We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Nanotechnological approaches for the treatment of placental dysfunction: recent trends and future perspectives

    Jian Zhao‡

    Delivery Rooms, Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Jungang Zhang‡

    General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Yan Xu‡

    Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Juan Dong

    Delivery Rooms, Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China

    ,
    Qichao Dong

    Delivery Rooms, Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China

    ,
    Guoqiang Zhao

    Delivery Rooms, Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China

    &
    Ying Shi

    *Author for correspondence:

    E-mail Address: shiyingtj@163.com

    Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China

    Published Online:https://doi.org/10.2217/nnm-2023-0194

    The transitory placenta develops during pregnancy and mediates the blood flow between the mother and the developing baby. Placental dysfunction, including but not limited to placenta accreta spectrum, fetal growth restriction, preeclampsia and gestational trophoblastic disease, arises from abnormal placental development and can result in significant adverse maternal and fetal health outcomes. Unfortunately, there is a lack of treatment alternatives for these disorders. Nanocarriers offer versatility, including extended circulation, organ-specific targeting and intracellular transport, finely tuning therapeutic placental interactions. This thorough review explores nanotechnological strategies for addressing placental disorders, encompassing dysfunction insights, potential drug-delivery targets and recent strides in placenta-targeted nanoparticle (NP) therapies, instilling hope for effective placental malfunction treatment.

    Plain language summary

    The placenta, essential for mother–baby blood exchange, may experience catastrophic abnormalities during pregnancy. Treating these issues is challenging since you must focus on the placenta while protecting the infant. Nanotechnology might be helpful in this scenario. Nanocarriers are small carriers that can transport medications to the placenta and other particular locations in the body. They can aid in the treatment of various placental issues. In our present review, we discuss nanotechnology's solutions to these issues. We discuss what goes wrong, potential therapeutic applications for nanocarriers and recent developments in their use. This might be a novel approach to treating placenta issues and maintaining the health of mothers and infants.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Chaddha V, Viero S, Huppertz B, Kingdom J. Developmental biology of the placenta and the origins of placental insufficiency. Semin. Fetal Neonatal Med. 9(5), 357–369 (2004).
    • 2. Mazarico E, Molinet-Coll C, Martinez-Portilla RJ, Figueras F. Heparin therapy in placental insufficiency: systematic review and meta-analysis. Acta Obstet. Gynecol. Scand. 99(2), 167–174 (2020).
    • 3. Agarwal R, Tiwari A, Wadhwa N, Radhakrishnan G. Placental histopathological findings in pre-term/term and early/late onset small for gestation age: are they significant? Indian J. Pathol. Microbiol. 60(2), 232 (2017).
    • 4. Ohgiya Y, Nobusawa H, Seino N et al. MR imaging of fetuses to evaluate placental insufficiency. Magn. Reson. Med. Sci. 15(2), 212–219 (2016).
    • 5. Chappell LC, David AL. Improving the pipeline for developing and testing pharmacological treatments in pregnancy. PLOS Med. 13(11), e1002161 (2016).
    • 6. Mitchell AA, Gilboa SM, Werler MM et al. Medication use during pregnancy, with particular focus on prescription drugs: 1976–2008. Am. J. Obstet. Gynecol. 205(1), 51.e51–51.e58 (2011).
    • 7. Buhimschi CS, Weiner CP. Medications in pregnancy and lactation. Obstet. Gynecol. 114(1), 167–168 (2009).
    • 8. Burkey BW, Holmes AP. Evaluating medication use in pregnancy and lactation: what every pharmacist should know. J. Pediatr. Pharmacol. Ther. 18(3), 247–258 (2013).
    • 9. Bragazzi NL. Nanomedicine: insights from a bibliometrics-based analysis of emerging publishing and research trends. Medicina 55(12), 785 (2019).
    • 10. Kim BY, Rutka JT, Chan WC. Nanomedicine. N. Engl. J. Med. 363(25), 2434–2443 (2010).
    • 11. Kannan RM, Kannan S. Emerging nanomedicine approaches in obstetrics. Am. J. Obstet. Gynecol. 216(3), 201–203 (2017).
    • 12. Glassman PM, Muzykantov VR. Pharmacokinetic and pharmacodynamic properties of drug delivery systems. J. Pharmacol. Exp. Ther. 370(3), 570–580 (2019).
    • 13. Lu W, Yao J, Zhu X, Qi Y. Nanomedicines: redefining traditional medicine. Biomed. Pharmacother. 134, 111103 (2021).
    • 14. Chen T, Liu W, Xiong S et al. Nanoparticles mediating the sustained puerarin release facilitate improved brain delivery to treat Parkinson's disease. ACS Appl. Mater. Interfaces 11(48), 45276–45289 (2019).
    • 15. Burton GJ, Fowden AL. The placenta: a multifaceted, transient organ. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 370(1663), 20140066 (2015). •• This manuscript discusses the placenta's transient and diverse role in fetal development. It emphasizes the placenta's anatomical modifications to efficiently exchange maternal and fetal circulations and give oxygen and nutrients to the developing fetus, especially the brain. The article also discusses how placental hormones affect maternal metabolism and energy reserves during pregnancy and postpartum.
    • 16. Burton G, Charnock-Jones D, Jauniaux E. Regulation of vascular growth and function in the human placenta. Reproduction 138(6), 895–902 (2009).
    • 17. Fisher SJ. Why is placentation abnormal in preeclampsia? Am. J. Obstet. Gynecol. 213(4), S115–S122 (2015).
    • 18. Romero R, Kusanovic JP, Chaiworapongsa T, Hassan SS. Placental bed disorders in pre-term labor, pre-term PROM, spontaneous abortion and abruptio placentae. Best Pract. Res. Clin. Obstet. Gynaecol. 25(3), 313–327 (2011).
    • 19. Weiner E, Feldstein O, Tamayev L et al. Placental histopathological lesions in correlation with neonatal outcome in preeclampsia with and without severe features. Pregnancy Hypertens. 12, 6–10 (2018).
    • 20. Silver RM. Examining the link between placental pathology, growth restriction, and stillbirth. Best Pract. Res. Clin. Obstet. Gynaecol. 49, 89–102 (2018).
    • 21. Barker DJ. The developmental origins of chronic adult disease. Acta Paediatr. 93, 26–33 (2004).
    • 22. Barker DJ. The developmental origins of well-being. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 359(1449), 1359–1366 (2004).
    • 23. Salafia C, Pezzullo J, Ghidini A, Lopez-Zeno J, Whittington S. Clinical correlations of patterns of placental pathology in pre-term preeclampsia. Placenta 19(1), 67–72 (1998). • Analyzing a database of nonanomalous singleton live births, they find links between severe maternal proteinuria and placental chronic inflammation and associations of lower platelet counts with placental abruption and infarct. Uteroplacental vascular lesions correlate with lower birthweight percentiles and lighter placentae, highlighting the intricate relationship between placental pathology and adverse pregnancy outcomes.
    • 24. Trudinger B, Cook C, Jones L, Giles W. A comparison of fetal heart rate monitoring and umbilical artery waveforms in the recognition of fetal compromise. Br. J. Obstet. Gynaecol. 93(2), 171–175 (1986).
    • 25. Trudinger BJ, Giles WB. Elaboration of stem villous vessels in growth restricted pregnancies with abnormal umbilical artery Doppler waveforms. Br. J. Obstet. Gynaecol. 103(5), 487–489 (1996).
    • 26. Gagnon R, Johnston L, Murotsuki J. Fetal placental embolization in the late-gestation ovine fetus: alterations in umbilical blood flow and fetal heart rate patterns. Am. J. Obstet. Gynecol. 175(1), 63–72 (1996).
    • 27. Phipps E, Prasanna D, Brima W, Jim B. Preeclampsia: updates in pathogenesis, definitions, and guidelines. Clin. J. Am. Soc. Nephrol. 11(6), 1102 (2016). •• Delves into pathogenesis, elucidating the roles of various factors such as angiogenic pathways, autoantibodies, nitric oxide and oxidative stress. The shift in viewing preeclampsia as a systemic disease with long-term cardiovascular implications is explored, leading to updated definitions, classification and treatment strategies, emphasizing improved care for at-risk women beyond childbirth.
    • 28. Redman CW, Sargent IL. Immunology of preeclampsia. Am. J. Reprod. Immunol. 63(6), 534–543 (2010).
    • 29. Redman CW, Sargent IL, Taylor RN. Immunology of normal pregnancy and preeclampsia. In: Chesley's Hypertensive Disorders in Pregnancy. Elsevier, London, UK 161–179 (2015).
    • 30. Ridder A, Giorgione V, Khalil A, Thilaganathan B. Preeclampsia: the relationship between uterine artery blood flow and trophoblast function. Int. J. Mol. Sci. 20(13), 3263 (2019). • Contrary to conventional understanding, the study highlights emerging evidence suggesting that abnormal placental perfusion could lead to trophoblast impairment; it emphasizes the critical role of maternal cardiovascular function in the pathophysiology of preeclampsia, shedding new light on the complex interplay between maternal factors and placental health.
    • 31. Burton G, Yung H, Murray A. Mitochondrial–endoplasmic reticulum interactions in the trophoblast: stress and senescence. Placenta 52, 146–155 (2017).
    • 32. Mayne BT, Leemaqz SY, Smith AK, Breen J, Roberts CT, Bianco-Miotto T. Accelerated placental aging in early onset preeclampsia pregnancies identified by DNA methylation. Epigenomics 9(3), 279–289 (2017).
    • 33. Sohlberg S, Mulic-Lutvica A, Lindgren P, Ortiz-Nieto F, Wikström A-K, Wikström J. Placental perfusion in normal pregnancy and early and late preeclampsia: a magnetic resonance imaging study. Placenta 35(3), 202–206 (2014).
    • 34. Jung E, Romero R, Yeo L et al. The etiology of preeclampsia. Am. J. Obstet. Gynecol. 226(2), S844–S866 (2022).
    • 35. Sibai BM. Preeclampsia as a cause of pre-term and late pre-term (near-term) births. Semin. Perinatol. 30(1), 16–19 (2006).
    • 36. Huppertz B. Placental origins of preeclampsia: challenging the current hypothesis. Hypertension 51(4), 970–975 (2008).
    • 37. Redman CW, Staff AC. Preeclampsia, biomarkers, syncytiotrophoblast stress, and placental capacity. Am. J. Obstet. Gynecol. 213(4), S9.e1–S9.e4 (2015).
    • 38. Burton GJ, Jauniaux E. Placental oxidative stress: from miscarriage to preeclampsia. J. Soc. Gynecol. Investig. 11(6), 342–352 (2004).
    • 39. 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. 218(2), S855–S868 (2018).
    • 40. Owens JA. Endocrine and substrate control of fetal growth: placental and maternal influences and insulin-like growth factors. Reprod. Fertil. Dev. 3(5), 501–517 (1991). •• Placental transfer capacity and utilization of substrates significantly influence substrate transfer from mother to fetus, orchestrated by factors like oxygen, glucose and IGF-I concentrations. Placental hormones and proteins further sculpt this delicate balance, ensuring optimal maternal metabolic adaptation and nourishment for fetal development during pregnancy.
    • 41. Vorherr H. Factors influencing fetal growth. Am. J. Obstet. Gynecol. 142(5), 577–588 (1982).
    • 42. Sharma D, Shastri S, Sharma P. Intrauterine growth restriction: antenatal and postnatal aspects. Clin. Med. Insights Pediatr. 10, doi:10.4137/CMPed.S40070 (2016).
    • 43. Burton GJ, Jauniaux E. Pathophysiology of placental-derived fetal growth restriction. Am. J. Obstet. Gynecol. 218(2), S745–S761 (2018).
    • 44. Fowden A, Forhead A. Endocrine mechanisms of intrauterine programming. Reproduction 127(5), 515–526 (2004).
    • 45. Murphy VE, Smith R, Giles WB, Clifton VL. Endocrine regulation of human fetal growth: the role of the mother, placenta, and fetus. Endocr. Rev. 27(2), 141–169 (2006).
    • 46. Agrogiannis GD, Sifakis S, Patsouris ES, Konstantinidou AE. Insulin-like growth factors in embryonic and fetal growth and skeletal development. Mol. Med. Rep. 10(2), 579–584 (2014).
    • 47. Netchine I, Azzi S, Le Bouc Y, Savage MO. IGF1 molecular anomalies demonstrate its critical role in fetal, postnatal growth and brain development. Best Pract. Res. Clin. Endocrinol. Metab. 25(1), 181–190 (2011).
    • 48. Morris RK, Bilagi A, Devani P, Kilby MD. Association of serum PAPP-A levels in first trimester with small for gestational age and adverse pregnancy outcomes: systematic review and meta-analysis. Prenat. Diagn. 37(3), 253–265 (2017).
    • 49. Bartels HC, Postle JD, Downey P, Brennan DJ. Placenta accreta spectrum: a review of pathology, molecular biology, and biomarkers. Dis. Markers 2018, 1–11(2018).
    • 50. Morlando M, Collins S. Placenta accreta spectrum disorders: challenges, risks, and management strategies. Int. J. Womens Health 2022, 1033–1045 (2020).
    • 51. Hecht JL, Baergen R, Ernst LM et al. Classification and reporting guidelines for the pathology diagnosis of placenta accreta spectrum (PAS) disorders: recommendations from an expert panel. Mod. Pathol. 33(12), 2382–2396 (2020).
    • 52. Jauniaux E, Collins S, Burton GJ. Placenta accreta spectrum: pathophysiology and evidence-based anatomy for prenatal ultrasound imaging. Am. J. Obstet. Gynecol. 218(1), 75–87 (2018).
    • 53. Linn RL, Miller ES, Lim G, Ernst LM. Adherent basal plate myometrial fibers in the delivered placenta as a risk factor for development of subsequent placenta accreta. Placenta 36(12), 1419–1424 (2015).
    • 54. Büke B, Akkaya H, Demir S et al. Relationship between first trimester aneuploidy screening test serum analytes and placenta accreta. J. Matern. Fetal Neonatal Med. 31(1), 59–62 (2018).
    • 55. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2(12), 751–760 (2007).
    • 56. Fliedel L, Alhareth K, Mignet N, Fournier T, Andrieux K. Placental models for evaluation of nanocarriers as drug delivery systems for pregnancy associated disorders. Biomedicines 10(5), 936 (2022).
    • 57. Valero L, Alhareth K, Gil S et al. Nanomedicine as a potential approach to empower the new strategies for the treatment of preeclampsia. Drug Discov. Today 23(5), 1099–1107 (2018).
    • 58. Burton G, Woods A, Jauniaux E, Kingdom J. Rheological and physiological consequences of conversion of the maternal spiral arteries for uteroplacental blood flow during human pregnancy. Placenta 30(6), 473–482 (2009).
    • 59. Adekiya TA, Kondiah PP, Choonara YE, Kumar P, Pillay V. A review of nanotechnology for targeted anti-schistosomal therapy. Front. Bioeng. Biotechnol. 8, 32 (2020).
    • 60. Pritchard N, Kaitu'u-Lino Tu, Harris L, Tong S, Hannan N. Nanoparticles in pregnancy: the next frontier in reproductive therapeutics. Hum. Reprod. Update 27(2), 280–304 (2021).
    • 61. Alhareth K, Sancey L, Tsapis N, Mignet N. How should we plan the future of nanomedicine for cancer diagnosis and therapy. Int. J. Pharm. 532(2), 657–659 (2017).
    • 62. Slingerland M, Guchelaar H-J, Gelderblom H. Liposomal drug formulations in cancer therapy: 15 years along the road. Drug Discov. Today 17(3–4), 160–166 (2012).
    • 63. King A, Ndifon C, Lui S et al. Tumor-homing peptides as tools for targeted delivery of payloads to the placenta. Sci. Adv. 2(5), e1600349 (2016).
    • 64. Cureton N, Korotkova I, Baker B et al. Selective targeting of a novel vasodilator to the uterine vasculature to treat impaired uteroplacental perfusion in pregnancy. Theranostics 7(15), 3715 (2017).
    • 65. 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 7(11), 2940 (2017).
    • 66. Zhang B, Tan L, Yu Y et al. Placenta-specific drug delivery by trophoblast-targeted nanoparticles in mice. Theranostics 8(10), 2765 (2018).
    • 67. Trac N, Ashraf A, Giblin J, Prakash S, Mitragotri S, Chung EJ. Spotlight on genetic kidney diseases: a call for drug delivery and nanomedicine solutions. ACS Nano 17(7), 6165–6177 (2023).
    • 68. Morton PD, Ishibashi N, Jonas RA. Neurodevelopmental abnormalities and congenital heart disease: insights into altered brain maturation. Circ. Res. 120(6), 960–977 (2017).
    • 69. Carr H, Cnattingius S, Granath F, Ludvigsson JF, Edstedt Bonamy A-K. Pre-term birth and risk of heart failure up to early adulthood. J. Am. Coll. Cardiol. 69(21), 2634–2642 (2017).
    • 70. Solimani F, Narenji F, Pourmohsen M, Khalrghinejad K, Mehran N. Cerebral palsy and patterns of magnetic resonance imaging (MRI): a review. Iran. Rehabil. J. 12(4), 59–64 (2014).
    • 71. Abd Ellah N, Taylor L, Troja W et al. Development of non-viral, trophoblast-specific gene delivery for placental therapy. PLOS ONE 10(10), e0140879 (2015).
    • 72. Dodd A, Natfji AA, Evangelinos A et al. Conjugation to PEG as a strategy to limit the uptake of drugs by the placenta: potential applications for drug administration in pregnancy. Mol. Pharm. 19(1), 345–353 (2021).
    • 73. Kuna M, Waller JP, Logue OC, Bidwell GL III. Polymer size affects biodistribution and placental accumulation of the drug delivery biopolymer elastin-like polypeptide in a rodent pregnancy model. Placenta 72, 20–27 (2018).
    • 74. N'Dea S, Nelson KM, Dang MN, Gleghorn JP, Day ES. Gold nanoparticle biodistribution in pregnant mice following intravenous administration varies with gestational age. Nanomed. Nanotechnol. Biol. Med. 36, 102412 (2021).
    • 75. Muoth C, Großgarten M, Karst U et al. Impact of particle size and surface modification on gold nanoparticle penetration into human placental microtissues. Nanomedicine (Lond.) 12(10), 1119–1133 (2017).
    • 76. Poulsen MS, Mose T, Maroun LL, Mathiesen L, Knudsen LE, Rytting E. Kinetics of silica nanoparticles in the human placenta. Nanotoxicology 9(Suppl. 1), 79–86 (2015).
    • 77. Alfaifi AA, Heyder RS, Bielski ER et al. Megalin-targeting liposomes for placental drug delivery. J. Control. Rel. 324, 366–378 (2020).
    • 78. Ho D, Leong JW, Crew RC et al. Maternal–placental–fetal biodistribution of multimodal polymeric nanoparticles in a pregnant rat model in mid and late gestation. Sci. Rep. 7(1), 2866 (2017).
    • 79. Juch H, Nikitina L, Reimann S et al. Dendritic polyglycerol nanoparticles show charge dependent bio-distribution in early human placental explants and reduce hCG secretion. Nanotoxicology 12(2), 90–103 (2018).
    • 80. Menjoge AR, Rinderknecht AL, Navath RS et al. Transfer of PAMAM dendrimers across human placenta: prospects of its use as drug carrier during pregnancy. J. Control. Rel. 150(3), 326–338 (2011).
    • 81. Sibai B, Dekker G, Kupferminc M. Preeclampsia. Lancet 365(9461), 785–799 (2005).
    • 82. McCarthy FP, Kenny LC. Hypertension in pregnancy. Obstet. Gynaecol. Reprod. Med. 22(6), 141–147 (2012).
    • 83. Tranquilli A, Dekker G, Magee L et al. The classification, diagnosis and management of the hypertensive disorders of pregnancy: a revised statement from the ISSHP. Pregnancy Hypertens. 4(2), 97–104 (2014).
    • 84. Maynard SE, Min J-Y, Merchan J et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J. Clin. Investig. 111(5), 649–658 (2003).
    • 85. Gilbert JS, Verzwyvelt J, Colson D, Arany M, Karumanchi SA, Granger JP. Recombinant vascular endothelial growth factor 121 infusion lowers blood pressure and improves renal function in rats with placentalischemia-induced hypertension. Hypertension 55(2), 380–385 (2010).
    • 86. Woods AK, Hoffmann DS, Weydert CJ et al. Adenoviral delivery of VEGF121 early in pregnancy prevents spontaneous development of preeclampsia in BPH/5 mice. Hypertension 57(1), 94–102 (2011).
    • 87. Logue OC, Mahdi F, Chapman H, George EM, Bidwell GL III. A maternally sequestered, biopolymer-stabilized vascular endothelial growth factor (VEGF) chimera for treatment of preeclampsia. J. Am. Heart Assoc. 6(12), e007216 (2017).
    • 88. Alexander BT, Cockrell KL, Massey MB, Bennett WA, Granger JP. Tumor necrosis factor-α-induced hypertension in pregnant rats results in decreased renal neuronal nitric oxide synthase expression. Am. J. Hypertens. 15(2), 170–175 (2002).
    • 89. Yu J, Jia J, Guo X, Chen R, Feng L. Modulating circulating sFlt1 in an animal model of preeclampsia using PAMAM nanoparticles for siRNA delivery. Placenta 58, 1–8 (2017).
    • 90. Turanov AA, Lo A, Hassler MR et al. RNAi modulation of placental sFLT1 for the treatment of preeclampsia. Nat. Biotechnol. 36(12), 1164–1173 (2018).
    • 91. Li L, Yang H, Chen P et al. Trophoblast-targeted nanomedicine modulates placental sFLT1 for preeclampsia treatment. Front. Bioeng. Biotechnol. 8, 64 (2020).
    • 92. Suhag A, Berghella V. Intrauterine growth restriction (IUGR): etiology and diagnosis. Curr. Obstet. Gynecol. Rep. 2(2), 102–111 (2013).
    • 93. Ferretti C, Bruni L, Dangles-Marie V, Pecking A, Bellet D. Molecular circuits shared by placental and cancer cells, and their implications in the proliferative, invasive and migratory capacities of trophoblasts. Hum. Reprod. Update 13(2), 121–141 (2007).
    • 94. Constância M, Hemberger M, Hughes J et al. Placental-specific IGF-II is a major modulator of placental and fetal growth. Nature 417(6892), 945–948 (2002).
    • 95. Forbes K, Westwood M, Baker PN, Aplin JD. Insulin-like growth factor I and II regulate the life cycle of trophoblast in the developing human placenta. Am. J. Physiol. Cell Physiol. 294(6), C1313–C1322 (2008).
    • 96. King A, Ndifon C, Lui S et al. Tumor-homing peptides as tools for targeted delivery of payloads to the placenta. Sci. Adv. 2(5), e1600349 (2016).
    • 97. 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 7(11), 2940 (2017).
    • 98. Cureton N, Korotkova I, Baker B et al. Selective targeting of a novel vasodilator to the uterine vasculature to treat impaired uteroplacental perfusion in pregnancy. Theranostics 7(15), 3715 (2017).
    • 99. Lyall F, Greer I, Young A, Myatt L. Nitric oxide concentrations are increased in the feto–placental circulation in intrauterine growth restriction. Placenta 17(2–3), 165–168 (1996).
    • 100. Myatt L, Eis A, Brockman D, Greer I, Lyall F. Endothelial nitric oxide synthase in placental villous tissue from normal, pre-eclamptic and intrauterine growth restricted pregnancies. Hum. Reprod. 12(1), 167–172 (1997).
    • 101. Morgan TK. Role of the placenta in pre-term birth: a review. Am. J. Perinatol. 33(03), 258–266 (2016).
    • 102. Blackwell SC, Gyamfi-Bannerman C, Biggio JR Jr et al. 17-OHPC to prevent recurrent pre-term birth in singleton gestations (PROLONG study): a multicenter, international, randomized double-blind trial. Am. J. Perinatol. 37(02), 127–136 (2020).
    • 103. Rubin R. Confirmatory trial for drug to prevent pre-term birth finds no benefit, so why is it still prescribed? J. Am. Med. Assoc. 323(13), 1229–1232 (2020).
    • 104. Norman JE, Marlow N, Messow C-M et al. Vaginal progesterone prophylaxis for pre-term birth (the OPPTIMUM study): a multicentre, randomised, double-blind trial. Lancet 387(10033), 2106–2116 (2016).
    • 105. Gandell DL, Randell MD, Gudeman JL. FDA approved vs pharmacy compounded 17-OHPC – current issues for obstetricians to consider in reducing recurrent pre-term birth. Curr. Med. Res. Opin. 36(8), 1393–1401 (2020).
    • 106. Refuerzo JS, Leonard F, Bulayeva N et al. Uterus-targeted liposomes for pre-term labor management: studies in pregnant mice. Sci. Rep. 6(1), 34710 (2016).
    • 107. Refuerzo JS, Alexander JF, Leonard F, Leon M, Longo M, Godin B. Liposomes: a nanoscale drug carrying system to prevent indomethacin passage to the fetus in a pregnant mouse model. Am. J. Obstet. Gynecol. 212(4), 508.e501–508.e507 (2015).
    • 108. Paul JW, Hua S, Ilicic M et al. Drug delivery to the human and mouse uterus using immunoliposomes targeted to the oxytocin receptor. Am. J. Obstet. Gynecol. 216(3), 283.e281–283.e214 (2017).
    • 109. Greenwood IA, Yeung S, Tribe R, Ohya S. Loss of functional K+ channels encoded by ether-à-go-go-related genes in mouse myometrium prior to labour onset. J. Physiol. 587(10), 2313–2326 (2009).
    • 110. Bulletti C, de Ziegler D, Flamigni C et al. Targeted drug delivery in gynaecology: the first uterine pass effect. Hum. Reprod. 12(5), 1073–1079 (1997).
    • 111. Zierden HC, Ortiz JI, DeLong K et al. Enhanced drug delivery to the reproductive tract using nanomedicine reveals therapeutic options for prevention of pre-term birth. Sci. Transl. Med. 13(576), eabc6245 (2021).
    • 112. Wang B, Navath RS, Menjoge AR et al. Inhibition of bacterial growth and intramniotic infection in a guinea pig model of chorioamnionitis using PAMAM dendrimers. Int. J. Pharm. 395(1–2), 298–308 (2010).
    • 113. Lei J, Rosenzweig JM, Mishra MK et al. Maternal dendrimer-based therapy for inflammation-induced pre-term birth and perinatal brain injury. Sci. Rep. 7(1), 6106 (2017).
    • 114. Epps TH III, Vi T, Sullivan MO. Design and development of a robust photo-responsive block copolymer framework for tunable nucleic acid delivery and efficient gene silencing. Polymer J. 50(8), 711–723 (2018).
    • 115. Riley RS, Dang MN, Billingsley MM, Abraham B, Gundlach L, Day ES. Evaluating the mechanisms of light-triggered siRNA release from nanoshells for temporal control over gene regulation. Nano Lett. 18(6), 3565–3570 (2018).
    • 116. Li C, Liu X, Liu Y et al. Glucose and H2O2 dual-sensitive nanogels for enhanced glucose-responsive insulin delivery. Nanoscale 11(18), 9163–9175 (2019).
    • 117. Zhang C, Hong S, Liu M-D et al. pH-sensitive MOF integrated with glucose oxidase for glucose-responsive insulin delivery. J. Control. Rel. 320, 159–167 (2020).
    • 118. Volpatti LR, Matranga MA, Cortinas AB et al. Glucose-responsive nanoparticles for rapid and extended self-regulated insulin delivery. ACS Nano 14(1), 488–497 (2019).
    • 119. Van Calsteren K, Heyns L, Smet FD et al. Cancer during pregnancy: an analysis of 215 patients emphasizing the obstetrical and the neonatal outcomes. J. Clin. Oncol. 28(4), 683–689 (2010).
    • 120. Stensheim H, Møller B, van Dijk T, Fosså SD. Cause-specific survival for women diagnosed with cancer during pregnancy or lactation: a registry-based cohort study. J. Clin. Oncol. 27(1), 45–51 (2009).
    • 121. Wang S-W, Ma L-L, Huang S, Liang L, Zhang J-R. Role of cervical cerclage and vaginal progesterone in the treatment of cervical incompetence with/without pre-term birth history. Chin. Med. J. 129(22), 2670–2675 (2016).
    • 122. Hassan S, Romero R, Vidyadhari D et al. Vaginal progesterone reduces the rate of pre-term birth in women with a sonographic short cervix: a multicenter, randomized, double-blind, placebo-controlled trial. Ultrasound Obstet. Gynecol. 38(1), 18–31 (2011).
    • 123. Kistner RW. The treatment of endometriosis by inducing pseudopregnancy with ovarian hormones: a report of fifty-eight cases. Fertil. Steril. 10(6), 539–556 (1959).
    • 124. Yuan P, Huang Y, Wu H, Teng Z, Zhang J, Xin X. Induction of a local pseudo-pregnancy via levonorgestrel-loaded microspheres for the treatment of endometriosis in a rabbit model. Hum. Reprod. 25(2), 462–469 (2010).
    • 125. Shetty AN, Pautler R, Ghaghada K et al. A liposomal Gd contrast agent does not cross the mouse placental barrier. Sci. Rep. 6(1), 27863 (2016).
    • 126. Badachhape AA, Kumar A, Ghaghada KB et al. Preclinical magnetic resonance imaging of retroplacental clear space throughout gestation. Placenta 77, 1–7 (2019).
    • 127. Badachhape AA, Devkota L, Stupin IV et al. Nanoparticle contrast-enhanced T1-mapping enables estimation of placental fractional blood volume in a pregnant mouse model. Sci. Rep. 9(1), 18707 (2019).
    • 128. Ghaghada KB, Starosolski ZA, Bhayana S et al. Preclinical evaluation of a nanoparticle-based blood-pool contrast agent for MR imaging of the placenta. Placenta 57, 60–70 (2017).