diaphragmatic herniation: antenatal detection and outcome. Br J Radiol. 2000; 73: 360–5.Google Scholar
Cohen, MS, Rychik, J, Bush, DM, Tian, ZY, Howell, LJ, Adzick, NS, et al. Influence of congenital heart disease on survival in children with congenital diaphragmatic hernia. J Pediatr. 2002; 141: 2530.Google Scholar
Siebert, JR, Haas, JE, Beckwith, JB. Left ventricular hypoplasia in congenital diaphragmatic hernia. J Pediatr Surg. 1984; 19: 567–71.Google Scholar
Van Mieghem, T, Cruz-Martinez, R, Allegaert, K, Dekoninck, P, Castanon, M, Sandaite, I, et al. Outcome of fetuses with congenital diaphragmatic hernia and associated intrafetal fluid effusions managed in the era of fetal surgery. Ultrasound Obstet Gynecol. 2012; 39: 50–5.Google Scholar
Yu, L, Wynn, J, Ma, L, Guha, S, Mychaliska, GB, Crombleholme, TM, et al. De novo copy number variants are associated with congenital diaphragmatic hernia. J Med Genet. 2012; 49: 650–9.Google Scholar
Pober, BR, Russell, MK, Ackerman, KG. Congenital Diaphragmatic Hernia Overview. In Adam, MP, Ardinger, HH, Pagon, RA, Wallace, SE, Bean, LJH, Stephens, K, et al., eds., GeneReviews. Seattle: University of Washington, 1993.Google Scholar
Holder, AM, Klaassens, M, Tibboel, D, de Klein, A, Lee, B, Scott, DA. Genetic factors in congenital diaphragmatic hernia. Am J Hum Genet. 2007; 80: 825–45.Google Scholar
Lurie, IW. Where to look for the genes related to diaphragmatic hernia? Genet Couns. 2003; 14: 7593.Google Scholar
Slavotinek, AM. The genetics of common disorders – congenital diaphragmatic hernia. Eur J Med Genet. 2014; 57: 418–23.Google Scholar
Kardon, G, Ackerman, KG. Congenital diaphragmatic hernias: from genes to mechanisms to therapies. Dis Model Mech. 2017; 10: 955–70.Google Scholar
Jani, J, Nicolaides, KH, Keller, RL, Benachi, A, Peralta, CF, Favre, R, et al. Observed to expected lung area to head circumference ratio in the prediction of survival in fetuses with isolated diaphragmatic hernia. Ultrasound Obstet Gynecol. 2007; 30: 6771.Google Scholar
Cruz-Martinez, R, Castanon, M, Moreno-Alvarez, O, Acosta-Rojas, R, Martinez, JM, Gratacós, E. Usefulness of lung-to-head ratio and intrapulmonary arterial Doppler in predicting neonatal morbidity in fetuses with congenital diaphragmatic hernia treated with fetoscopic tracheal occlusion. Ultrasound Obstet Gynecol. 2013; 41: 5965.Google Scholar
Garcia, AV, Fingeret, AL, Thirumoorthi, AS, Hahn, E, Leskowitz, MJ, Aspelund, G, et al. Lung to head ratio in infants with congenital diaphragmatic hernia does not predict long term pulmonary hypertension. J Pediatr Surg. 2013; 48: 154–7.Google Scholar
Lipshutz, GS, Albanese, CT, Feldstein, VA, Jennings, RW, Housley, HT, Beech, R, et al. Prospective analysis of lung-to-head ratio predicts survival for patients with prenatally diagnosed congenital diaphragmatic hernia. J Pediatr Surg. 1997; 32: 1634–6.Google Scholar
Peralta, CF, Cavoretto, P, Csapo, B, Vandecruys, H, Nicolaides, KH. Assessment of lung area in normal fetuses at 12–32 weeks. Ultrasound Obstet Gynecol. 2005; 26: 718–24.Google Scholar
Hidaka, N, Murata, M, Sasahara, J, Ishii, K, Mitsuda, N. Correlation between lung to thorax transverse area ratio and observed/expected lung area to head circumference ratio in fetuses with left-sided diaphragmatic hernia. Congenit Anom. 2015; 55: 81–4.Google Scholar
Quintero, RA, Quintero, LF, Chmait, R, Gomez Castro, L, Korst, LM, Fridman, M, et al. The quantitative lung index (QLI): a gestational age-independent sonographic predictor of fetal lung growth. Am J Obstet Gynecol. 2011; 205: 544. e1–8.Google Scholar
Ruano, R, Takashi, E, da Silva, MM, Campos, JA, Tannuri, U, Zugaib, M. Prediction and probability of neonatal outcome in isolated congenital diaphragmatic hernia using multiple ultrasound parameters. Ultrasound Obstet Gynecol. 2012; 39: 42–9.Google Scholar
Russo, FM, Cordier, AG, De Catte, L, Saada, J, Benachi, A, Deprest, J, et al. Proposal for standardized prenatal ultrasound assessment of the fetus with congenital diaphragmatic hernia by the European Reference Network on Rare Inherited and Congenital Anomalies (ERNICA). Prenat Diagn. 2018; 38: 629–37.Google Scholar
Harrison, MR, Langer, JC, Adzick, NS, Golbus, MS, Filly, RA, Anderson, RL, et al. Correction of congenital diaphragmatic hernia in utero, V. Initial clinical experience. J Pediatr Surg. 1990; 25: 4755; discussion 56–7.Google Scholar
Albanese, CT, Lopoo, J, Goldstein, RB, Filly, RA, Feldstein, VA, Calen, PW, et al. Fetal liver position and perinatal outcome for congenital diaphragmatic hernia. Prenat Diagn. 1998; 18: 1138–42.Google Scholar
Rypens, F, Metens, T, Rocourt, N, Sonigo, P, Brunelle, F, Quere, MP, et al. Fetal lung volume: estimation at MR imaging-initial results. Radiology. 2001; 219: 236–41.Google Scholar
Jani, J, Cannie, M, Done, E, Van Mieghem, T, Van Schoubroeck, D, Gucciardo, L, et al. Relationship between lung area at ultrasound examination and lung volume assessment with magnetic resonance imaging in isolated congenital diaphragmatic hernia. Ultrasound Obstet Gynecol. 2007; 30: 855–60.Google Scholar
Gorincour, G, Bouvenot, J, Mourot, MG, Sonigo, P, Chaumoitre, K, Garel, C, et al. Prenatal prognosis of congenital diaphragmatic hernia using magnetic resonance imaging measurement of fetal lung volume. Ultrasound Obstet Gynecol. 2005; 26: 738–44.Google Scholar
Cannie, M, Jani, JC, De Keyzer, F, Devlieger, R, Van Schoubroeck, D, Witters, I, et al. Fetal body volume: use at MR imaging to quantify relative lung volume in fetuses suspected of having pulmonary hypoplasia. Radiology. 2006; 241: 847–53.Google Scholar
Nawapun, K, Eastwood, M, Sandaite, I, DeKoninck, P, Claus, F, Richter, J, et al. Correlation of observed-to-expected total fetal lung volume with intrathoracic organ herniation on magnetic resonance imaging in fetuses with isolated left-sided congenital diaphragmatic hernia. Ultrasound Obstet Gynecol. 2015; 46: 162–7.Google Scholar
Cannie, MM, Cordier, AG, De Laveaucoupet, J, Franchi-Abella, S, Cagneaux, M, Prodhomme, O, et al. Liver-to-thoracic volume ratio: use at MR imaging to predict postnatal survival in fetuses with isolated congenital diaphragmatic hernia with or without prenatal tracheal occlusion. Eur Radiol. 2013; 23: 1299–305.Google Scholar
Bebbington, M, Victoria, T, Danzer, E, Moldenhauer, J, Khalek, N, Johnson, M, et al. Comparison of ultrasound and magnetic resonance imaging parameters in predicting survival in isolated left-sided congenital diaphragmatic hernia. Ultrasound Obstet Gynecol. 2014; 43: 670–4.Google Scholar
Schaible, T, Busing, KA, Felix, JF, Hop, WC, Zahn, K, Wessel, L, et al. Prediction of chronic lung disease, survival and need for ECMO therapy in infants with congenital diaphragmatic hernia: additional value of fetal MRI measurements? Eur J Radiol. 2012; 81: 1076–82.Google Scholar
Mayer, S, Klaritsch, P, Petersen, S, Done, E, Sandaite, I, Till, H, et al. The correlation between lung volume and liver herniation measurements by fetal MRI in isolated congenital diaphragmatic hernia: a systematic review and meta-analysis of observational studies. Prenat Diagn. 2011; 31: 1086–96.Google Scholar
Claus, F, Sandaite, I, Dekoninck, P, Moreno, O, Cruz Martinez, R, Van Mieghem, T, et al. Prenatal anatomical imaging in fetuses with congenital diaphragmatic hernia. Fetal Diagn Ther. 2011; 29: 88100.Google Scholar
Benachi, A, Cordier, AG, Cannie, M, Jani, J. Advances in prenatal diagnosis of congenital diaphragmatic hernia. Semin Fetal Neonatal Med. 2014; 19: 331–7.Google Scholar
Basta, AM, Lusk, LA, Keller, RL, Filly, RA. Fetal stomach position predicts neonatal outcomes in isolated left-sided congenital diaphragmatic Hernia. Fetal Diagn Ther. 2016; 39: 248–55.Google Scholar
Cordier, AG, Jani, JC, Cannie, MM, Rodo, C, Fabietti, I, Persico, N, et al. Stomach position in prediction of survival in left-sided congenital diaphragmatic hernia with or without fetoscopic endoluminal tracheal occlusion. Ultrasound Obstet Gynecol. 2015; 46: 155–61.Google Scholar
Sananes, N, Britto, I, Akinkuotu, AC, Olutoye, OO, Cass, DL, Sangi-Haghpeykar, H, et al. Improving the prediction of neonatal outcomes in isolated left-sided congenital diaphragmatic hernia by direct and indirect sonographic assessment of liver herniation. J Ultrasound Med. 2016; 35: 1437–43.Google Scholar
Cruz-Martinez, R, Figueras, F, Moreno-Alvarez, O, Martinez, JM, Gomez, O, Hernandez-Andrade, E, et al. Learning curve for lung area to head circumference ratio measurement in fetuses with congenital diaphragmatic hernia. Ultrasound Obstet Gynecol. 2010; 36: 32–6.Google Scholar
Senat, MV, Bouchghoul, H, Stirnemann, J, Vaast, P, Boubnova, J, Begue, L, et al. Prognosis of isolated congenital diaphragmatic hernia using lung-to-head circumference ratio: variability across centers in a national perinatal network. Ultrasound Obstet Gynecol. 2018; 51: 208–13.Google Scholar
Graham, G, Devine, PC. Antenatal diagnosis of congenital diaphragmatic hernia. Semin Perinatol. 2005; 29: 6976.Google Scholar
Dott, MM, Wong, LY, Rasmussen, SA. Population-based study of congenital diaphragmatic hernia: risk factors and survival in Metropolitan Atlanta, 1968-1999. Birth Defects Res A Clin Mol Teratol. 2003; 67: 261–7.Google Scholar
Slavotinek, AM. Fryns syndrome: a review of the phenotype and diagnostic guidelines. Am J Med Genet A. 2004; 124A: 427–33.Google Scholar
Wat, MJ, Veenma, D, Hogue, J, Holder, AM, Yu, Z, Wat, JJ, et al. Genomic alterations that contribute to the development of isolated and non-isolated congenital diaphragmatic hernia. J Med Genet. 2011; 48: 299307.Google Scholar
Magoulas, PL, El-Hattab, AW. Chromosome 15q24 microdeletion syndrome. Orphanet J Rare Dis. 2012; 7: 2.Google Scholar
Slavotinek, AM, Moshrefi, A, Davis, R, Leeth, E, Schaeffer, GB, Burchard, GE, et al. Array comparative genomic hybridization in patients with congenital diaphragmatic hernia: mapping of four CDH-critical regions and sequencing of candidate genes at 15q26.1-15q26.2. Eur J Hum Genet. 2006; 14: 9991008.Google Scholar
Qidwai, K, Pearson, DM, Patel, GS, Pober, BR, Immken, LL, Cheung, SW, et al. Deletions of Xp provide evidence for the role of holocytochrome C-type synthase (HCCS) in congenital diaphragmatic hernia. Am J Med Genet A. 2010; 152A: 1588–90.Google Scholar
Tautz, J, Veenma, D, Eussen, B, Joosen, L, Poddighe, P, Tibboel, D, et al. Congenital diaphragmatic hernia and a complex heart defect in association with Wolf-Hirschhorn syndrome. Am J Med Genet A. 2010; 152A: 2891–4.Google Scholar
Chen, CP, Lee, CC, Pan, CW, Kir, TY, Chen, BF. Partial trisomy 8q and partial monosomy 15q associated with congenital hydrocephalus, diaphragmatic hernia, urinary tract anomalies, congenital heart defect and kyphoscoliosis. Prenat Diagn. 1998; 18: 1289–93.Google Scholar
Pober, BR, Russell, MK, Ackerman, KG. Congenital Diaphragmatic Hernia Overview. In Adam, MP, Ardinger, HH, Pagon, RA, Wallace, SE, Bean, LJH, Stephens, K, et al., eds. GeneReviews. Seattle: University of Washington, 1993.Google Scholar
Wilkens, A, Liu, H, Park, K, Campbell, LB, Jackson, M, Kostanecka, A, et al. Novel clinical manifestations in Pallister-Killian syndrome: comprehensive evaluation of 59 affected individuals and review of previously reported cases. Am J Med Genet A. 2012; 158A: 3002–17.Google Scholar
Slavotinek, AM. Single gene disorders associated with congenital diaphragmatic hernia. Am J Med Genet C Semin Med Genet. 2007; 145C: 172–83.Google Scholar
Han, XY, Wu, SS, Conway, DH, Pawel, BR, Punnett, HH, Martin, RA, et al. Truncus arteriosus and other lethal internal anomalies in Goltz syndrome. Am J Med Genet. 2000; 90: 45–8.Google Scholar
Enns, GM, Cox, VA, Goldstein, RB, Gibbs, DL, Harrison, MR, Golabi, M. Congenital diaphragmatic defects and associated syndromes, malformations, and chromosome anomalies: a retrospective study of 60 patients and literature review. Am J Med Genet. 1998; 79: 215–25.Google Scholar
Li, M, Shuman, C, Fei, YL, Cutiongco, E, Bender, HA, Stevens, C, et al. GPC3 mutation analysis in a spectrum of patients with overgrowth expands the phenotype of Simpson-Golabi-Behmel syndrome. Am J Med Genet. 2001; 102: 161–8.Google Scholar
Russo, FM, De Coppi, P, Allegaert, K, Toelen, J, van der Veeken, L, Attilakos, G, et al. Current and future antenatal management of isolated congenital diaphragmatic hernia. Semin Fetal Neonatal Med. 2017; 22: 383–90.Google Scholar

References

EUROCAT (2015). EUROCAT Statistical Monitoring Report – 2012. www.eurocat-network.eu/content/Stat-Mon-Report-2012.pdfGoogle Scholar
Harting, MT, Lally, KP. The Congenital Diaphragmatic Hernia Study Group registry update. Semin Fetal Neonatal Med. 2014; 19: 370–5.Google Scholar
Claus, F, Sandaite, I, Dekoninck, P, Moreno, O, Cruz Martinez, R, Van Mieghem, T, et al. Prenatal anatomical imaging in fetuses with congenital diaphragmatic hernia. Fetal Diagn Ther. 2011; 29: 88100.Google Scholar
Harrison, MR, Langer, JC, Adzick, NS, Golbus, MS, Filly, RA, Anderson, RL, et al. Correction of congenital diaphragmatic hernia in utero, V. Initial clinical experience. J Pediatr Surg. 1990; 25: 4755; discussion 6–7.Google Scholar
Wilson, JM, DiFiore, JW, Peters, CA. Experimental fetal tracheal ligation prevents the pulmonary hypoplasia associated with fetal nephrectomy: possible application for congenital diaphragmatic hernia. J Pediatr Surg. 1993; 28: 1433–9; discussion 1439–40.Google Scholar
Hedrick, MH, Estes, JM, Sullivan, KM, Bealer, JF, Kitterman, JA, Flake, AW, et al. Plug the lung until it grows (PLUG): a new method to treat congenital diaphragmatic hernia in utero. J Pediatr Surg. 1994; 29: 612–17.Google Scholar
Flageole, H, Evrard, VA, Piedboeuf, B, Laberge, JM, Lerut, TE, Deprest, JA. The plug-unplug sequence: an important step to achieve type II pneumocyte maturation in the fetal lamb model. J Pediatr Surg. 1998; 33: 299303.Google Scholar
Khan, PA, Cloutier, M, Piedboeuf, B. Tracheal occlusion: a review of obstructing fetal lungs to make them grow and mature. Am J Med Genet C Semin Med Genet. 2007; 145C: 125–38.Google Scholar
Marwan, AI, Shabeka, U, Dobrinskikh, E. Suggested mechanisms of tracheal occlusion mediated accelerated fetal lung growth: a case for heterogeneous topological zones. Front Pediatr. 2017; 5: 295.Google Scholar
Deprest, J, Gratacós, E, Nicolaides, KH, Group, FT. Fetoscopic tracheal occlusion (FETO) for severe congenital diaphragmatic hernia: evolution of a technique and preliminary results. Ultrasound Obstet Gynecol. 2004; 24: 121–6.Google Scholar
Metkus, AP, Filly, RA, Stringer, MD, Harrison, MR, Adzick, NS. Sonographic predictors of survival in fetal diaphragmatic hernia. J Pediatr Surg. 1996; 31: 148–51; discussion 151–2.Google Scholar
Gallot, D, Boda, C, Ughetto, S, Perthus, I, Robert-Gnansia, E, Francannet, C, et al. Prenatal detection and outcome of congenital diaphragmatic hernia: a French registry-based study. Ultrasound Obstet Gynecol. 2007; 29: 276–83.Google Scholar
Jani, J, Nicolaides, KH, Keller, RL, Benachi, A, Peralta, CF, Favre, R, et al. Observed to expected lung area to head circumference ratio in the prediction of survival in fetuses with isolated diaphragmatic hernia. Ultrasound Obstet Gynecol. 2007; 30: 6771.Google Scholar
Jani, JC, Nicolaides, KH, Gratacós, E, Valencia, CM, Done, E, Martinez, JM, et al. Severe diaphragmatic hernia treated by fetal endoscopic tracheal occlusion. Ultrasound Obstet Gynecol. 2009; 34: 304–10.Google Scholar
DeKoninck, P, Gomez, O, Sandaite, I, Richter, J, Nawapun, K, Eerdekens, A, et al. Right-sided congenital diaphragmatic hernia in a decade of fetal surgery. BJOG. 2015; 122: 940–6.Google Scholar
Al-Maary, J, Eastwood, MP, Russo, FM, Deprest, JA, Keijzer, R. Fetal tracheal occlusion for severe pulmonary hypoplasia in isolated congenital diaphragmatic hernia: a systematic review and meta-analysis of survival. Ann Surg. 2016; 264: 929–33.Google Scholar
Seravalli, V, Jelin, EB, Miller, JL, Tekes, A, Vricella, L, Baschat, AA. Fetoscopic tracheal occlusion for treatment of non-isolated congenital diaphragmatic hernia. Prenat Diagn. 2017; 37: 1046–9.Google Scholar
Van Mieghem, T, Cruz-Martinez, R, Allegaert, K, Dekoninck, P, Castanon, M, Sandaite, I, et al. Outcome of fetuses with congenital diaphragmatic hernia and associated intrafetal fluid effusions managed in the era of fetal surgery. Ultrasound Obstet Gynecol. 2012; 39: 50–5.Google Scholar
Van Mieghem, T, Cruz-Martinez, R, Allegaert, K, Dekoninck, P, Castanon, M, Sandaite, I, et al. Outcome of fetuses with congenital diaphragmatic hernia and associated intrafetal fluid effusions managed in the era of fetal surgery. Ultrasound Obstet Gynecol. 2012; 39: 50-5.Google Scholar
Deprest, J, Brady, P, Nicolaides, K, Benachi, A, Berg, C, Vermeesch, J, et al. Prenatal management of the fetus with isolated congenital diaphragmatic hernia in the era of the TOTAL trial. Semin Fetal Neonatal Med. 2014; 19: 338–48.Google Scholar
Friede, T, Kieser, M. Sample size recalculation in internal pilot study designs: a review. Biom J. 2006; 48: 537–55.Google Scholar
Friede, T, Mitchell, C, Muller-Velten, G. Blinded sample size reestimation in non-inferiority trials with binary endpoints. Biom J. 2007; 49: 903–16.Google Scholar
O’Brien, PC, Fleming, TR. A multiple testing procedure for clinical trials. Biometrics. 1979; 35: 549–56.Google Scholar
Rodrigues, HC, Deprest, J, Berg, PP. When referring physicians and researchers disagree on equipoise: the TOTAL trial experience. Prenat Diagn. 2011; 31: 589–94.Google Scholar
Van der Veeken, L, Russo, FM, De Catte, L, Gratacós, E, Benachi, A, Ville, Y, et al. Fetoscopic endoluminal tracheal occlusion and reestablishment of fetal airways for congenital diaphragmatic hernia. Gynecol Surg. 2018; 15: 9.Google Scholar
Deprest, J, Jani, J, Cannie, M, Debeer, A, Vandevelde, M, Done, E, et al. Prenatal intervention for isolated congenital diaphragmatic hernia. Curr Opin Obstet Gynecol. 2006; 18: 355–67.Google Scholar
Peralta, CF, Jani, JC, Van Schoubroeck, D, Nicolaides, KH, Deprest, JA. Fetal lung volume after endoscopic tracheal occlusion in the prediction of postnatal outcome. Am J Obstet Gynecol. 2008; 198: 60. e1–5.Google Scholar
Done, E, Gratacós, E, Nicolaides, KH, Allegaert, K, Valencia, C, Castanon, M, et al. Predictors of neonatal morbidity in fetuses with severe isolated congenital diaphragmatic hernia undergoing fetoscopic tracheal occlusion. Ultrasound Obstet Gynecol. 2013; 42: 7783.Google Scholar
Jimenez, JA, Eixarch, E, DeKoninck, P, Bennini, JR, Devlieger, R, Peralta, CF, et al. Balloon removal after fetoscopic endoluminal tracheal occlusion for congenital diaphragmatic hernia. Am J Obstet Gynecol. 2017; 217: 78. e1–11.Google Scholar
Vijfhuize, S, Schaible, T, Kraemer, U, Cohen-Overbeek, TE, Tibboel, D, Reiss, I. Management of pulmonary hypertension in neonates with congenital diaphragmatic hernia. Eur J Pediatr Surg. 2012; 22: 374–83.Google Scholar
Villamor, E, Le Cras, TD, Horan, MP, Halbower, AC, Tuder, RM, Abman, SH. Chronic intrauterine pulmonary hypertension impairs endothelial nitric oxide synthase in the ovine fetus. Am J Physiol. 1997; 272: L1013–20.Google Scholar
Hanson, KA, Ziegler, JW, Rybalkin, SD, Miller, JW, Abman, SH, Clarke, WR. Chronic pulmonary hypertension increases fetal lung cGMP phosphodiesterase activity. Am J Physiol. 1998; 275: L931–41.Google Scholar
Barnett, CF, Machado, RF. Sildenafil in the treatment of pulmonary hypertension. Vasc Health Risk Manag. 2006; 2: 411–22.Google Scholar
Wharton, J, Strange, JW, Moller, GM, Growcott, EJ, Ren, X, Franklyn, AP, et al. Antiproliferative effects of phosphodiesterase type 5 inhibition in human pulmonary artery cells. Am J Respir Crit Care Med. 2005; 172: 105–13.Google Scholar
Barst, RJ, Ivy, DD, Gaitan, G, Szatmari, A, Rudzinski, A, Garcia, AE, et al. A randomized, double-blind, placebo-controlled, dose-ranging study of oral sildenafil citrate in treatment-naive children with pulmonary arterial hypertension. Circulation. 2012; 125: 324–34.Google Scholar
Barst, RJ, Beghetti, M, Pulido, T, Layton, G, Konourina, I, Zhang, M, et al. STARTS-2: long-term survival with oral sildenafil monotherapy in treatment-naive pediatric pulmonary arterial hypertension. Circulation. 2014; 129: 1914–23.Google Scholar
Steinhorn, RH, Kinsella, JP, Pierce, C, Butrous, G, Dilleen, M, Oakes, M, et al. Intravenous sildenafil in the treatment of neonates with persistent pulmonary hypertension. J Pediatr. 2009; 155: 841–7. e1.Google Scholar
Sanchez Luna, M, Franco, ML, Bernardo, B. Therapeutic strategies in pulmonary hypertension of the newborn: where are we now? Curr Med Chem. 2012; 19: 4640–53.Google Scholar
Lacassie, HJ, Germain, AM, Valdes, G, Fernandez, MS, Allamand, F, Lopez, H. Management of Eisenmenger syndrome in pregnancy with sildenafil and L-arginine. Obstet Gynecol. 2004; 103: 1118–20.Google Scholar
Molelekwa, V, Akhter, P, McKenna, P, Bowen, M, Walsh, K. Eisenmenger’s syndrome in a 27 week pregnancy –management with bosentan and sildenafil. Ir Med J. 2005; 98: 87–8.Google Scholar
Streit, M, Speich, R, Fischler, M, Ulrich, S. Successful pregnancy in pulmonary arterial hypertension associated with systemic lupus erythematosus: a case report. J Med Case Rep. 2009; 3: 7255.Google Scholar
Samangaya, RA, Mires, G, Shennan, A, Skillern, L, Howe, D, McLeod, A, et al. A randomised, double-blinded, placebo-controlled study of the phosphodiesterase type 5 inhibitor sildenafil for the treatment of preeclampsia. Hypertens Pregnancy. 2009; 28: 369–82.Google Scholar
von Dadelszen, P, Dwinnell, S, Magee, LA, Carleton, BC, Gruslin, A, Lee, B, et al. Sildenafil citrate therapy for severe early-onset intrauterine growth restriction. BJOG. 2011; 118: 624–8.Google Scholar
Maher, MA, Sayyed, TM, Elkhouly, N. Sildenafil citrate therapy for oligohydramnios: a randomized controlled trial. Obstet Gynecol. 2017; 129: 615–20.Google Scholar
Luong, C, Rey-Perra, J, Vadivel, A, Gilmour, G, Sauve, Y, Koonen, D, et al. Antenatal sildenafil treatment attenuates pulmonary hypertension in experimental congenital diaphragmatic hernia. Circulation. 2011; 123: 2120–31.Google Scholar
Yamamoto, Y, Thebaud, B, Vadivel, A, Eaton, F, Jain, V, Hornberger, LK. Doppler parameters of fetal lung hypoplasia and impact of sildenafil. Am J Obstet Gynecol. 2014; 211: 263. e1–8.Google Scholar
Kattan, J, Cespedes, C, Gonzalez, A, Vio, CP. Sildenafil stimulates and dexamethasone inhibits pulmonary vascular development in congenital diaphragmatic hernia rat lungs. Neonatology. 2014; 106: 7480.Google Scholar
Lemus-Varela Mde, L, Soliz, A, Gomez-Meda, BC, Zamora-Perez, AL, Ornelas-Aguirre, JM, Melnikov, V, et al. Antenatal use of bosentan and/or sildenafil attenuates pulmonary features in rats with congenital diaphragmatic hernia. World J Pediatr. 2014; 10: 354–9.Google Scholar
Burgos, CM, Pearson, EG, Davey, M, Riley, J, Jia, H, Laje, P, et al. Improved pulmonary function in the nitrofen model of congenital diaphragmatic hernia following prenatal maternal dexamethasone and/or sildenafil. Pediatr Res. 2016; 80: 577–85.Google Scholar
Mous, DS, Kool, HM, Buscop-van Kempen, MJ, Koning, AH, Dzyubachyk, O, Wijnen, RM, et al. Clinically relevant timing of antenatal sildenafil treatment reduces pulmonary vascular remodeling in congenital diaphragmatic hernia. Am J Physiol Lung Cell Mol Physiol. 2016; 311: L734–42.Google Scholar
Russo, FM, Toelen, J, Eastwood, MP, Jimenez, J, Miyague, AH, Vande Velde, G, et al. Transplacental sildenafil rescues lung abnormalities in the rabbit model of diaphragmatic hernia. Thorax. 2016; 71: 517–25.Google Scholar
Russo, FM, Toelen, J, Eastwood, MP, Jimenez, J, Miyague, AH, Vande Velde, G, et al. Transplacental sildenafil rescues lung abnormalities in the rabbit model of diaphragmatic hernia. Thorax. 2016; 71: 517–25.Google Scholar
Russo, et al. Am J Obstet Gynecol. 2018 (will be added in galley proof phase)Google Scholar
Russo, FM, Conings, S, Allegaert, K, van Mieghem, T, Toelen, J, van Calsteren, K, et al. Sildenafil crosses the placenta at therapeutic levels in a dually perfused human cotyledon model. Am J Obstet Gynecol. 2018; 219: 619. e1–619. e10.Google Scholar
Russo, FM, Benachi, A, Van Mieghem, T, De Hoon, J, Van Calsteren, K, Annaert, P, et al. Antenatal sildenafil administration to prevent pulmonary hypertension in congenital diaphragmatic hernia (SToP-PH): study protocol for a phase I/IIb placenta transfer and safety study. Trials. 2018; 19: 524.Google Scholar
Khoshgoo, N, Kholdebarin, R, Iwasiow, BM, Keijzer, R. MicroRNAs and lung development. Pediatr Pulmonol. 2013; 48: 317–23.Google Scholar
Pereira-Terra, P, Deprest, JA, Kholdebarin, R, Khoshgoo, N, DeKoninck, P, Munck, AA, et al. Unique tracheal fluid microRNA signature predicts response to FETO in patients with congenital diaphragmatic Hernia. Ann Surg. 2015; 262: 1130–40.Google Scholar
Khoshgoo, N, Kholdebarin, R, Pereira-Terra, P, Mahood, TH, Falk, L, Day, CA, et al. Prenatal microRNA miR-200b therapy improves nitrofen-induced pulmonary hypoplasia associated with congenital diaphragmatic hernia. Ann Surg. 2019; 269: 979–87.Google Scholar
Eastwood, MP, Deprest, J, Russo, FM, Wang, H, Mulhall, D, Iwasiow, B, et al. MicroRNA 200b is upregulated in the lungs of fetal rabbits with surgically induced diaphragmatic hernia. Prenat Diagn. 2018; 38: 645–53.Google Scholar
Jani, JC, Nicolaides, KH, Gratacós, E, Vandecruys, H, Deprest, JA, Group, FT. Fetal lung-to-head ratio in the prediction of survival in severe left-sided diaphragmatic hernia treated by fetal endoscopic tracheal occlusion (FETO). Am J Obstet Gynecol. 2006; 195: 1646–50.Google Scholar
Russo, FM, De Coppi, P, Allegaert, K, Toelen, J, van der Veeken, L, Attilakos, G, et al. Current and future antenatal management of isolated congenital diaphragmatic hernia. Semin Fetal Neonatal Med. 2017; 22: 383–90.Google Scholar
Bancalari, E, Claure, N. Definitions and diagnostic criteria for bronchopulmonary dysplasia. Semin Perinatol. 2006; 30: 164–70.Google Scholar

References

Kassim, AA, Sharma, D. Hematopoietic stem cell transplantation for sickle cell disease: the changing landscape. Hematol Oncol Stem Cell Ther. 2017; 10: 259–66.Google Scholar
Raje, N, Dinakar, C. Overview of immunodeficiency disorders. Immunol Allergy Clin North Am. 2015; 35: 599623.Google Scholar
Sagar, R, Walther-Jallow, L, David, AL, Götherström, C, Westgren, M. Fetal mesenchymal stromal cells: an opportunity for prenatal cellular therapy. Curr Stem Cell Rep. 2018; 4: 61–8.Google Scholar
Squillaro, T, Peluso, G, Galderisi, U. Clinical trials with mesenchymal stem cells: an update. Cell Transplant. 2016; 25: 829–48.Google Scholar
Nijagal, A, Flake, AW, MacKenzie, TC. In utero hematopoietic cell transplantation for the treatment of congenital anomalies. Clin Perinatol. 2012; 39: 301–10.Google Scholar
Tiblad, E, Westgren, M. Fetal stem-cell transplantation. Best Pract Res Clin Obstet Gynaecol. 2008; 22 : 189201.Google Scholar
Touraine, JL, Raudrant, D, Royo, C, Rebaud, A, Roncarolo, MG, Souillet, G, et al. In-utero transplantation of stem cells in bare lymphocyte syndrome. Lancet. 1989; 1: 1382.Google Scholar
Vrecenak, JD, Flake, AW. In utero hematopoietic cell transplantation –recent progress and the potential for clinical application. Cytotherapy. 2013; 15: 525–35.Google Scholar
Flake, AW, Zanjani, ED. In utero hematopoietic stem cell transplantation: ontogenic opportunities and biologic barriers. Blood. 1999; 94: 2179–91.Google Scholar
Westgren, M, Ringdén, O, Eik-Nes, S, Ek, S, Anvret, M, Brubakk, AM, et al. Lack of evidence of permanent engraftment after in utero fetal stem cell transplantation in congenital hemoglobinopathies. Transplantation. 1996; 61: 1176–9.Google Scholar
Loewendorf, AI, Csete, M, Flake, A. Immunological considerations in in utero hematopoetic stem cell transplantation (IUHCT). Front Pharmacol. 2014; 5: 282.Google Scholar
Perez, B, Vilageliu, L, Grinberg, D, Desviat, LR. Antisense mediated splicing modulation for inherited metabolic diseases: challenges for delivery. Nucleic Acid Ther. 2014; 24: 4856.Google Scholar
Baris, HN, Cohen, IJ, Mistry, PK. Gaucher disease: the metabolic defect, pathophysiology, phenotypes and natural history. Pediatr Endocrinol Rev. 2014; 12 (Suppl. 1): 7281.Google Scholar
Charrow, J, Scott, CR. Long-term treatment outcomes in Gaucher disease. Am J Hematol. 2015; 90 (Suppl. 1): S19–24.Google Scholar
Orvisky, E, Sidransky, E, McKinney, CE, Lamarca, ME, Samimi, R, Krasnewich, D, et al. Glucosylsphingosine accumulation in mice and patients with type 2 Gaucher disease begins early in gestation. Pediatr Res. 2000; 48: 233–7.Google Scholar
Müller, I, Kustermann-Kuhn, B, Holzwarth, C, Isensee, G, Vaegler, M, Harzer, K, et al.. In vitro analysis of multipotent mesenchymal stromal cells as potential cellular therapeutics in neurometabolic diseases in pediatric patients.. Exp Hematol. 2006; 34: 1413–19.Google Scholar
Weiss, K, Gonzalez, A, Lopez, G, Pedoeim, L, Groden, C, Sidransky, E. The clinical management of Type 2 Gaucher disease. Mol Genet Metab. 2015; 114: 110–22.Google Scholar
Meregalli, M, Farini, A, Belicchi, M, Parolini, D, Cassinelli, L, Razini, P, et al. Perspectives of stem cell therapy in Duchenne muscular dystrophy. FEBS J. 2013; 280: 4251–62.Google Scholar
Flanigan, KM. Duchenne and Becker muscular dystrophies. Neurol Clin. 2014; 32: 671–88, viii.Google Scholar
Mendell, JR, Rodino-Klapac, LR, Sahenk, Z, Roush, K, Bird, L, Lowes, LP, et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol. 2013; 74: 637–47.Google Scholar
Aartsma-Rus, A, Goemans, N. A Sequel to the eteplirsen saga: eteplirsen is approved in the United States but was not approved in Europe. Nucleic Acid Ther. 2019; 29: 1315.Google Scholar
Falzarano, MS, Scotton, C, Passarelli, C, Ferlini, A. Duchenne muscular dystrophy: from diagnosis to Therapy. Molecules. 2015; 20: 18168–84.Google Scholar
Cordova, G, Negroni, E, Cabello-Verrugio, C, Mouly, V, Trollet, C. Combined therapies for duchenne muscular dystrophy to optimize treatment efficacy. Front Genet. 2018; 9: 114.Google Scholar
Chan, J, Waddington, SN, O’Donoghue, K, Kurata, H, Guillot, PV, Götherström, C, et al. Widespread distribution and muscle differentiation of human fetal mesenchymal stem cells after intrauterine transplantation in dystrophic mdx mouse. Stem Cells. 2007; 25: 875–84.Google Scholar
de Carvalho, SC, Hindi, SM, Kumar, A, Marques, MJ. Effects of omega-3 on matrix metalloproteinase-9, myoblast transplantation and satellite cell activation in dystrophin-deficient muscle fibers. Cell Tissue Res. 2017; 369: 591602.Google Scholar
Aminzadeh, MA, Rogers, RG, Fournier, M, Tobin, RE, Guan, X, Childers, MK, et al. Exosome-mediated benefits of cell therapy in mouse and human models of duchenne muscular dystrophy. Stem Cell Reports. 2018; 10: 942–55.Google Scholar
Silva, MC, Magalhaes, TA, Meira, ZM, Rassi, CH, Andrade, AC, Gutierrez, PS, et al. Myocardial fibrosis progression in duchenne and becker muscular dystrophy: a randomized clinical trial. JAMA Cardiol. 2017; 2: 190–9.Google Scholar
Marini, JC, Forlino, A, Bachinger, HP, Bishop, NJ, Byers, PH, Paepe, A, et al. Osteogenesis imperfecta. Nat Rev Dis Primers. 2017; 3: 17052.Google Scholar
Folkestad, L, Hald, JD, Canudas-Romo, V, Gram, J, Hermann, AP, Langdahl, B, et al. Mortality and causes of death in patients with osteogenesis imperfecta: a register-based nationwide cohort study. J Bone Miner Res. 2016; 31: 2159–66.Google Scholar
Hald, JD, Evangelou, E, Langdahl, BL, Ralston, SH. Bisphosphonates for the prevention of fractures in osteogenesis imperfecta: meta-analysis of placebo-controlled trials. J Bone Miner Res. 2015; 30: 929–33.Google Scholar
Dwan, K, Phillipi, CA, Steiner, RD, Basel, D. Bisphosphonate therapy for osteogenesis imperfecta. Cochrane Database Syst Rev. 2014; 7: CD005088.Google Scholar
Chan, JK, Götherström, C. Prenatal transplantation of mesenchymal stem cells to treat osteogenesis imperfecta. Front Pharmacol. 2014; 5: 223.Google Scholar
Horwitz, EM, Prockop, DJ, Gordon, PL, Koo, WW, Fitzpatrick, LA, Neel, MD, et al. Clinical responses to bone marrow transplantation in children with severe osteogenesis imperfecta. Blood. 2001; 97: 1227–31.Google Scholar
Horwitz, EM, Gordon, PL, Koo, WK, Marx, JC, Neel, MD, McNall, RY, et al. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: implications for cell therapy of bone. Proc Natl Acad Sci U S A. 2002; 99: 8932–7.Google Scholar
Le Blanc, K, Götherström, C, Ringden, O, Hassan, M, McMahon, R, Horwitz, E, et al. Fetal mesenchymal stem-cell engraftment in bone after in utero transplantation in a patient with severe osteogenesis imperfecta. Transplantation. 2005; 79: 1607–14.Google Scholar
Götherström, C, Westgren, M, Shaw, SW, Astrom, E, Biswas, A, Byers, PH, et al. Pre- and postnatal transplantation of fetal mesenchymal stem cells in osteogenesis imperfecta: a two-center experience. Stem Cells Transl Med. 2014; 3: 255–64.Google Scholar
Götherström, C, Hermerén, G, Johansson, M, Sahlin, N-E, Westgren, M. Stem cells and fetal therapy: is it a reality? Obstet Gynecol Reprod Med. 2017; 27: 166–7.Google Scholar
MacKenzie, TC, David, AL, Flake, AW, Almeida-Porada, G. Consensus statement from the first international conference for in utero stem cell transplantation and gene therapy. Front Pharmacol. 2015; 6: 15.Google Scholar

References

Deprest, JA, Lerut, TE, Vandenberghe, K. Operative fetoscopy: new perspective in fetal therapy? Prenat Diagn. 1997; 17: 1247–60.Google Scholar
Gratacós, E, Sanin-Blair, J, Lewi, L, et al. A histological study of fetoscopic membrane defects to document membrane healing. Placenta. 2006; 27: 452–6.Google Scholar
Crowley, AE, Grivell, RM, Dodd, JM. Sealing procedures for preterm prelabour rupture of membranes. Cochrane Database Syst Rev. 2016; 7: CD010218.Google Scholar
Theoret, C. Physiology of wound healing. In Theoret, C, Schumacher, J, eds., Equine Wound Management. Ames: John Wiley & Sons, Inc. 2017, 113.Google Scholar
Devlieger, D, Millar, LK, Bryant-Greenwood, G, Lewi, L, Deprest, JA. Fetal membrane healing after spontaneous and iatrogenic membrane rupture: a review of current evidence. Am J Obstet Gynecol. 2006; 195: 1512–20.Google Scholar
Sopher, D. The response of rat fetal membranes to injury. Ann R Coll Surg Engl. 1972; 51: 240–9.Google Scholar
Devlieger, R, Riley, SC, Verbist, L, Leask, R, Pijnenborg, R, Deprest, JA. Matrix metalloproteinases-2 and -9 and their endogenous tissue inhibitors in tissue remodeling after sealing of the fetal membranes in a sheep model of fetoscopic surgery. J Soc Gynecol Investig. 2002; 9: 137–45.Google Scholar
Papanna, R, Mann, LK, Tseng, SC, et al. Cryopreserved human amniotic membrane and a bioinspired underwater adhesive to seal and promote healing of iatrogenic fetal membrane defect sites. Placenta. 2015; 36: 888–94.Google Scholar
Mogami, H, Hari Kishore, A, Akgul, Y, Word, RA. Healing of preterm ruptured fetal membranes. Sci Rep. 2017; 7: 13139.Google Scholar
Papanna, R, Mann, LK, Moise, KJ, et al. Histologic changes of the fetal membranes after fetoscopic laser surgery for twin-twin transfusion syndrome. Pediatr Res. 2015; 78: 247–55.Google Scholar
Carvalho, S, Moron, AF, Menon, R, et al. Histological evidence of reparative activity in chorioamniotic membrane following open fetal surgery for myelomeningocele. Exp Ther Med. 2017; 14: 3732–6.Google Scholar
Engels, AC, Bauters, D, Rynkevic, R, et al. Thrombin generation by fetoscopic trauma to the fetal membranes: an in vivo and in vitro Study. Fetal Diagn Ther. 2016; 39: 261–8.Google Scholar
Bilic, G, Ochsenbein-Kolble, N, Hall, H, Huch, R, Zimmermann, R. In vitro lesion repair by human amnion epithelial and mesenchymal cells. Am J Obstet Gynecol. 2004; 190: 8792.Google Scholar
Ochsenbein-Kölble, N, Bilic, G, Hall, H, Huch, R, Zimmermann, R. Inducing proliferation of human amnion epithelial and mesenchymal cells for prospective engineering of membrane repair. J Perinat Med. 2003; 31: 287.Google Scholar
Lim, R, Chan, ST, Tan, JL, Mockler, JC, Murphy, SV, Wallace, EM. Preterm human amnion epithelial cells have limited reparative potential. Placenta. 2013; 34: 486–92.Google Scholar
Barrett, DW, David, AL, Thrasivoulou, C, Mata, A, Becker, DL, Engels, AC, et al. Connexin 43 is overexpressed in human fetal membrane defects after fetoscopic surgery. Prenat Diagn. 2014; 36: 942–52.Google Scholar
Barrett, DW, Keethes, A, Thrasivoulou, C, et al. Trauma induces overexpression of Cx43 in human fetal membrane defects. Prenat Diagn. 2017; 37: 899906.Google Scholar
Genz, HG. Treatment of premature rupture of the fetal membranes by means of fibrin adhesion. Med Welt. 1979; 30: 1557–9.Google Scholar
Baumgarten, K, Moser, S. The technique of fibrin adhesion for premature rupture of the membranes during pregnancy. J Perinat Med. 1986; 14: 43–9.Google Scholar
Quintero, RA, Morales, WJ, Allen, M, Bornick, PW, Arroyo, J, LeParc, G. Treatment of iatrogenic previable premature rupture of membranes with intra-amniotic injection of platelets and cryoprecipitate (amniopatch): preliminary experience. Am J Obstet Gynecol. 1999; 181: 744–9.Google Scholar
Reddy, UM, Shah, SS, Nemiroff, RL, et al. In vitro sealing of punctured fetal membranes: potential treatment for midtrimester premature rupture of membranes. Am J Obstet Gynecol. 2001; 185: 1090–3.Google Scholar
Harmanli, OH, Wapner, RJ, Lontz, JF. Efficacy of fibrin glue for in vitro sealing of human chorioamniotic membranes. J Reprod Med. 1998; 43: 986–90.Google Scholar
Young, BK, Roman, AS, MacKenzie, AP, et al. The closure of iatrogenic membrane defects after amniocentesis and endoscopic intrauterine procedures. Fetal Diagn Ther. 2004; 19: 296300.Google Scholar
Sciscione, AC, Manley, JS, Pollock, M, et al. Intracervical fibrin sealants: a potential treatment for early preterm premature rupture of the membranes. Am J Obstet Gynecol. 2001; 184: 368–73.Google Scholar
Quintero, RA. New horizons in the treatment of preterm premature rupture of membranes. Clin Perinatol. 2001; 28: 861–75.Google Scholar
Chmait, RH, Kontopoulos, EV, Chon, AH, Korst, LM, Llanes, A, Quintero, RA. Amniopatch treatment of iatrogenic preterm premature rupture of membranes (iPPROM) after fetoscopic laser surgery for twin-twin transfusion syndrome. J Matern Fetal Neonatal Med. 2017; 30: 1349–54.Google Scholar
Sung, JH, Kuk, JY, Cha, HH, et al. Amniopatch treatment for preterm premature rupture of membranes before 23 weeks’ gestation and factors associated with its success. Taiwan J Obstet Gynecol. 2017; 56: 599605.Google Scholar
Luks, FI, Deprest, JA, Peers, KH, Steegers, EA, van Der Wildt, B. Gelatin sponge plug to seal fetoscopy port sites: technique in ovine and primate models. Am J Obstet Gynecol. 1999; 181: 995–6.Google Scholar
Quintero, RA, Morales, WJ, Bornick, PW, Allen, M, Garabelis, N. Surgical treatment of spontaneous rupture of membranes: the amniograft – first experience. Am J Obstet Gynecol. 2002; 186: 155–7.Google Scholar
O’Brien, JM, Barton, JR, Milligan, DA. An aggressive interventional protocol for early midtrimester premature rupture of the membranes using gelatin sponge for cervical plugging. Am J Obstet Gynecol. 2002; 187: 1143–6.Google Scholar
O’Brien, JM, Mercer, BM, Barton, JR, Milligan, DA. An in vitro model and case report that used gelatin sponge to restore amniotic fluid volume after spontaneous premature rupture of the membranes. Am J Obstet Gynecol. 2001; 185: 1094–7.Google Scholar
Deprest, JA, Papadopulos, NA, Decaluw, H, Yamamoto, H, Lerut, TE, Gratacós, E. Closure techniques for fetoscopic access sites in the rabbit at mid-gestation. Hum Reprod. 1999; 14: 1730–4.Google Scholar
Liekens, D, Lewi, L, Jani, J, et al. Enrichment of collagen plugs with platelets and amniotic fluid cells increases cell proliferation in sealed iatrogenic membrane defects in the fetal rabbit model. Prenat Diagn. 2008; 28: 503–7.Google Scholar
Papadopulos, NA, Kyriakidis, DI, Schillinger, U, Totis, A, Henke, J, Kovacs, L, et al. Successful anatomic repair of fetoscopic access sites in the mid-gestational rabbit model using amnion cell engineering. In Vivo. 2010; 24: 745–50.Google Scholar
Engels, AC, Hoylaerts, MF, Endo, M, et al. In vitro sealing of iatrogenic fetal membrane defects by a collagen plug imbued with fibrinogen and plasma. Prenat Diagn. 2013: 33: 162–7.Google Scholar
Papanna, R, Mann, LK, Moise, KY, Johnson, A, Moise, KJ. Absorbable gelatin plug does not prevent iatrogenic preterm premature rupture of membranes after fetoscopic laser surgery for twin-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2013; 42: 456–60.Google Scholar
Papanna, R, Molina, S, Moise, KY, Moise, KJ Jr., Johnson, A. Chorioamnion plugging and the risk of preterm premature rupture of membranes after laser surgery in twin-twin transfusion syndrome. Ultrasound Obstet Gynecol. 2010; 35: 337–43.Google Scholar
Engels, AC, Van Calster, B, Richter, J, et al. Collagen plug sealing of iatrogenic fetal membrane defects after fetoscopic surgery for congenital diaphragmatic hernia. Ultrasound Obstet Gynecol. 2014; 43: 54–9.Google Scholar
Mallik, AS, Fichter, MA, Rieder, S, et al. Fetoscopic closure of punctured fetal membranes with acellular human amnion plugs in a rabbit model. Obstet Gynecol. 2007; 110: 1121–9.Google Scholar
Devlieger, R, Ardon, H, Verbist, L, Gratacós, E, Pijnenborg, R, Deprest, JA. Increased polymorphonuclear infiltration and iatrogenic amniotic band after closure of fetoscopic access sites with a bioactive membrane in the rabbit at midgestation. Am J Obstet Gynecol. 2003; 188: 844–8.Google Scholar
Ochsenbein-Kolble, N, Jani, J, Lewi, L, et al. Enhancing sealing of fetal membrane defects using tissue engineered native amniotic scaffolds in the rabbit model. Am J Obstet Gynecol. 2007; 196: 263. e1–7.Google Scholar
Engels, AC, Joyeux, L, Van der Merwe, J, Jimenez, J, Prapanus, S, Barrett, DW, et al. Tissuepatch is biocompatible and seals iatrogenic membrane defects in a rabbit model. Prenat Diagn. 2018; 38: 99105.Google Scholar
Kivelio, A, Dekoninck, P, Perrini, M, et al. Mussel mimetic tissue adhesive for fetal membrane repair: initial in vivo investigation in rabbits. Eur J Obstet Gynecol Reprod Med. 2013; 171: 240–5.Google Scholar
Haller, CM, Buerzle, W, Brubaker, CE, et al. Mussel-mimetic tissue adhesive for fetal membrane repair: a standardized ex vivo evaluation using elastomeric membranes. Prenat Diagn. 2011; 31: 654–60.Google Scholar
Mann, LK, Papanna, R, Moise, KJ Jr., et al. Fetal membrane patch and biomimetic adhesive coacervates as a sealant for fetoscopic defects. Acta Biomater. 2012; 8: 2160–5.Google Scholar
Bilic, G, Brubaker, C, Messersmith, PB, et al. Injectable candidate sealants for fetal membrane repair: bonding and toxicity in vitro. Am J Obstet Gynecol. 2010; 202: 85. e1–9.Google Scholar
Parolini, O, Alviano, F, Bagnara, GP, et al. Concise review: isolation and characterization of cells from human term placenta: outcome of the First International Workshop on Placenta Derived Stem Cells. Stem Cells. 2008; 26: 300–11.Google Scholar
Miki, T, Lehmann, T, Cai, H, Stolz, DB, Strom, SC. Stem cell characteristics of amniotic epithelial cells. Stem Cells. 2005; 23: 1549–59.Google Scholar
Mi, S, David, AL, Chowdhury, B, Jones, RR, Hamley, IW, Squires, AM, Connon, CJ. Tissue engineering a fetal membrane.Tissue Eng Part A. 2012; 18: 373–81.Google Scholar
Papanna, R, Fletcher, S, Moise, KJ Jr., et al. Cryopreserved human umbilical cord patch for in-utero spina bifida repair. Ultrasound Obstet Gynecol. 2016; 47: 168–76.Google Scholar
Roman, S, Bullock, AJ, Anumba, DO, MacNeil, S. Development of an implantable synthetic membrane for the treatment of preterm premature rupture of fetal membranes. J Biomater Appl. 2016; 30: 9951003.Google Scholar
Perrini, M, Mauri, A, Ehret, AE, et al. Mechanical and microstructural investigation of the cyclic behavior of human amnion. J Biomech Eng. 2015; 137: 061010.Google Scholar
Chowdhury, B, David, AL, Thrasivoulou, C, Becker, DL, Bader, DL, Chowdhury, TT. Tensile strain increased COX-2 expression and PGE2 release leading to weakening of the human amniotic membrane. Placenta. 2014; 35: 1057–64.Google Scholar
Sooranna, SR, Lee, Y, Kim, LU, Mohan, AR, Bennett, PR, Johnson, MR. Mechanical stretch activates type 2 cyclooxygenase via activator protein-1 transcription factor in human myometrial cells. Mol Hum Reprod. 2004; 10: 109–13.Google Scholar
Adams Waldorf, KM. Uterine overdistention induces preterm labor mediated by inflammation: observations in pregnant women and nonhuman primates. Am J Obstet Gynecol. 2015; 213: 830. e1–830. e19.Google Scholar
Mauri, A, Perrini, M, Mateos, JM, et al. Second harmonic generation microscopy of fetal membranes under deformation: normal and altered morphology. Placenta. 2013; 34: 1020–6.Google Scholar
Kumar, D, Moore, RM, Nash, A, et al. Decidual GM-CSF is a critical common intermediate necessary for thrombin and TNF induced in-vitro fetal membrane weakening. Placenta. 2014; 35: 1049–56.Google Scholar
Petersen, SG, Gibbons, KS. The impact of entry technique and access diameter on prelabour rupture of membranes following primary fetoscopic laser treatment for twin-twin transfusion syndrome. Fetal Diagn Ther. 2016; 40: 100–9.Google Scholar
Beck, V, Lewi, P, Gucciardo, L, Devlieger, R. Preterm prelabor rupture of membranes and fetal survival after minimally invasive fetal surgery: a systematic review of the literature. Fetal Diagn Ther. 2012; 31: 19.Google Scholar

References

Amit, M, Shariki, C, Margulets, V, Itskovitz-Eldor, J. Feeder layer and serum-free culture of human embryonic stem cells. Biol Reprod. 2004; 70: 837–45.Google Scholar
Fong, H, Hohenstein, KA, Donovan, PJ. Regulation of self-renewal and pluripotency by Sox2 in human embryonic stem cells. Stem Cells. 2008; 26: 1931–8.Google Scholar
Klimanskaya, I, Chung, Y, Becker, S, et al. Human embryonic stem cell lines derived from single blastomeres. Nature. 2006; 444: 481–5.Google Scholar
Chung, Y, Klimanskaya, I, Becker, S, Marh, J, Lu, SJ, Johnson, J, et al. Embryonic and extraembryonic stem cell lines derived from single mouse blastomeres. Nature. 2006; 439: 216–19.Google Scholar
Deb, KD, Sarda, K. Human embryonic stem cells: preclinical perspectives. J Transl Med. 2008; 6: 7.Google Scholar
Briggs, R, King, TJ. Transplantation of living nuclei from blastula cells into enucleated frogs’ eggs. Proc Natl Acad Sci USA. 1952; 38: 455–63.Google Scholar
Campbell, KH, McWhir, J, Ritchie, WA, Wilmut, I. Sheep cloned by nuclear transfer from a cultured cell line. Nature. 1996; 380: 64–6.Google Scholar
Takahashi, K, Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006; 126: 663–76.Google Scholar
Slamecka, J, Salimova, L, McClellan, S, van Kelle, M, Kehl, D, Laurini, J, et al. Non-integrating episomal plasmid-based reprogramming of human amniotic fluid stem cells into induced pluripotent stem cells in chemically defined conditions. Cell Cycle. 2016; 15: 234–49.Google Scholar
Velasquez-Mao, AJ, Tsao, CJM, Monroe, MN, Legras, X, Bissig-Choisat, B, Bissig, K-D, et al. Differentiation of spontaneously contracting cardiomyocytes from non-virally reprogrammed human amniotic fluid stem cells. PLoS One. 2017; 12: e0177824.Google Scholar
Chang, C-Y, Ting, H-C, Su, H-L, Jeng, J-R. Combining induced pluripotent stem cells and genome editing technologies for clinical applications. Cell Transplant. 2018; 27: 379–92.Google Scholar
De Coppi, P, Pozzobon, M, Piccoli, M, Gazzola, M, Boldrin, L, Slanzi, E, et al. Isolation of mesenchymal stem cells from human vermiform appendix. J Surg Res. 2006; 135: 8591.Google Scholar
Loukogeorgakis, SP, De Coppi, P. Stem cells from amniotic fluid – potential for regenerative medicine. Best Pract Res Clin Obstet Gynaecol. 2016; 31: 4557.Google Scholar
De Coppi, P. Tissue engineering and stem cell research. In Puri, P, ed., Newborn Surgery, 4th edn. Boca Raton: CRC Press, 2017, pp. 301–14.Google Scholar
Caves, JM, Kumar, VA, Martinez, AW, Kim, J, Ripberger, CM, Haller, CA, et al. The use of microfiber composites of elastin-like protein matrix reinforced with synthetic collagen in the design of vascular grafts. Biomaterials. 2010; 31: 7175–82.Google Scholar
Gasior, AC, St. Peter, SD. A review of patch options in the repair of congenital diaphragm defects. Pediatr Surg Int. 2012; 28: 327–33.Google Scholar
Mayer, S, Decaluwe, H, Ruol, M, Manodoro, S, Kramer, M, Till, H, et al. Diaphragm repair with a novel cross-linked collagen biomaterial in a growing rabbit model. PLoS One. 2015; 10: e0132021.Google Scholar
Huang, AH, Niklason, LE. Engineering Biological-Based Vascular Grafts Using a Pulsatile Bioreactor. J Vis Exp. 2011; 52: 2646.Google Scholar
Lu, H, Feng, Z, Gu, Z, Liu, C. Growth of outgrowth endothelial cells on aligned PLLA nanofibrous scaffolds. J Mater Sci Mater Med. 2009; 20: 1937–44.Google Scholar
Tillman, BW, Yazdani, SK, Lee, SJ, Geary, RL, Atala, A, Yoo, JJ. The in vivo stability of electrospun polycaprolactone-collagen scaffolds in vascular reconstruction. Biomaterials. 2009; 30: 583–8.Google Scholar
Mathews, A, Colombus, S, Krishnan, VK, Krishnan, LK. Vascular tissue construction on poly(epsilon-caprolactone) scaffolds by dynamic endothelial cell seeding: effect of pore size. J Tissue Eng Regen Med. 2012; 6: 451–61.Google Scholar
Zhao, J, Qiu, H, Chen, D, Zhang, W, Zhang, D, Li, M. Development of nanofibrous scaffolds for vascular tissue engineering. Int J Biol Macromol. 2013; 56: 106–13.Google Scholar
Huang, Y-C, Kuo, Y, Huang, Y, Chen, C, Ho, D, Shi, C-S. The effects of adipose-derived stem cells in a rat model of tobacco-associated erectile dysfunction. PLoS One. 2016; 11: e0156725.Google Scholar
Satake, R, Komura, M, Komura, H, Kodaka, T, Terawaki, K, Ikebukuro, K, et al. Patch tracheoplasty in body tissue engineering using collagenous connective tissue membranes (biosheets). J Pediatr Surg. 2016; 51: 244–8.Google Scholar
Suzuki, K, Komura, M, Terawaki, K, Kodaka, T, Gohara, T, Komura, H, et al. Engineering and repair of diaphragm using biosheet (a collagenous connective tissue membrane) in rabbits. J Pediatr Surg. 2018; 53: 330–4.Google Scholar
Mitchell, IC, Garcia, NM, Barber, R, Ahmad, N, Hicks, BA, Fischer, AC. Permacol: a potential biologic patch alternative in congenital diaphragmatic hernia repair. J Pediatr Surg. 2008; 43: 2161–4.Google Scholar
Balayssac, D, Poinas, AC, Pereira, B, Pezet, D. Use of permacol in parietal and general surgery: a bibliographic review. Surg Innov. 2013; 20: 176–82.Google Scholar
Cheng, AW, Abbas, MA, Tejirian, T. Outcome of abdominal wall hernia repair with biologic mesh: PermacolTM versus StratticeTM. Am Surg. 2014; 80: 9991002.Google Scholar
Gilpin, A, Yang, Y. Decellularization strategies for regenerative medicine: from processing techniques to applications. Biomed Res Int. 2017; 2017: 9831534.Google Scholar
Wainwright, DJ. Use of an acellular allograft dermal matrix (AlloDerm) in the management of full-thickness burns. Burns. 1995; 21: 243–8.Google Scholar
Elliott, MJ, De Coppi, P, Speggiorin, S, Roebuck, D, Butler, CR, Samuel, E, et al. Stem-cell-based, tissue engineered tracheal replacement in a child: a 2-year follow-up study. Lancet. 2012; 380: 9941000.Google Scholar
Flake, AW, Roncarolo, M-G, Puck, JM, Almeida-Porada, G, Evans, MI, Johnson, MP, et al. Treatment of X-linked severe combined immunodeficiency by in utero transplantation of paternal bone marrow. N Engl J Med. 1996; 335: 1806–10.Google Scholar
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: 9931004.Google Scholar
Götherström, C, Westgren, M, Shaw, SWS, Åström, E, Biswas, A, Byers, PH, et al. Pre- and postnatal transplantation of fetal mesenchymal stem cells in osteogenesis imperfecta: a two-center experience. Stem Cells Transl Med. 2014; 3: 255–64.Google Scholar
Deprest, J, Brady, P, Nicolaides, K, Benachi, A, Berg, C, Vermeesch, J, et al. Prenatal management of the fetus with isolated congenital diaphragmatic hernia in the era of the TOTAL trial. Semin Fetal Neonatal Med. 2014; 19: 338–48.Google Scholar
Haynes, BF. Human thymic epithelium and T cell development: current issues and future directions. Thymus. 1990; 16: 143–57.Google Scholar
Pawlowski, TJ, Staerz, UD. Thymic education – T cells do it for themselves. Trends Immunol. 1994; 15: 205–9.Google Scholar
Liechty, KW, MacKenzie, TC, Shaaban, AF, Radu, A, Moseley, AM, Deans, R, et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med. 2000; 6: 1282–6.Google Scholar
Maselli, KM, Badillo, A. Advances in fetal surgery. Ann Transl Med. 2016; 4: 394.Google Scholar
Kitagawa, H, Pringle, KC. Fetal surgery: a critical review. Pediatr Surg Int. 2017; 33: 421–33.Google Scholar
Partridge, EA, Davey, MG, Hornick, MA, McGovern, PE, Mejaddam, AY, Vrecenak, JD, et al. An extra-uterine system to physiologically support the extreme premature lamb. Nat Commun. 2017; 8: 15112.Google Scholar
Larson, BJ, Longaker, MT, Lorenz, HP. Scarless fetal wound healing: a basic science review. Plast Reconstr Surg. 2010; 126: 1172–80.Google Scholar
Yagi, LH, Watanuki, LM, Isaac, C, Gemperli, R, Nakamura, YM, Ladeira, PRS. Human fetal wound healing: a review of molecular and cellular aspects. Eur J Plastic Surg. 2016; 39: 239–46.Google Scholar
Hosper, NA, Eggink, AJ, Roelofs, LAJ, Wijnen, RMH, van Luyn, MJA, Bank, RA, et al. Intra-uterine tissue engineering of full-thickness skin defects in a fetal sheep model. Biomaterials. 2010; 31: 3910–19.Google Scholar
Xia, W, Noimark, S, Ourselin, S, West, SJ, Finlay, MC, David, AL, et al. Ultrasonic needle tracking with a fibre-optic ultrasound transmitter for guidance of minimally invasive fetal surgery. Med Image Comput Comput Assist Interv. 2017; 10434: 637–45.Google Scholar
Peter, L, Tella-Amo, M, Shakir, DI, Attilakos, G, Wimalasundera, R, Deprest, J, et al. Retrieval and registration of long-range overlapping frames for scalable mosaicking of in vivo fetoscopy. Int J Comput Assist Radiol Surg. 2018; 13: 713–20.Google Scholar
Watanabe, M, Kim, AG, Flake, AW. Tissue engineering strategies for fetal myelomeningocele repair in animal models. Fetal Diagn Ther. 2015; 37: 197205.Google Scholar
Fauza, DO, Jennings, RW, Teng, YD, Snyder, EY. Neural stem cell delivery to the spinal cord in an ovine model of fetal surgery for spina bifida. Surgery. 2008; 144: 367–73.Google Scholar
Peiro, JL, Fontecha, CG, Ruano, R, Esteves, M, Fonseca, C, Marotta, M, et al. Single-Access Fetal Endoscopy (SAFE) for myelomeningocele in sheep model I: Amniotic carbon dioxide gas approach. Surg Endosc Other Interv Tech. 2013; 27: 3835–40.Google Scholar
Brown, EG, Saadai, P, Pivetti, CD, Beattie, MS, Bresnahan, JC, Wang, A, et al. In utero repair of myelomeningocele with autologous amniotic membrane in the fetal lamb model. J Pediatr Surg. 2014; 49: 133–7; discussion 137–8.Google Scholar
Chen, YJ, Chung, K, Pivetti, C, Lankford, L, Kabagambe, SK, Vanover, M, et al. Fetal surgical repair with placenta-derived mesenchymal stromal cell engineered patch in a rodent model of myelomeningocele. J Pediatr Surg. 2018; 53: 183–88.Google Scholar
Roelofs, LAJ, Eggink, AJ, Hulsbergen-van de Kaa, CA, van den Berg, PP, van Kuppevelt, TH, van Moerkerk, HTB, et al. Fetal abdominal wall repair with a collagen biomatrix in an experimental sheep model for gastroschisis. Tissue Eng Part A. 2008; 14: 2033–40.Google Scholar
Roelofs, LAJ, Geutjes, PJ, Van De Kaa, CAH, Eggink, AJ, Van Kuppevelt, TH, Daamen, WF, et al. Prenatal coverage of experimental gastroschisis with a collagen scaffold to protect the bowel. J Pediatr Surg. 2013; 48: 516–24.Google Scholar
Coughlin, MA, Werner, NL, Gajarski, R, Gadepalli, S, Hirschl, R, Barks, J, et al. Prenatally diagnosed severe CDH: mortality and morbidity remain high. J Pediatr Surg. 2016; 51: 1091–5.Google Scholar
Deprest, J, Gucciardo, L, Eastwood, P, Zia, S, Jimenez, J, Russo, F, et al. Medical and regenerative solutions for congenital diaphragmatic hernia: A perinatal perspective. Eur J Pediatr Surg. 2014; 24: 270–7.Google Scholar
Fuchs, JR, Kaviani, A, Oh, JT, LaVan, D, Udagawa, T, Jennings, RW, et al. Diaphragmatic reconstruction with autologous tendon engineered from mesenchymal amniocytes. J Pediatr Surg. 2004; 39: 834–8.Google Scholar
Turner, CG, Klein, JD, Steigman, SA, Armant, M, Nicksa, GA, Zurakowski, D, et al. Preclinical regulatory validation of an engineered diaphragmatic tendon made with amniotic mesenchymal stem cells. J Pediatr Surg. 2011; 46: 5761.Google Scholar
Pederiva, F, Ghionzoli, M, Pierro, A, De Coppi, P, Tovar, JA. Amniotic fluid stem cells rescue both in vitro and in vivo growth, innervation, and motility in nitrofen-exposed hypoplastic rat lungs through paracrine effects. Cell Transplant. 2013; 22: 1683–94.Google Scholar
Lim, R, Malhotra, A, Tan, J, Chan, ST, Lau, S, Zhu, D, et al. First-in-human administration of allogeneic amnion cells in premature infants with bronchopulmonary dysplasia: a safety study. Stem Cells Transl Med. 2018; 7: 628–35.Google Scholar
Zani, A, Pierro, A, Elvassore, N, De Coppi, P. Tissue engineering: an option for esophageal replacement? Semin Pediatr Surg. 2009; 18: 5762.Google Scholar
Lee, E, Milan, A, Urbani, L, De Coppi, P, Lowdell, MW. Decellularized material as scaffolds for tissue engineering studies in long gap esophageal atresia. Expert Opin Biol Ther. 2017; 17: 573–84.Google Scholar
Scottoni, F, Urbani, L, Camilli, C. D1.4 Oesophageal tissue engineering: preliminary evaluation of a 2 stage surgical approach in a mouse model. Arch Dis Child. 2017; 102 (A6).Google Scholar
Okuyama, H, Umeda, S, Takama, Y, Terasawa, T, Nakayama, Y. Patch esophagoplasty using an in-body-tissue-engineered collagenous connective tissue membrane. J Pediatr Surg. 2018; 53: 223–6.Google Scholar
Poghosyan, T, Sfeir, R, Michaud, L, Bruneval, P, Domet, T, Vanneaux, V, et al. Circumferential esophageal replacement using a tube-shaped tissue-engineered substitute: an experimental study in minipigs. Surgery. 2015; 158: 266–77.Google Scholar
Catry, J, Luong-Nguyen, M, Arakelian, L, Poghosyan, T, Bruneval, P, Domet, T, et al. Circumferential esophageal replacement by a tissue-engineered substitute using mesenchymal stem cells: an experimental study in mini pigs. Cell Transplant. 2017; 26: 1831–9.Google Scholar
Petsche Connell, J, Camci-Unal, G, Khademhosseini, A, Jacot, JG. Amniotic fluid-derived stem cells for cardiovascular tissue engineering applications. Tissue Eng Part B Rev. 2013; 19: 368–79.Google Scholar
Velasquez-Mao, AJ, Tsao, CJM, Monroe, MN, Legras, X, Bissig-Choisat, B, Bissig, KD, et al. Differentiation of spontaneously contracting cardiomyocytes from non-virally reprogrammed human amniotic fluid stem cells. PLoS One. 2017; 12: e0177824.Google Scholar
Nolan, HR, Gurria, J, Peiro, JL, Tabbah, S, Diaz-Primera, R, Polzin, W, et al. Congenital high airway obstruction syndrome (CHAOS): Natural history, prenatal management strategies, and outcomes at a single comprehensive fetal center. J Pediatr Surg. 2019; 54:11531158.Google Scholar
Lange, P, Fishman, JM, Elliott, MJ, De Coppi, P, Birchall, MA. What can regenerative medicine offer for infants with laryngotracheal agenesis? Otolaryngol Head Neck Surg. 2011; 145: 544–50.Google Scholar
Atala, A, Bauer, SB, Soker, S, Yoo, JJ, Retik, AB. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet. 2006; 367: 1241–6.Google Scholar
Corre, P, Merceron, C, Longis, J, Khonsari, RH, Pilet, P, Thi, TN, et al. Direct comparison of current cell-based and cell-free approaches towards the repair of craniofacial bone defects – a preclinical study. Acta Biomater. 2015; 26: 306–17.Google Scholar
Steigman, SA, Ahmed, A, Shanti, RM, Tuan, RS, Valim, C, Fauza, DO. Sternal repair with bone grafts engineered from amniotic mesenchymal stem cells. J Pediatr Surg. 2009; 44: 1120–6.Google Scholar
Klein, JD, Turner, CGB, Ahmed, A, Steigman, SA, Zurakowski, D, Fauza, DO. Chest wall repair with engineered fetal bone grafts: an efficacy analysis in an autologous leporine model. J Pediatr Surg. 2010; 45: 1354–60.Google Scholar
Turner, CG, Klein, JD, Gray, FL, Ahmed, A, Zurakowski, D, Fauza, DO. Craniofacial repair with fetal bone grafts engineered from amniotic mesenchymal stem cells. J Surg Res. 2012; 178: 785–90.Google Scholar
Ranzoni, AM, Corcelli, M, Hau, K-L, Kerns, JG, Vanleene, M, Shefelbine, S, et al. Counteracting bone fragility with human amniotic mesenchymal stem cells. Sci Rep. 2016; 6: 39656.Google Scholar
Flake, AW, Harrison, MR, Adzick, NS, Zanjani, ED. Transplantation of fetal hematopoietic stem cells in utero: the creation of hematopoietic chimeras. Science. 1986; 233: 776–8.Google Scholar
Liechty, KW, Mackenzie, TC, Shaaban, AF, Radu, A, Moseley, AMB, Deans, R, et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med. 2000; 6: 1282–6.Google Scholar

References

Waddington, SN, Nivsarkar, MS, Mistry, AR, Buckley, SMK, Kemball-Cook, G, Mosley, KL, et al. Permanent phenotypic correction of hemophilia B in immunocompetent mice by prenatal gene therapy. Blood. 2004; 104: 2714–21.Google Scholar
Tran, ND, Porada, CD, Almeida-Porada, G, Glimp, HA, French Anderson, W, Zanjani, ED. Induction of stable prenatal tolerance to β-galactosidase by in utero gene transfer into preimmune sheep fetuses. Blood. 2001; 97: 3417–23.Google Scholar
US National Institutes of Health Recombinant DNA Advisory Committee. Prenatal gene transfer: scientific, medical, and ethical issues: a report of the Recombinant DNA Advisory Committee. Hum Gene Ther. 2000; 11: 1211–29.Google Scholar
Sabatino, DE, MacKenzie, TC, Peranteau, W, Edmonson, S, Campagnoli, C, Liu, YL, et al. Persistent expression of hF.IX after tolerance induction by in utero or neonatal administration of AAV-1-F.IX in hemophilia B mice. Mol Ther. 2007; 15: 1677–85.Google Scholar
Porada, CD, Tran, N, Eglitis, M, Moen, RC, Troutman, L, Flake, AW, et al. In utero gene therapy: transfer and long-term expression of the bacterial neo(r) gene in sheep after direct injection of retroviral vectors into preimmune fetuses. Hum Gene Ther. 1998; 9: 1571–85.Google Scholar
Yang, EY, Cass, DL, Sylvester, KG, Wilson, JM, Adzick, NS. Fetal gene therapy: efficacy, toxicity, and immunologic effects of early gestation recombinant adenovirus. J Pediatr Surg. 1999; 34: 235–41.Google Scholar
David, A, Peebles, D, Miah, M, Themis, M, Nivsarkar, M, Tucker, N, et al. Ultrasound-guided delivery of viral vectors encoding the beta-galactosidase and human factor IX genes to early gestation fetal sheep in utero. Hum Gene Ther. 2002; 364: 353–64.Google Scholar
Nathwani, AC, Gray, JT, Ng, CYC, Zhou, J, Spence, Y, Waddington, SN, et al. Self-complementary adeno-associated virus vectors containing a novel liver-specific human factor IX expression cassette enable highly efficient transduction of murine and nonhuman primate liver. Blood. 2006; 107: 2653–61.Google Scholar
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: 419–26.Google Scholar
Mattar, C, Nathwani, A, Waddington, S, 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: 1950–60.Google Scholar
Mattar, CN, Gil-Farina, I, Rosales, C, Johana, N, Yi Wan Tan, Y, McIntosh, J, et al. In utero transfer of adeno-associated viral vectors produces long-term factor IX levels in a cynomolgus macaque model. Mol Ther. 2017; 25: 1843–53.Google Scholar
Mattar, CN, Wong, AMS, Hoefer, K, Alonso-Ferrero, ME, Buckley, SMK, Howe, SJ, et al. Systemic gene delivery following intravenous administration of AAV9 to fetal and neonatal mice and late-gestation nonhuman primates. FASEB J. 2015; 29: 3876–88.Google Scholar
Nathwani, AC, Tuddenham, EGD, Rangarajan, S, Rosales, C, McIntosh, J, Linch, DC, et al. Adenovirus-associated virus vector–mediated gene transfer in hemophilia B. N Engl J Med. 2011; 365: 2357–65.Google Scholar
George, LA, Sullivan, SK, Giermasz, A, Rasko, JEJ, Samelson-Jones, BJ, Ducore, J, et al. Hemophilia B gene therapy with a high-specific-activity factor IX variant. N Engl J Med. 2017; 377: 2215–27.Google Scholar
Rangarajan, S, Walsh, L, Lester, W, Perry, D, Madan, B, Laffan, M, et al. AAV5–Factor VIII gene transfer in severe hemophilia A. N Engl J Med. 2017; 377: 2519–30.Google Scholar
McVey, JH, Boswell, E, Mumford, AD, Kemball-Cook, G, Tuddenham, EG. Factor VII deficiency and the FVII mutation database. Hum Mutat. 2001; 17: 317.Google Scholar
Binny, C, McIntosh, J, Della Peruta, M, Kymalainen, H, Tuddenham, EGD, Buckley, SMK, et al. AAV-mediated gene transfer in the perinatal period results in expression of FVII at levels that protect against fatal spontaneous hemorrhage. Blood. 2012; 119: 957–66.Google Scholar
Modell, B, Darlison, M. Global epidemiology of haemoglobin disorders and derived service indicators. Bull World Health Organ. 2008; 86: 480–7.Google Scholar
Lucarelli, G, Isgrò, A, Sodani, P, Gaziev, J. Hematopoietic stem cell transplantation in thalassemia and sickle cell anemia. Cold Spring Harb Perspect Med. 2012; 2: a011825.Google Scholar
Pawliuk, R, Westerman, KA, Fabry, ME, Payen, E, Tighe, R, Bouhassira, EE, et al. Correction of sickle cell disease in transgenic mouse models by gene therapy. Science. 2001; 294: 2368–71.Google Scholar
Rivella, S, May, C, Chadburn, A, Rivière, I, Sadelain, M. A novel murine model of Cooley anemia and its rescue by lentiviral-mediated human β-globin gene transfer. Blood. 2003; 101: 2932–9.Google Scholar
Han, X-D, Lin, C, Chang, J, Sadelain, M, Kan, YW. Fetal gene therapy of alpha-thalassemia in a mouse model. Proc Natl Acad Sci U S A. 2007; 104: 907–11.Google Scholar
Cavazzana-Calvo, M, Payen, E, Negre, O, Wang, G, Hehir, K, Fusil, F, et al. Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia. Nature. 2010; 467: 318–22.Google Scholar
Thompson, AA, Walters, MC, Kwiatkowski, J, et al. Gene therapy in patients with transfusion-dependent β-thalassemia. N Engl J Med. 2018; 378: 1479–93.Google Scholar
Shaw, SWS, 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: 122–32.Google Scholar
Alton, EWFW, Armstrong, DK, Ashby, D, Bayfield, KJ, Bilton, D, Bloomfield, EV, et al. Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Respir Med. 2015; 3: 684–91.Google Scholar
Griesenbach, U, Inoue, M, Meng, C, Farley, R, Chan, M, Newman, NK, et al. Assessment of F/HN-pseudotyped lentivirus as a clinically relevant vector for lung gene therapy. Am J Respir Crit Care Med. 2012; 186: 846–56.Google Scholar
Alton, EWFW, Beekman, JM, Boyd, AC, Brand, J, Carlon, MS, Connolly, MM, et al. Preparation for a first-in-man lentivirus trial in patients with cystic fibrosis. Thorax. 2017; 72: 137–47.Google Scholar
Larson, JE, Morrow, SL, Happel, L, Sharp, JF, Cohen, JC. Reversal of cystic fibrosis phenotype in mice by gene therapy in utero. Lancet. 1997; 349: 619–20.Google Scholar
Buckley, SMK, Waddington, SN, Jezzard, S, Bergau, A, Themis, M, MacVinish, LJ, et al. Intra-amniotic delivery of CFTR-expressing adenovirus does not reverse cystic fibrosis phenotype in inbred CFTR-knockout mice. Mol Ther. 2008; 16: 819–24.Google Scholar
Davies, LA, Varathalingam, A, Painter, H, Lawton, AE, Sumner-Jones, SG, Nunez-Alonso, GA, et al. Adenovirus-mediated in utero expression of CFTR does not improve survival of CFTR knockout mice. Mol Ther. 2008; 16: 812–18.Google Scholar
Buckley, SMK, Howe, SJ, Sheard, V, Ward, NJ, Coutelle, C, Thrasher, AJ, et al. Lentiviral transduction of the murine lung provides efficient pseudotype and developmental stage-dependent cell-specific transgene expression. Gene Ther. 2008; 15: 1167–75.Google Scholar
Buckley, SMK, Waddington, SN, Jezzard, S, Lawrence, L, Schneider, H, Holder, M V, et al. Factors influencing adenovirus-mediated airway transduction in fetal mice. Mol Ther. 2005; 12: 484–92.Google Scholar
Moss, IR, Scarpelli, EM. Stimulatory effect of theophylline on regulation of fetal breathing movements. Pediatr Res. 1981; 15: 870–3.Google Scholar
Henriques-Coelho, T, Gonzaga, S, Endo, M, Zoltick, PW, Davey, M, Leite-Moreira, AF, et al. Targeted gene transfer to fetal rat lung interstitium by ultrasound-guided intrapulmonary injection. Mol Ther. 2007; 15: 340–7.Google Scholar
Toelen, J, Deroose, CM, Gijsbers, R, Reumers, V, Sbragia, LN, Vets, S, et al. Fetal gene transfer with lentiviral vectors: long-term in vivo follow-up evaluation in a rat model. Am J Obstet Gynecol. 2007; 196. 352. e1–6.Google Scholar
Tarantal, AF, Lee, CI, Ekert, JE, McDonald, R, Kohn, DB, Plopper, CG, et al. Lentiviral vector gene transfer into fetal rhesus monkeys (Macaca mulatta): lung-targeting approaches. Mol Ther. 2001; 4: 614–21.Google Scholar
Tarantal, AF, McDonald, RJ, Jimenez, DF, Lee, CCI, O’Shea, CE, Leapley, AC, et al. Intrapulmonary and intramyocardial gene transfer in rhesus monkeys (Macaca mulatta): safety and efficiency of HIV-1-derived lentiviral vectors for fetal gene delivery. Mol Ther. 2005; 12: 8798.Google Scholar
David, AL, Peebles, DM, Gregory, L, Themis, M, Cook, T, Coutelle, C, et al. Percutaneous ultrasound-guided injection of the trachea in fetal sheep: A novel technique to target the fetal airways. Fetal Diagn Ther. 2003; 18: 385–90.Google Scholar
Peebles, D, Gregory, LG, David, A, Themis, M, Waddington, SN, Knapton, HJ, et al. Widespread and efficient marker gene expression in the airway epithelia of fetal sheep after minimally invasive tracheal application of recombinant adenovirus in utero. Gene Ther. 2004; 11: 70–8.Google Scholar
Gregory, LG, Harbottle, RP, Lawrence, L, Knapton, HJ, Themis, M, Coutelle, C. Enhancement of adenovirus-mediated gene transfer to the airways by DEAE dextran and sodium caprate in vivo. Mol Ther. 2003; 7: 1926.Google Scholar
David, AL, Peebles, DM, Gregory, L, Waddington, SN, Themis, M, Weisz, B, et al. Clinically applicable procedure for gene delivery to fetal gut by ultrasound-guided gastric injection: toward prenatal prevention of early-onset intestinal diseases. Hum Gene Ther. 2006; 17 : 767–79.Google Scholar
Saada, J, Oudrhiri, N, Bonnard, A, de Lagausie, P, Aissaoui, A, Hauchecorne, M, et al. Combining keratinocyte growth factor transfection into the airways and tracheal occlusion in a fetal sheep model of congenital diaphragmatic hernia. J Gene Med. 2010; 12: 413–22.Google Scholar
Berges, BK, Yellayi, S, Karolewski, BA, Miselis, RR, Wolfe, JH, Fraser, NW. Widespread correction of lysosomal storage in the mucopolysaccharidosis type VII mouse brain with a herpes simplex virus type I vector expressing beta-glucuronidase. Mol Ther. 2006; 13: 859–69.Google Scholar
Ciron, C, Desmaris, N, Colle, MA, Raoul, S, Joussemet, B, Vérot, L, et al. Gene therapy of the brain in the dog model of Hurler’s syndrome. Ann Neurol. 2006; 60: 204–13.Google Scholar
Shen, JS, Meng, XL, Yokoo, T, Sakurai, K, Watabe, K, Ohashi, T, et al. Widespread and highly persistent gene transfer to the CNS by retrovirus vector in utero: implication for gene therapy to Krabbe disease. J Gene Med. 2005; 7: 540–51.Google Scholar
Karolewski, BA, Wolfe, JH. Genetic correction of the fetal brain increases the lifespan of mice with the severe multisystemic disease mucopolysaccharidosis type VII. Mol Ther. 2006; 14: 1424.Google Scholar
Tarantal, AF, Chu, F, O’Brien, WD, Hendrickx, AG. Sonographic heat generation in vivo in the gravid long-tailed macaque (Macaca fascicularis). J Ultrasound Med. 1993; 12: 285–95.Google Scholar
Massaro, G, Mattar, CNZ, Wong, AMS, Sirka, E, Buckley, SMK, Herbert, BR, et al. Fetal gene therapy for neurodegenerative disease of infants. Nat Med. 2018; 24: 1317–23.Google Scholar
Mendell, JR, Al-Zaidy, S, Shell, R, Arnold, WD, Rodino-Klapac, LR, Prior, TW, et al. Single-dose gene-replacement therapy for spinal muscular atrophy. N Engl J Med. 2017; 377: 1713–22.Google Scholar
Foust, KD, Nurre, E, Montgomery, CL, Hernandez, A, Chan, CM, Kaspar, BK. Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes. Nat Biotechnol. 2009; 27: 5965.Google Scholar
Duque, S, Joussemet, B, Riviere, C, Marais, T, Dubreil, L, Douar, A-M, et al. Intravenous administration of self-complementary AAV9 enables transgene delivery to adult motor neurons. Mol Ther. 2009; 17: 1187–96.Google Scholar
Manfredsson, FP, Rising, AC, Mandel, RJ. AAV9: A potential blood-brain barrier buster. Mol Ther. 2009; 17: 403–5.Google Scholar
Rahim, AA, Wong, AMS, Hoefer, K, Buckley, SMK, Mattar, CN, Cheng, SH, et al. Intravenous administration of AAV2/9 to the fetal and neonatal mouse leads to differential targeting of CNS cell types and extensive transduction of the nervous system. FASEB J. 2011; 25: 3505–18.Google Scholar
Le Guiner, C, Servais, L, Montus, M, Larcher, T, Fraysse, B, Moullec, S, et al. Long-term microdystrophin gene therapy is effective in a canine model of Duchenne muscular dystrophy. Nat Commun. 2017; 8: 16105.Google Scholar
MacKenzie, TC, Kobinger, GP, Louboutin, JP, Radu, A, Javazon, EH, Sena-Esteves, M, et al. Transduction of satellite cells after prenatal intramuscular administration of lentiviral vectors. J Gene Med. 2005; 7: 50–8.Google Scholar
Reay, DP, Bilbao, R, Koppanati, BM, Cai, L, O’Day, TL, Jiang, Z, et al. Full-length dystrophin gene transfer to the mdx mouse in utero. Gene Ther. 2008; 15: 531–6.Google Scholar
Gregory, LG, Waddington, SN, Holder, MV, Mitrophanous, KA, Buckley, SMK, Mosley, KL, et al. Highly efficient EIAV-mediated in utero gene transfer and expression in the major muscle groups affected by Duchenne muscular dystrophy. Gene Ther. 2004; 11: 1117–25.Google Scholar
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: 1355–62.Google Scholar
Weisz, B, David, AL, Gregory, LG, Perocheau, D, Ruthe, A, Waddington, SN, et al. Targeting the respiratory muscles of fetal sheep for prenatal gene therapy for Duchenne muscular dystrophy. Am J Obstet Gynecol. 2005; 193: 1105–9.Google Scholar
Mühle, C, Neuner, A, Park, J, Pacho, F, Jiang, Q, Waddington, SN, et al. Evaluation of prenatal intra-amniotic LAMB3 gene delivery in a mouse model of Herlitz disease. Gene Ther. 2006; 13: 1665–76.Google Scholar
Sato, M, Tanigawa, M, Kikuchi, N. Nonviral gene transfer to surface skin of mid-gestational murine embryos by intraamniotic injection and subsequent electroporation. Mol Reprod Dev. 2004; 69: 268–77.Google Scholar
Endoh, M, Koibuchi, N, Sato, M, Morishita, R, Kanzaki, T, Murata, Y, et al. Fetal gene transfer by intrauterine injection with microbubble-enhanced ultrasound. Mol Ther. 2002; 5: 501–8.Google Scholar
Yoshizawa, J, Li, XK, Fujino, M, Kimura, H, Mizuno, R, Hara, A, et al. Successful in utero gene transfer using a gene gun in midgestational mouse fetuses. J Pediatr Surg. 2004; 39: 81–4.Google Scholar
Endo, M, Zoltick, PW, Peranteau, WH, Radu, A, Muvarak, N, Ito, M, et al. Efficient in vivo targeting of epidermal stem cells by early gestational intraamniotic injection of lentiviral vector driven by the keratin 5 promoter. Mol Ther. 2008; 16: 131–7.Google Scholar
Endo, M, Zoltick, PW, Radu, A, Qiujie, J, Matsui, C, Marinkovich, PM, et al. Early intra-amniotic gene transfer using lentiviral vector improves skin blistering phenotype in a murine model of Herlitz junctional epidermolysis bullosa. Gene Ther. 2012; 19: 561–9.Google Scholar
Schneider, H, Mallepell, SS, Körber, I, Wohlfart, S, Dick, A, Wahlbuhl, M, et al. Prenatal correction of X-linked hypohidrotic ectodermal dysplasia. N Engl J Med. 2018; 378: 1604–10.Google Scholar
Suff, N, Karda, R, Bajaj-Elliott, M, Buckley, SMK, Tangney, M, Waddington, SN, et al. Cervical gene delivery of human beta-defensin-3(HBD-3) prevents ascending bacterial infection in pregnant mice. Reprod Sci. 2017; 24 (Suppl. 1): 55A.Google Scholar
Miller, SL, Loose, JM, Jenkin, G, Wallace, EM. The effects of sildenafil citrate (Viagra) on uterine blood flow and well being in the intrauterine growth-restricted fetus. Am J Obstet Gynecol. 2009; 200: 102. e1–7.Google Scholar
David, AL, Torondel, B, Zachary, I, Wigley, V, Nader, KA, 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: 1344–50.Google Scholar
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-A165 in the uterine arteries of pregnant sheep. Gene Ther. 2012; 19: 925–35.Google Scholar
Mehta, V, Abi-Nader, KN, Shangaris, P, Shaw, SWS, Filippi, E, Benjamin, E, et al. Local over-expression of VEGF-DΔNΔC in the uterine arteries of pregnant sheep results in long-term changes in uterine artery contractility and angiogenesis. PLoS One. 2014; 9: e100021.Google Scholar
Carr, DJ, Wallace, JM, Aitken, RP, Milne, JS, Mehta, V, Martin, JF, et al. Uteroplacental adenovirus vascular endothelial growth factor gene therapy increases fetal growth velocity in growth-restricted sheep pregnancies. Hum Gene Ther. 2014; 25: 375–84.Google Scholar
Carr, DJ, Wallace, JM, Aitken, RP, Milne, JS, Martin, JF, Zachary, IC, et al. Peri- and postnatal effects of prenatal adenoviral VEGF gene therapy in growth-restricted sheep. Biol Reprod. 2016; 94: 142.Google Scholar
Swanson, AM, Rossi, CA, Ofir, K, Mehta, V, Boyd, M, Barker, H, et al. Maternal therapy with Ad.VEGF-A165 increases fetal weight at term in a guinea pig model of fetal growth restriction. Hum Gene Ther. 2016; 27: 9971007.Google Scholar
Vaughan, OR, Rossi, CA, Ginsberg, Y, White, A, Hristova, M, Sebire, NJ, et al. Perinatal and long term effects of maternal uterine artery adenoviral VEGF-A165 gene therapy in the growth restricted guinea pig fetus. Am J Physiol Regul Integr Comp Physiol. 2018; 315: R344–53.Google Scholar
Gancberg, D, Hoeveler, A, Draghia-Akli, R. Gene therapy and gene transfer projects of the 7th Framework Programme for Research and Technological Development of the European Union. Hum Gene Ther Clin Dev. 2015; 26: 77.Google Scholar
Guo, ZS, Li, Q, Bartlett, DL, Yang, JY, Fang, B. Gene transfer: the challenge of regulated gene expression. Trends Mol Med. 2008; 14: 410–18.Google Scholar
Coutelle, C, Waddington, SN. Vector systems for prenatal gene therapy: choosing vectors for different Applications. In Prenatal Gene Therapy. Totowa, NJ: Humana Press, 2012, pp. 4153.Google Scholar
Merten, OW, Hebben, M, Bovolenta, C. Production of lentiviral vectors. Mol Ther Methods Clin Dev. 2016; 3: 16017.Google Scholar
Manceur, AP, Kim, H, Misic, V, Andreev, N, Dorion-Thibaudeau, J, Lanthier, S, et al. Scalable lentiviral vector production using stable HEK293SF producer cell lines. Hum Gene Ther Methods. 2017; 28: 330–9.Google Scholar
Douar, AM, Themis, M, Sandig, V, Friedmann, T, Coutelle, C. Effect of amniotic fluid on cationic lipid mediated transfection and retroviral infection. Gene Ther. 1996; 3: 789–96.Google Scholar
Engelstädter, M, Buchholz, CJ, Bobkova, M, Steidl, S, Merget-Millitzer, H, Willemsen, RA, et al. Targeted gene transfer to lymphocytes using murine leukaemia virus vectors pseudotyped with spleen necrosis virus envelope proteins. Gene Ther. 2001; 8: 1202–6.Google Scholar
Challita, P-M, Kohn, DB. Lack of expression from a retroviral vector after transduction of murine hematopoietic stem cells is associated with methylation in vivo. Proc Natl Acad Sci USA. 1994; 91: 2567–71.Google Scholar
Jinek, M, Chylinski, K, Fonfara, I, Hauer, M, Doudna, JA, Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science . 2012; 337: 816–21.Google Scholar
Cong, L, Ran, FA, Cox, D, Lin, S, Barretto, R, Habib, N, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013; 339: 819–23.Google Scholar
Fu, Y, Sander, JD, Reyon, D, Cascio, VM, Joung, JK. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat Biotechnol. 2014; 32: 279–84.Google Scholar
Slaymaker, IM, Gao, L, Zetsche, B, Scott, DA, Yan, WX, Zhang, F. Rationally engineered Cas9 nucleases with improved specificity. Science. 2016; 351: 84–8.Google Scholar
Kleinstiver, BP, Pattanayak, V, Prew, MS, Tsai, SQ, Nguyen, NT, Zheng, Z, et al. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature. 2016; 529: 490–5.Google Scholar
Ran, FA, Cong, L, Yan, WX, Scott, DA, Gootenberg, JS, Kriz, AJ, et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature. 2015; 520: 186–91.Google Scholar
Liang, P, Xu, Y, Zhang, X, Ding, C, Huang, R, Zhang, Z, et al. CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes. Protein Cell. 2015; 6: 363–72.Google Scholar
Fogarty, NME, McCarthy, A, Snijders, KE, Powell, BE, Kubikova, N, Blakeley, P, et al. Genome editing reveals a role for OCT4 in human embryogenesis. Nature. 2017; 550: 6773.Google Scholar
Ma, H, Marti-Gutierrez, N, Park, S-W, Wu, J, Lee, Y, Suzuki, K, et al. Correction of a pathogenic gene mutation in human embryos. Nature. 2017; 548: 413–19.Google Scholar
Suzuki, T, Asami, M, Perry, ACF. Asymmetric parental genome engineering by Cas9 during mouse meiotic exit. Sci Rep. 2014; 4: 7621.Google Scholar
David, AL, Weisz, B, Gregory, L, Themis, M, Cook, T, Roubliova, X, et al. Ultrasound-guided injection and occlusion of the trachea in fetal sheep. Ultrasound Obstet Gynecol. 2006; 28: 82–8.Google Scholar
Jiménez, JA, Eixarch, E, DeKoninck, P, Bennini, JR, Devlieger, R, Peralta, CF, et al. Balloon removal after fetoscopic endoluminal tracheal occlusion for congenital diaphragmatic hernia. Am J Obstet Gynecol. 2017; 217: 78. e1–78. e11.Google Scholar
Coutelle, C, Themis, M, Waddington, SN, Buckley, SMK, Gregory, LG, Nivsarkar, MS, et al. Gene therapy progress and prospects: fetal gene therapy – first proofs of concept – some adverse effects. Gene Ther. 2005; 12: 1601–7.Google Scholar
Huard, J, Lochmuller, H, Acsadi, G, Jani, A, Holland, P, Guerin, C, et al. Differential short-term transduction efficiency of adult versus newborn mouse tissues by adenoviral recombinants. Exp Mol Pathol. 1995; 62: 131–43.Google Scholar
Endo, M, Henriques-Coelho, T, Zoltick, PW, Stitelman, DH, Peranteau, WH, Radu, A, et al. The developmental stage determines the distribution and duration of gene expression after early intra-amniotic gene transfer using lentiviral vectors. Gene Ther. 2010; 17: 6171.Google Scholar
Brown, BD, Gentner, B, Cantore, A, Colleoni, S, Amendola, M, Zingale, A, et al. Endogenous microRNA can be broadly exploited to regulate transgene expression according to tissue, lineage and differentiation state. Nat Biotechnol. 2007; 25: 1457–67.Google Scholar
Shaw, SWS, 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: 109–16.Google Scholar
Steven 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: 1015–31.Google Scholar
Shaw, SWS, 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: 122–32.Google Scholar
Ellis, J. Silencing and variegation of gammaretrovirus and lentivirus vectors. Hum Gene Ther. 2005; 16: 1241–6.Google Scholar
Jerebtsova, M, Batshaw, ML, Ye, X. Humoral immune response to recombinant adenovirus and adeno-associated virus after in utero administration of viral vectors in mice. Pediatr Res. 2002; 52: 95104.Google Scholar
Seppen, J, van Til, NP, van der Rijt, R, Hiralall, JK, Kunne, C, Oude Elferink, RPJ. Immune response to lentiviral bilirubin UDP-glucuronosyltransferase gene transfer in fetal and neonatal rats. Gene Ther. 2006; 13: 672–7.Google Scholar
Manno, CS, Pierce, GF, Arruda, VR, Glader, B, Ragni, M, Rasko, JJ, et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med. 2006; 12: 342–7.Google Scholar
Wenstrom, KD, Andrews, WW, Bowles, NE, Towbin, JA, Hauth, JC, Goldenberg, RL. Intrauterine viral infection at the time of second trimester genetic amniocentesis. Obstet Gynecol. 1998; 92: 420–4.Google Scholar
Porada, CD, Park, PJ, Tellez, J, Ozturk, F, Glimp, HA, Almeida-Porada, G, et al. Male germ-line cells are at risk following direct-injection retroviral-mediated gene transfer in utero. Mol Ther. 2005; 12: 754–62.Google Scholar
Heikkilä, A, Hiltunen, MO, Turunen, MP, Keski-Nisula, L, Turunen, A-M, Räsänen, H, et al. Angiographically guided utero-placental gene transfer in rabbits with adenoviruses, plasmid/liposomes and plasmid/polyethyleneimine complexes. Gene Ther. 2001; 8: 784–8.Google Scholar
MacCalman, CD, Furth, EE, Omigbodun, A, Kozarsky, KF, Coutifaris, C, Strauss, JF. Transduction of human trophoblast cells by recombinant adenoviruses is differentiation dependent. Biol Reprod. 1996; 54: 682–91.Google Scholar
Desforges, M, Rogue, A, Pearson, N, Rossi, C, Olearo, E, Forster, R, et al. In vitro human placental studies to support adenovirus-mediated VEGF-DΔNΔC maternal gene therapy for the treatment of severe early-onset fetal growth restriction. Hum Gene Ther Clin Dev. 2018; 29: 1023.Google Scholar
Koi, H, Zhang, J, Makrigiannakis, A, Getsios, S, MacCalman, CD, Kopf, GS, et al. Differential expression of the coxsackievirus and adenovirus receptor regulates adenovirus infection of the placenta. Biol Reprod. 2001; 64: 1001–9.Google Scholar
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: 148–58.Google Scholar
Bedrosian, JC, Gratton, MA, Brigande, JV, Tang, W, Landau, J, Bennett, J. In vivo delivery of recombinant viruses to the fetal murine cochlea: transduction characteristics and long-term effects on auditory function. Mol Ther. 2006; 14: 328–35.Google Scholar
Hacein-Bey-Abina, S, Pai, S-Y, Gaspar, HB, Armant, M, Berry, CC, Blanche, S, et al. A modified γ-retrovirus vector for X-linked severe combined immunodeficiency. N Engl J Med. 2014; 371: 1407–17.Google Scholar
David, RM, Doherty, AT. Viral Vectors: The road to reducing genotoxicity. Toxicol Sci. 2017; 155: 315–25.Google Scholar
Themis, M, Waddington, SN, Schmidt, M, von Kalle, C, Wang, Y, Al-Allaf, F, et al. Oncogenesis following delivery of a nonprimate lentiviral gene therapy vector to fetal and neonatal mice. Mol Ther. 2005; 12: 763–71.Google Scholar
Chandler, RJ, LaFave, MC, Varshney, GK, Trivedi, NS, Carrillo-Carrasco, N, Senac, JS, et al. Vector design influences hepatic genotoxicity after adeno-associated virus gene therapy. J Clin Invest. 2015; 125: 870–80.Google Scholar
Morrow, SL, Larson, JE, Nelson, S, Sekhon, HS, Ren, T, Cohen, JC. Modification of development by the CFTR gene in utero. Mol Genet Metab. 1998; 65: 203–12.Google Scholar
Larson, JE, Delcarpio, JB, Farberman, MM, Morrow, SL, Cohen, JC. CFTR modulates lung secretory cell proliferation and differentiation. Am J Physiol Lung Cell Mol Physiol. 2000; 279: L333–41.Google Scholar
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: 346–55.Google Scholar
Tarantal, AF, Chen, H, Shi, TT, Lu, CH, Fang, AB, Buckley, S, et al. Overexpression of transforming growth factor-β1 in fetal monkey lung results in prenatal pulmonary fibrosis. Eur Respir J. 2010; 36: 907–14.Google Scholar
Pahal, GS, Jauniaux, E, Kinnon, C, Thrasher, AJ, Rodeck, CH. Normal development of human fetal hematopoiesis between eight and seventeen weeks’ gestation. Am J Obstet Gynecol. 2000; 183: 1029–34.Google Scholar
Sheppard, M, Spencer, RN, Ashcroft, R, David, AL. Ethics and social acceptability of a proposed clinical trial using maternal gene therapy to treat severe early-onset fetal growth restriction. Ultrasound Obstet Gynecol. 2016; 47: 484–91.Google Scholar
European Medicines Agency. (2018). Guideline on the quality, non-clinical and clinical aspects of gene therapy medicinal products. https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-quality-non-clinical-clinical-aspects-gene-therapy-medicinal-products_en.pdfGoogle Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×