New hope for preventing preterm birth: The promise of vaginal nanoformulations

Vaginal nanoformulations for preterm birth

Authors

  • Sandra Reznik Department of Pharmaceutical Sciences, St. John’s University, Queens 11439, NY, USA

DOI:

https://doi.org/10.54844/prm.2022.0100

References

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;388:3027–3035.

Blencowe H, Cousens C, Oestergaard MZ, Chou D, Moller AB, Narwal R, et al. National, regional and worldwide estimates of preterm birth. Lancet 2012;379:2162–2172.

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:S2.

Martin JA, Osterman MJK. Describing the increase in preterm births in the United States, 2014-2016. NCHS Data Brief 2018:1–8.

MacKay DF, Smith GCS, Dobbie R, Pell JP. Gestational age at delivery and special educational need: retrospective cohort study of 407,503 schoolchildren. PLoS Med 2010;7:e1000289.

Blackwell SC, Gyamfi-Bannerman C, Biggio JR Jr, Chauhan SP, Hughes BL, Louis JM, et al. 17-OHPC to prevent recurrent preterm birth in singleton gestations (PROLONG study):a multicenter, international, randomized double-blind trial. Am J Perinatol 2020;37:127–136.

Jarde A, Lutsiv O, Beyene J, McDonald SD. Vaginal progesterone, oral progesterone, 17-OHPC, cerclage, and pessary for preventing preterm birth in at-risk singleton pregnancies: an updated systematic review and network meta-analysis. BJOG 2019;126:556–567.

Romero R, Conde-Agudelo A, Da Fonseca E, O’Brien JM, Cetingoz E, Creasy GW, et al. Vaginal progesterone for preventing preterm birth and adverse perinatal outcomes in singleton gestations with a short cervix: a meta-analysis of individual patient data. Am J Obstet Gynecol 2018;218:161–180.

Lamont RF, Jørgensen JS. Safety and efficacy of tocolytics for the treatment of spontaneous preterm labour. Curr Pharm Des 2019;25:577–592.

Coler BS, Shynlova O, Boros-Rausch A, Lye S, McCartney S, Leimert KB, et al. Landscape of preterm birth therapeutics and a path forward. J Clin Med 2021;10:2912.

Srikrishna S, Cardozo L. The vagina as a route for drug delivery: a review. Int Urogynecol J 2013;24:537–543.

Bulletti C, de Ziegler D, Flamigni C, Giacomucci E, Polli V, Bolelli G, et al. Targeted drug delivery in gynaecology: the first uterine pass effect. Hum Reprod 1997;12:1073–1079.

Cicinelli E, de Ziegler D. Transvaginal progesterone: evidence for a new functional 'portal system' flowing from the vagina to the uterus. Hum Reprod Update 1999;5:365–372.

Alexander NJ, Baker E, Kaptein M, Karck U, Miller L, Zampaglione E. Why consider vaginal drug administration? Fertil Steril 2004;82:1–12.

Amabebe E, Anumba DOC. The vaginal microenvironment: the physiologic role of Lactobacilli. Front Med (Lausanne) 2018;5:181.

Adnane M, Meade KG, O’Farrelly C. Cervico-vaginal mucus (CVM)–an accessible source of immunologically informative biomolecules. Vet Res Commun 2018;42:255–263.

Vagios S, Mitchell CM. Mutual preservation: a review of interactions between cervicovaginal mucus and microbiota. Front Cell Infect Microbiol 2021;11:676114.

Bayda S, Adeel M, Tuccinardi T, Cordani M, Rizzolio F. The history of nanoscience and nanotechnology: from chemical-physical applications to nanomedicine. Molecules 2019;25:E112.

Nel AE MD/DSc. Transformational impact of nanomedicine: reconciling outcome with promise. Nano Lett 2020;20:5601–5603.

Lloyd-Parry O, Downing C, Aleisaei E, Jones C, Coward K. Nanomedicine applications in women’s health: state of the art. Int J Nanomedicine 2018;13:1963–1983.

Boddupalli BM, Mohammed ZNK, Nath RA, Banji D. Mucoadhesive drug delivery system: an overview. J Adv Pharm Technol Res 2010;1:381–387.

Lai SK, Wang YY, Hanes J. Mucus-penetrating nanoparticles for drug and gene delivery to mucosal tissues. Adv Drug Deliv Rev 2009;61:158–171.

Giusto K, Patki M, Koya J, Ashby CR Jr, Munnangi S, Patel K, et al. A vaginal nanoformulation of a SphK inhibitor attenuates lipopolysaccharide-induced preterm birth in mice. Nanomedicine 2019;14:2835–2851.

Patki M, Giusto K, Gorasiya S, Reznik SE, Patel K. 17-α hydroxyprogesterone nanoemulsifying preconcentrate-loaded vaginal tablet: a novel non-invasive approach for the prevention of preterm birth. Pharmaceutics 2019;11:E335.

Vyas V, Ashby CR Jr, Olgun NS, Sundaram S, Salami O, Munnangi S, et al. Inhibition of sphingosine kinase prevents lipopolysaccharide-induced preterm birth and suppresses proinflammatory responses in a murine model. Am J Pathol 2015;185:862–869.

Vyas V, Ashby CR Jr, Reznik SE. Sphingosine kinase: a novel putative target for the prevention of infection-triggered preterm birth. Obstet Gynecol Int 2013;2013:302952.

Sundaram S, Ashby CR Jr, Pekson R, Sampat V, Sitapara R, Mantell L, et al. N, N-dimethylacetamide regulates the proinflammatory response associated with endotoxin and prevents preterm birth. Am J Pathol 2013;183:422–430.

Pekson R, Poltoratsky V, Gorasiya S, Sundaram S, Ashby CR, Vancurova I, et al. N, N-dimethylacetamide significantly attenuates LPS- and TNFα-induced proinflammatory responses via inhibition of the nuclear factor kappa B pathway. Mol Med 2016;22:747–758.

Gorasiya S, Mushi J, Pekson R, Yoganathan S, Reznik SE. Repurposing N, N-dimethylacetamide (DMA), a pharmaceutical excipient, as a prototype novel anti-inflammatory agent for the prevention and/or treatment of preterm birth. Curr Pharm Des 2018;24:989–992.

Roberts DJ. New hope for the prevention of preterm birth. Am J Path 2013;83:330-32.

Zierden HC, Ortiz JI, DeLong K, Yu J, Li G, Dimitrion P, et al. Enhanced drug delivery to the reproductive tract using nanomedicine reveals therapeutic options for prevention of preterm birth. Sci Transl Med 2021;13:eabc6245.

Published

2022-10-18

How to Cite

1.
Reznik S. New hope for preventing preterm birth: The promise of vaginal nanoformulations: Vaginal nanoformulations for preterm birth. PRM. 2022;1. doi:10.54844/prm.2022.0100

Issue

Section

Perspective

Downloads

Download data is not yet available.
PERSPECTIVE

New hope for preventing preterm birth: The promise of vaginal nanoformulations


Sandra E. Reznik1,2

1Department of Pharmaceutical Sciences, St. John’s University, Queens 11439, NY, USA;

2Departments of Pathology and Obstetrics and Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx 10461, NY, USA


Address for correspondence:

Dr. Sandra E. Reznik, E-mail: rezniks@stjohns.edu

Received: 17 May 2022

Accepted: 07 September 2022

Published: 18 October 2022



Defined as birth before 37 completed weeks of gestation, preterm birth (PTB) accounts for the vast majority of perinatal morbidity and mortality.[1,2,3] Sadly, data collected by the World Health Organization indicate that the annual rate of PTB worldwide has not improved over the last several decades and is greater than 10% in most countries.[4] PTB rates are generally higher in the developing world; among industrialized nations, the United States (US) has the highest rate of PTB.[2] In the US particularly, advances in neonatology have occurred more rapidly than in the field of obstetrics, so that more and more neonates born closer and closer to the cusp of viability survive their neonatal intensive care unit stay, only to live with a constellation of medical challenges. The acute sequelae of PTB, such as respiratory distress syndrome and necrotizing enterocolitis, are replaced by lifelong respiratory, metabolic and neurologic abnormalities, including retinopathy of prematurity and cerebral palsy.[5] The personal and societal costs of PTB are enormous.


Currently, the only US Food and Drug Administration (FDA) approved drug for the prevention of PTB is hydroxyprogesterone caproate, aka Makena. However, the FDA is now calling for withdrawal of the approval of this drug, because of the lack of efficacy shown in the PROLONG trial.[6] Although not FDA approved, vaginal progesterone is the most successful drug therapy to prevent PTB and is administered to women who have had a PTB before and are at risk for delivering preterm. However, vaginal progesterone only reduces the incidence of PTB by one third.[7,8] As far as tocolytics, the three most commonly used are magnesium sulfate, indomethacin and nifedipine. However, these drugs only delay birth for approximately 48 hours,[9,10] which provides an opportunity to administer antenatal corticosteroids to promote lung maturity but does not increase gestational age or improve neonatal outcomes significantly. Beta mimetics, such as ritodrine and terbutaline, originally thought promising agents to prevent PTB, are now contraindicated due to toxicity.[10]


The paucity of pharmacotherapy to prevent PTB has resulted, at least partially, from concerns about potential teratogenic effects on the fetus and toxic effects on the mother, both of which result from off-target actions of drugs meant to function in either the cervix or the uterus. A useful first step for avoiding these complications is using the vaginal route of administration. Advantages of administering a drug vaginally include the presence of a dense network of blood vessels, large surface area, and ability to circumvent hepatic first-pass metabolism.[11] Most importantly, this route of administration takes advantage of the so-called “uterine first pass effect”. The uterine first pass effect allows drugs introduced into the vagina to be carried by the vaginal-cervical-uterine portal vascular system and accounts for why high concentrations of a vaginally administered drug accumulate in the cervix and uterus, while low concentrations accumulate in the systemic circulation.[12,13] Still, there are several disadvantages related to the vaginal route of administration. These include potentially poor absorption,[14] alterations to the vaginal microbiome[15] and a thick cervico-vaginal mucous layer—a natural barrier rich in glycoproteins and lipids preventing drugs from reaching their target tissues higher up in the gynecologic tract.[16,17]


The most promising advance to overcome these challenges is the revolution in pharmacology and pharmaceutics known as nanomedicine.[18,19,20] Vaginally administered nanoparticles loaded with drug cargo, specifically engineered for mucoadhesion[21] or mucopenetration,[22] can overcome the cervico-vaginal mucous barrier and be directed to the target tissue with reduced systemic drug levels and reduced penetration of the placental barrier. Several preclinical trials have demonstrated the utility of the vaginal nanoformulation approach.


In two different preclinical studies, we have shown that a vaginally administered self-nanoemulsifying drug delivery system (SNEDDS) can delay the onset of inflammation induced PTB.[23,24] After reporting that a sphingosine kinase inhibitor (SKI II, Cayman Chemicals, MI, USA) can prevent PTB when administered intraperitoneally,[25,26] we showed that a SNEDDS composed of oil, co-solvent and SKI II drug cargo significantly increased the number of pups rescued from PTB in lipopolysaccharide-induced mice.[23] In a second study, we showed that a similar vaginal formulation loaded with 17-alpha hydroxyprogesterone caproate (aka Makena) also delayed the onset of PTB and rescued a significant number of pups.[24] Work is now underway to test a vaginal nanoformulation of N,N-dimethylacetamide, a widely used drug excipient, which we have shown to be a candidate for re-purposing for PTB.[27,28,29,30] Finally, Ensign et al. have recently reported that a vaginal nanoformulation loaded with the histone deacetylase inhibitor Trichostatin A plus progesterone (P4) decreased inflammation-induced PTB by 50% in their murine model.[31]


The ability to direct efficacious levels of drug cargo to the cervix and uterus, while maintaining very low concentrations in the maternal systemic circulation and preventing significant amounts from crossing the placental barrier, heralds a new era in the long and unrewarding quest for drugs to delay PTB. The advent of nanomedicine raises the possibility of re-purposing existing drugs and testing new drugs that would otherwise not be candidates for obstetrical disorders. Efforts in the PTB drug development field should be refocused and vaginal nanoformulations should be placed in the spotlight.


Source of Funding

This perspective is supported by funding from NIH to Sandra E. Reznik (1R16GM145586).


Conflict of Interest

The author has a pending patent (13/536,94), titled “Administration of N,N-Dimethylacetamide and Its Monomethylated Metabolites for the Treatment of Inflammatory Disorders”. The author is an Editorial Board Member of the journal. The article was subject to the journal’s standard procedures, with peer review handled independently of this member and her research group.


  1. 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;388:3027–3035. DOI: 10.1016/S0140-6736(16)31593-8
  2. Blencowe H, Cousens C, Oestergaard MZ, Chou D, Moller AB, Narwal R, et al. National, regional and worldwide estimates of preterm birth. Lancet 2012;379:2162–2172. DOI: 10.1016/S0140-6736(12)60820-4
  3. 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:S2. DOI: 10.1186/1742-4755-10-S1-S2
  4. Martin JA, Osterman MJK. Describing the increase in preterm births in the United States, 2014-2016. NCHS Data Brief 2018:1–8. DOI: 10.1542/peds.140.1_MeetingAbstract.72
  5. MacKay DF, Smith GCS, Dobbie R, Pell JP. Gestational age at delivery and special educational need: retrospective cohort study of 407,503 schoolchildren. PLoS Med 2010;7:e1000289. DOI: 10.1371/journal.pmed.1000289
  6. Blackwell SC, Gyamfi-Bannerman C, Biggio JR Jr, Chauhan SP, Hughes BL, Louis JM, et al. 17-OHPC to prevent recurrent preterm birth in singleton gestations (PROLONG study): a multicenter, international, randomized double-blind trial. Am J Perinatol 2020;37:127–136. DOI: 10.1055/s-0039-3400227
  7. Jarde A, Lutsiv O, Beyene J, McDonald SD. Vaginal progesterone, oral progesterone, 17-OHPC, cerclage, and pessary for preventing preterm birth in at-risk singleton pregnancies: an updated systematic review and network meta-analysis. BJOG 2019;126:556–567. DOI: 10.1111/1471-0528.15566
  8. Romero R, Conde-Agudelo A, Da Fonseca E, O’Brien JM, Cetingoz E, Creasy GW, et al. Vaginal progesterone for preventing preterm birth and adverse perinatal outcomes in singleton gestations with a short cervix: a meta-analysis of individual patient data. Am J Obstet Gynecol 2018;218:161–180. DOI: 10.1016/j.ajog.2017.11.576
  9. Lamont RF, Jørg ensen JS. Safety and efficacy of tocolytics for the treatment of spontaneous preterm labour. Curr Pharm Des 2019;25:577–592. DOI: 10.2174/1381612825666190329124214
  10. Coler BS, Shynlova O, Boros-Rausch A, Lye S, McCartney S, Leimert KB, et al. Landscape of preterm birth therapeutics and a path forward. J Clin Med 2021;10:2912. DOI: 10.3390/jcm10132912
  11. Srikrishna S, Cardozo L. The vagina as a route for drug delivery: a review. Int Urogynecol J 2013;24:537–543. DOI: 10.1007/s00192-012-2009-3
  12. Bulletti C, de Ziegler D, Flamigni C, Giacomucci E, Polli V, Bolelli G, et al. Targeted drug delivery in gynaecology: the first uterine pass effect. Hum Reprod 1997;12:1073–1079. DOI: 10.1093/humrep/12.5.1073
  13. Cicinelli E, de Ziegler D. Transvaginal progesterone: evidence for a new functional ‘portal system’ flowing from the vagina to the uterus. Hum Reprod Update 1999;5:365–372. DOI: 10.1093/humupd/5.4.365
  14. Alexander NJ, Baker E, Kaptein M, Karck U, Miller L, Zampaglione E. Why consider vaginal drug administration? Fertil Steril 2004;82:1–12. DOI: 10.1016/j.fertnstert.2004.01.025
  15. Amabebe E, Anumba DOC. The vaginal microenvironment: the physiologic role of Lactobacilli. Front Med (Lausanne) 2018;5:181. DOI: 10.3389/fmed.2018.00181
  16. Adnane M, Meade KG, O’Farrelly C. Cervico-vaginal mucus (CVM)–an accessible source of immunologically informative biomolecules. Vet Res Commun 2018;42:255–263. DOI: 10.1007/s11259-018-9734-0
  17. Vagios S, Mitchell CM. Mutual preservation: a review of interactions between cer vicovaginal mucus and microbiota. Front Cell Infect Microbiol 2021;11:676114. DOI: 10.3389/fcimb.2021.676114
  18. Bayda S, Adeel M, Tuccinardi T, Cordani M, Rizzolio F. The history of nanoscience and nanotechnology: from chemical-physical applications to nanomedicine. Molecules 2019;25:E112. DOI: 10.3390/molecules25010112
  19. Nel AE MD/DSc. Transformational impact of nanomedicine: reconciling outcome with promise. Nano Lett 2020;20:5601–5603. DOI: 10.1021/acs.nanolett.0c02738
  20. Lloyd-Parry O, Downing C, Aleisaei E, Jones C, Coward K. Nanomedicine applications in women’s health: state of the art. Int J Nanomedicine 2018;13:1963–1983. DOI: 10.2147/IJN.S97572
  21. Boddupalli BM, Mohammed ZNK, Nath RA, Banji D. Mucoadhesive drug delivery system: an overview. J Adv Pharm Technol Res 2010;1:381–387. DOI: 10.4103/0110-5558.76436
  22. Lai SK, Wang YY, Hanes J. Mucus-penetrating nanoparticles for drug and gene delivery to mucosal tissues. Adv Drug Deliv Rev 2009;61:158–171. DOI: 10.1016/j.addr.2008.11.002
  23. Giusto K, Patki M, Koya J, Ashby CR Jr, Munnangi S, Patel K, et al. A vaginal nanoformulation of a SphK inhibitor attenuates lipopolysaccharide-induced preterm birth in mice. Nanomedicine 2019;14:2835–2851. DOI: 10.2217/nnm-2019-0243
  24. Patki M, Giusto K, Gorasiya S, Reznik SE, Patel K. 17-α hydroxyprogesterone nanoemulsifying preconcentrate-loaded vaginal tablet: a novel non-invasive approach for the prevention of preterm birth. Pharmaceutics 2019;11:E335. DOI: 10.3390/pharmaceutics11070335
  25. Vyas V, Ashby CR Jr, Olgun NS, Sundaram S, Salami O, Munnangi S, et al. Inhibition of sphingosine kinase prevents lipopolysaccharide-induced preterm birth and suppresses proinflammatory responses in a murine model. Am J Pathol 2015;185:862–869. DOI: 10.1016/j.ajpath.2014.10.026
  26. Vyas V, Ashby CR Jr, Reznik SE. Sphingosine kinase: a novel putative target for the prevention of infection-triggered preterm birth. Obstet Gynecol Int 2013;2013:302952. DOI: 10.1155/2013/302952
  27. Sundaram S, Ashby CR Jr, Pekson R, Sampat V, Sitapara R, Mantell L, et al. N, N-dimethylacetamide regulates the proinflammatory response associated with endotoxin and prevents preterm birth. Am J Pathol 2013;183:422–430. DOI: 10.1016/j.ajpath.2013.05.006
  28. Pekson R, Poltoratsky V, Gorasiya S, Sundaram S, Ashby CR, Vancurova I, et al. N, N-dimethylacetamide significantly attenuates LPS- and TNFα- induced proinflammatory responses via inhibition of the nuclear factor kappa B pathway. Mol Med 2016;22:747–758. DOI: 10.2119/molmed.2016.00017
  29. Gorasiya S, Mushi J, Pekson R, Yoganathan S, Reznik SE. Repurposing N, N-dimethylacetamide (DMA), a pharmaceutical excipient, as a prototype novel anti-inflammatory agent for the prevention and/or treatment of preterm birth. Curr Pharm Des 2018;24:989–992. DOI: 10.2174/1381612824666180130121706
  30. Roberts DJ. New hope for the prevention of preterm birth. Am J Path 2013;83:330-32. DOI: 10.1016/j.ajpath.2013.06.001
  31. Zierden HC, Or tiz JI, DeLong K, Yu J, Li G, Dimitrion P, et al. Enhanced drug delivery to the reproductive tract using nanomedicine reveals therapeutic options for prevention of preterm birth. Sci Transl Med 2021;13:eabc6245. DOI: 10.1126/scitranslmed.abc6245