Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Aug 1.
Published in final edited form as: Climacteric. 2018 May 23;21(4):315–320. doi: 10.1080/13697137.2018.1463982

Non-clinical studies of progesterone

R Sitruk-Ware 1
PMCID: PMC6281289  NIHMSID: NIHMS1512555  PMID: 29790373

Abstract

Progesterone is a steroid hormone that is essential for the regulation of reproductive function. Progesterone has been approved for several indications including the treatment of anovulatory menstrual cycles, assisted reproductive technology, contraception during lactation and, when combined with estrogen, for the prevention of endometrial hyperplasia in postmenopausal hormonal therapy. In addition to its role in reproduction, progesterone regulates a number of biologically distinct processes in other tissues, particularly in the nervous system.

This physiological hormone is poorly absorbed when administered in a crystalline form and is not active when given orally, unless in micronized form, or from different non-oral delivery systems that allow a more constant delivery rate. A limited number of preclinical studies have been conducted to document the toxicity, carcinogenicity and overall animal safety of progesterone delivered from different formulations, and these rather old studies showed no safety concern. More recently, it has been shown in animal experiments that progesterone, its metabolite allopregnanolone and structurally related progestins have positive effects on neuroregeneration and repair of brain damage, as well as myelin repair. These recent preclinical findings have the potential to accelerate therapeutic translation for multiple unmet neurological needs.

Keywords: Progesterone, pharmacodynamics, toxicology studies, carcinogenicity, neuroregeneration

Introduction

Progesterone plays an essential role in many aspects of reproduction, including ovulation, transformation of the endometrium, and pregnancy maintenance. The multiple and complex effects of progesterone on the female reproductive process are the result of its binding to and activation of progesterone receptors (PRs), including sub-units PR-A and PR-B13 and membrane receptors4,5.

Progesterone effects the secretory transformation of an estrogen-primed endometrium, a process which prepares the endometrium to receive and nourish a fertilized ovum. In addition, depending on the level of progesterone secretion and the duration of the luteal phase, progesterone can prevent the over-proliferation of endometrial tissue6.

The main indications for which progesterone has been approved include the treatment of anovulatory menstrual cycles, assisted reproductive technology (ART), and, when combined with estrogen, the prevention of endometrial hyperplasia in postmenopausal hormonal replacement therapy regimens68.

A few products delivering the physiological hormone progesterone have been developed and approved for various indications including hormonal therapy. These include micronized progesterone given orally or via a vaginal insert, and products designed to deliver micronized progesterone directly from the vaginal mucosa: a vaginal ring9, a vaginal gel10 and a vaginal insert11. Micronized progesterone is identical to the naturally occurring hormone secreted by the ovary, placenta, and adrenal gland. Therefore, in contrast to new chemical entities, there were few preclinical studies conducted with these products911.

Non-clinical pharmacology

Receptor binding

Progesterone has been shown to bind to progesterone receptors (PR-A and PR-B) in reproductive organs, including ovary, fallopian tubes, uterus, and mammary gland13, as well as to PRs in the anterior pituitary and hypothalamus12,13. PR-A appears to be necessary for progesterone-dependent aspects of female fertility, while PR-B is involved in progesterone-related differentiation of mammary glandular tissue3. Nuclear PR has also been found in thymus, vascular smooth muscle, bone, and the peripheral nervous system3,13 as well as in primate and human male reproductive organs, including the epididymis, prostate, and male mammary gland14,15. Membrane PRs have been found in the reproductive and non-reproductive tissues of rat and primate4,5.

Pharmacodynamics

Pharmacokinetics (absorption, distribution, and excretion) determine how much of the steroid administered is available to tissues, primarily by measuring its blood level, and the amount that enters the cells is determined by the extent to which it is bound to carrier proteins that cannot cross the cell membranes. After progesterone enters the systemic circulation, it is distributed between blood and tissues by passive diffusion. The pattern of distribution is mainly regulated by its binding to transport proteins and tissue receptors. A relatively small amount of progesterone is bound with high affinity and low capacity to corticosteroid-binding globulin but not to sex hormone-binding globulin6.

Primary pharmacodynamics

The primary pharmacodynamics of natural progesterone given by non-oral routes of administration has been evaluated in several animal species1618.

The effects of progesterone delivered via a vaginal ring on vaginal bleeding, ovarian function, and the endometrium were evaluated in the rhesus monkey16. In this study, 25 mature females were assigned to treatment with intravaginal rings delivering progesterone (235 or 1770 μg/day), norethisterone (39 or 340 μg/day), or placebo for 52 weeks. At end of treatment, suppression of ovarian function was observed in treated groups. The endometrium was atrophic in all monkeys in the high-dose progesterone and norethisterone groups16.

The effect of progesterone delivered by an intrauterine system (IUS) on endometrial transformation and fertility was studied in the female rabbit18. An IUS delivering progesterone (10, 65, or 150 μg per day) was implanted in one uterine horn while the contralateral horn served as a control. The endometrium in both horns exhibited progestational effects in the 65- or 150-μg/day groups. Ovulation occurred in all subjects except those in the 65-μg/day group. Fertility returned quickly in all animals after IUS removal18.

Secondary pharmacodynamics

Pregnant female rats were treated with progesterone (1.5 mg/kg subcutaneous) or vehicle control from day 8 to day 21 of gestation, a dose comparable to a human dose of 100 mg/day19. Prenatal exposure to progesterone had no significant effect on reproductive success, neonatal mortality, and physical maturation. Progesterone-treated offspring weighed less than control animals initially (p < 0.05), a difference that disappeared by days 37–38. Brain DNA content was significantly greater among progesterone-treated offspring (p < 0.02, Student’s t-test). The only statistically significant behavioral difference detected was a reduction in mean head-lifting responses on the open field test among progesterone-treated offspring (p < 0.01, test for trend). It was concluded that prenatal exposure to progesterone had no consistent effects on the central nervous system in male rats19.

Pharmacokinetics and bioavailability

The pharmacokinetics of exogenous progesterone has been explored in a few studies in animal models.

In female ovariectomized rats treated with subcutaneous capsules containing progesterone (20, 40, 110, or 220 mg of crystalline progesterone), serum levels increased and then declined, reaching a steady-state value from day 7 to day 24 after capsule insertion17.

In dogs, progesterone bioavailability (relative to intravenous dosing) was significantly higher following administration of 25 mg of progesterone by vaginal tablet versus vaginal suppository20. In six female dogs, a single progesterone dose of 2 mg/kg, given by intramuscular injection, resulted in a progesterone mean (± standard deviation) serum Cmax of 34.3 (± 7.8) ng/ml at 1.8 (± 0.2) h after dosing; serum progesterone declined to pre-administration values (0.9 (± 0.2) ng/ml) by 72 h after the dose21.

Absorption, distribution, metabolism and excretion

In ovariectomized female rats, administration of a single dose of progesterone, 500 μg/kg intravenously, was followed by widespread distribution and rapid elimination; the mean (± standard deviation) distribution and elimination half-lives (t1/2) were 0.13 (± 0.024) and 1.21 (± 0.21) h, respectively. At steady state, the volume of distribution was 2.36 (± 0.23) l/kg. Elimination of the steroid was rapid with a total clearance of 2.75 (± 0.42) l/h·kg22.

In adult female rats in proestrus, 3H-progesterone was administered by gastric intubation, subcutaneous injection, or uterine intraluminal instillation. Absorption by the endometrium was rapid, and progesterone was eliminated from uterine tissue in a biphasic pattern with half-lives of 6.5 and 230 min for the α and β phases23.

Following intravenous injection of 3H-progesterone to non-pregnant monkeys, the labeled hormone disappeared from the circulation by 3 h after administration; between 0.5 and 1.75 h after 3H-progesterone disappeared, it reappeared in the circulation, reaching a maximum mean value of 20% of the initial Cmax. Progesterone metabolites also reappeared, suggesting that the parent compound was released from tissue stores since progesterone does not undergo enterohepatic recirculation24.

After oral administration, progesterone is approximately 96–99% bound to serum proteins, primarily to serum albumin and corticosteroid binding globulin6,11. Progesterone is metabolized primarily by the liver, largely to pregnanediols and pregnanolones6. Pregnanediols and pregnanolones are conjugated in the liver to glucuronide and sulfate metabolites. Progesterone undergoes renal and biliary elimination. Following injection of labeled progesterone, 50–60% of the excretion of metabolites occurs via the kidney; approximately 10% occurs via the bile and feces. Overall recovery of the labeled material accounts for 70% of an administered dose. Only a small portion of unchanged progesterone is excreted in the bile6,11.

Toxicology

A vaginal progesterone gel10 and a vaginal progesterone insert11 are currently approved for clinical use and a progesterone vaginal ring is approved in a few countries in Central and Latin America25,26. As progesterone is a physiological hormone, few preclinical studies have been conducted with these formulations and one preclinical toxicity study has been conducted with a progesterone vaginal ring16. Several reports issued by various governmental agencies or published literature have evaluated the carcinogenic and mutagenic potential of progesterone as well as its potential to cause reproductive toxicity16,2730.

In a review of the reproductive and developmental toxicity of progesterone developed for the California Environmental Protection Agency, no clear relationship had been demonstrated between exogenous progesterone and developmental or reproductive toxicity in humans30.

Acute toxicity

In mice, the median lethal dose (LD50) of progesterone, given by intravenous injection, is 100 mg/kg; in rats the LD50 is 327 mg/kg when the hormone is given by intraperitoneal injection. In rabbits, the LD50 was 26.5 mg/kg of body weight30,31.

Chronic toxicity

In a study in the rhesus monkey, 25 mature females were assigned to treatment with intravaginal rings delivering progesterone (235 or 1770 μg/day), norethisterone (39 or 340 μg/day), or placebo for 52 weeks16. Body weight gain and food and water consumption were not affected by treatment. Vaginal ring insertion was associated with a change in the balance of vaginal microflora in both active and control groups. The progesterone and norethisterone groups experienced a reduction in acid-tolerant bacteria; the balance was restored in all groups except the high-dose norethisterone group by the end of the study; no increase in the incidence of Candida species was observed in any group. A significant increase in fibrinogen in the high-dose norethisterone group versus the control group was the only hematologic change (253.4 ± 248.8 vs. 184.6 ± 184.6 mg%; p < 0.01)16.

Carcinogenicity studies

The International Agency for Research on Cancer (IARC) reviewed evidence on the carcinogenicity of progesterone and designated progesterone as ‘possibly carcinogenic to humans’ (Group 2B), although evidence for carcinogenicity to humans was considered inadequate27. The US National Toxicology Program (NTP) has classified progesterone as an agent ‘reasonably anticipated to be a human carcinogen’ since 1985; this classification is based primarily on evidence from the IARC reports28. Neither the IARC nor NTP has found sufficient evidence to conclude that progesterone exposure is linked to the development of cancer in humans.

Newborn female rats receiving progesterone subcutaneous injections and subsequent treatment with a carcinogen DMBA (7,12-dimethylbenz(a)anthracene) had an increased incidence of mammary adenocarcinoma as compared with control animals31.

In mice treated with subcutaneous implants releasing 59–900 μg of progesterone per day for 18 months, ovarian granulosa cell tumors were found in 27 of 83 treated mice and in one of 33 controls; the tumors were small (< 0.5 mm in diameter). In a similar study in mice treated with subcutaneous implants releasing 18–900 μg/day of progesterone for 18 months, uterine sarcomas were found in 15 of 142 treated mice and no control mice at the end of the study32.

As noted in the earliest IARC review32, when combined with known carcinogens (chiefly polycyclic aromatic hydrocarbons), progesterone influenced the incidence and histological type of mammary, uterine, and vaginal tumors in mice, rats, and rabbits32. An increase in tumor incidence was only observed when progesterone was given after, but not before, the carcinogen32. A review of rodent studies noted that, in the rat, progesterone increases the frequency of mammary tumors induced by DMBA or MCA (3,4-benzopyrene, 3-methylcholanthrene) in intact and ovariectomized animals but not in ovariectomized-adrenalectomized animals33. Further, chronic administration of progesterone to neonatally androgenized rats at various times after DMBA increased the frequency and growth of mammary carcinoma but moderate-to-high doses of progesterone plus high doses of estrogen inhibited mammary carcinoma in this model33.

In beagle dogs, long-term (74 weeks) subcutaneous progesterone injections in doses of 0.8–22.5 mg/day caused endometrial hyperplasia; no tumors were found on necropsy after the last dose, but fibroadenomatous nodules were found in two of five dogs receiving the highest dose.

In 52 rabbits given subcutaneous injections of progesterone 10 mg twice-weekly and then exposed to vaginal strings containing MCA, vaginal tumor incidence was similar to that of controls after 20 months (4/30 vs. 5/23)32.

Progesterone or medroxyprogesterone acetate (MPA) appears to have different potentials for carcinogenicity. In virgin female BALB/c mice, the incidence of mammary tumors was lower in those treated with progesterone in silastic subcutaneous pellets than in those treated with MPA, given by the same route at the same dose. In the progesterone-treated group, 67% of tumors were lobular and 33% were ductal; the proportions were reversed in the MPA-treated group. The ductal tumors were estrogen receptor-positive and PR-positive, while the lobular tumors were not, suggesting different mechanisms for carcinogenesis for the two progestins34.

Several studies in nulliparous, intact female rats have demonstrated that a combination of an estrogen and progesterone may be protective against the induction of mammary cancer by N-methyl-N-nitrosourea35,36. This effect mimics the protective effect of early pregnancy on subsequent development of mammary tumors.

Genotoxicity

In the Salmonella mutagenicity assay (Ames test), progesterone was not mutagenic37. In the mouse lymphoma assay, evidence for mutagenic activity was difficult to interpret38. Progesterone did not induce dominant lethal mutations in mice or chromosomal aberrations in rats treated in vivo and did not cause chromosomal aberrations in human or rodent cells or sister chromatid exchanges in cultured human cells. Studies on transformation of rodent cells in vitro were inconclusive: a clearly positive result was obtained for rat embryo, but results were weakly positive and negative in mouse and Syrian hamster cells, respectively27.

Reproductive and developmental toxicity

The offspring of pregnant Wistar rats injected with progesterone (1.5 mg/kg), between gestation days 8 and 21, weighed less at weaning and showed reduced performance on one measure of open field exploration and a slight increase in brain DNA as compared with controls; the effects on brain development were not considered to be drug-related39.

In pregnant Wistar rats given progesterone 0.1, 1, 5, or 20 mg/kg by intramuscular injection at various times during gestation, the two lowest doses were not associated with effects on fetal or placenta weights, although the frequency of early resorptions increased in rats given 1 mg/kg on gestation days 1–6 and 2.5% of the surviving fetuses had a morphological defect40. The highest dose, 20 mg/kg dose, given on gestation days 1–6, led to intrauterine death of all fetuses40, while the 5 mg/kg dose led to reduced birth weights and placental weights and to an increase in fetal defects (3.0%) in surviving fetuses40.

Effect of prenatal exposure on sexual development

The effects of prenatal progesterone on female sexual development have been studied in rats40, rabbits41, mice42, rhesus monkeys43,44 and guinea pigs45. In mice, the female offspring of pregnant mice treated with 0.25 mg or 0.5 mg subcutaneous progesterone, given on gestation days 12–16 had increased anogenital distances compared with control42. When the progesterone-treated female offspring were mated, fertility and pregnancy outcome were unaffected but an increase in aggressive behavior toward male mice was observed during lactation42.

In a study in rhesus and cynomolgus monkeys, intramuscular progesterone plus estradiol benzoate were given from gestation days 20 to 50 at 0.1–25 times the human dose equivalent (HDE). Embryo lethality was observed at doses 10 and 25 times the HDE; one dead female fetus (progesterone dose: 10 times HDE) exhibited masculinized external genitalia43.

The effects of prenatal exposure to exogenous progesterone on male sexual development have not been studied extensively in animal models. A change in anogenital distance in male, as well as female, offspring of rabbits exposed to prenatal progesterone was reported41 but no ‘feminization’ was reported in three male monkey infants exposed to progesterone44.

In one study, pregnant female mice were given progesterone 2 mg/day by subcutaneous injection on gestational days 14–16. Anogenital distance and body weight were unaffected by progesterone exposure. At 90 days, however, male mice exposed to prenatal progesterone showed deficiencies in mating behavior, marked by failure to achieve mounting or intromission46.

Progesterone as a neurosteroid and preclinical studies in the brain

More recently, most preclinical work was geared at identifying the unique role of progesterone on neurogenesis and brain effects.

Progesterone receptors have been identified in several areas of the brain47 and have been implicated in a variety of functions, including cognition48, neuroprotection49,50, and dendritic remodeling51. Progesterone reduces the incidence of epileptic activity both directly52 and through conversion to allopregnanolone53. In addition, progesterone has neuroprotective effects after ischemic or traumatic brain injury, both alone and after estrogen priming54.

Despite positive preclinical studies, failure of clinical trials of traumatic brain injury were discouraging. However, extrapolation from pre-clinical data to humans needs careful dose-finding, and suboptimal dosing was suggested as a factor of failure50.

In preclinical studies, progesterone and some structurally related progestins have shown neuroprotective properties mediated via the PR5558.

Preclinical studies showed that progesterone and Nestorone, with or without estradiol, stimulate oligodendrocytes and myelin repair in both in vitro55 and in vivo56 experiments and decrease the severity of the disease induced in experimental autoimmune encephalomyelitis57, an animal model for multiple sclerosis, with consistent responses in the same dose range. Progesterone and Nestorone were shown to promote myelin repair by binding to PRs, thereby stimulating recruitment and maturation of oligodendrocyte progenitor cells. In contrast, other commonly used synthetic progestins such as levonorgestrel and medroxyprogesterone acetate have potential adverse outcomes on the brain’s regenerative capacity55,58.

Brinton’s laboratory58 reported the efficacy of different progestins used alone or in combination with 17β-estradiol on adult rat neural progenitor cell (rNPC) proliferation and hippocampal cell viability. In vitro analyses indicated that progesterone, norgestimate, Nestorone, norethynodrel, norethisterone, and levonorgestrel significantly increased rNPC proliferation, whereas norethisterone acetate was without effect, and MPA inhibited rNPC proliferation. Proliferative progestins in vitro were also neuroprotective. Acute in vivo exposure to progesterone and Nestorone significantly increased proliferating cells.

Experimental studies also showed progesterone efficiency in reducing infarct volume and improving functional recovery in a model of transient middle cerebral artery occlusion5961.

Allopregnanolone may also protect the brain against ischemic damage by other signaling mechanisms not involving PRs62. Liu and colleagues63 have shown that Nestorone was also effective in that model. PRs are mediators of neuroprotection, as progesterone is not effective in PRKO mice63. Also, progesterone and Nestorone were less efficient in heterozygous mice, expressing reduced levels of PRs56,63.

In addition, Brinton’s laboratory showed the efficacy of allopregnanolone as a regenerative agent for Alzheimer’s disease and with potential to treat multiple neurological disorders6466.

Conclusion

Progesterone is a physiological hormone to which women are exposed at high levels during pregnancy. The various formulations developed for therapy, either oral or from other delivery systems, had few preclinical studies conducted but the clinical data support the benefits of this therapy in the approved indications.

It is possible that progesterone and related molecules may also confer novel therapeutic benefits in the nervous system for the treatment of stroke, brain injury, myelin repair and Alzheimer’s disease.

Footnotes

Conflict of interest The author reports no conflict of interest. The author alone is responsible for the content and writing of this paper.

References

  • 1.Conneely OM, Lydon JP. Progesterone receptors in reproduction: functional impact of the A and B isoforms. Steroids 2000;65:571–7 [DOI] [PubMed] [Google Scholar]
  • 2.Conneely OM, Mulac-Jericevic B, Lydon JP, et al. Reproductive functions of the progesterone receptor isoforms: lessons from knock-out mice. Mol Cell Endocrinol 2001;179:97–103 [DOI] [PubMed] [Google Scholar]
  • 3.Conneely OM, Mulac-Jericevic B, Lydon JP. Progesterone-dependent regulation of female reproductive activity by two distinct progesterone receptor isoforms. Steroids 2003;68:771–8 [DOI] [PubMed] [Google Scholar]
  • 4.Nutu M, Weijdegard B, Thomas P, et al. Membrane progesterone receptor gamma: tissue distribution and expression in ciliated cells in the fallopian tube. Mol Reprod Dev 2007;74:843–50 [DOI] [PubMed] [Google Scholar]
  • 5.Hochner-Celnikier D, Marandici A, Iohan F, et al. Estrogen and progesterone receptors in the organs of prenatal cynomolgus monkey and laboratory mouse. Biol Reprod 1986;35:633–40 [DOI] [PubMed] [Google Scholar]
  • 6.Stanczyk FZ, Hapgood JP, Winer S, et al. Progestogens used in postmenopausal hormone therapy: differences in their pharmacological properties, intracellular actions, and clinical effects. Endocr Rev 2013;34:171–208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Noé G, Sitruk-Ware R, Zegers-Hochschild F, et al. Endometrial effect of progesterone delivered by vaginal rings in estrogen-treated postmenopausal women. Climacteric 2010;13:433–41 [DOI] [PubMed] [Google Scholar]
  • 8.Nath A, Sitruk-Ware R. Parenteral administration of progestins for hormonal replacement therapy. Eur J Contracept Reprod Health Care 2009;14:88–96 [DOI] [PubMed] [Google Scholar]
  • 9.Massai R, Quinteros E, Reyes MV, et al. Extended use of a progesterone-releasing vaginal ring in nursing women: a phase II clinical trial. Contraception 2005;72:352–7 [DOI] [PubMed] [Google Scholar]
  • 10.Crinone® 4% and Crinone® 8% (progesterone gel) [package insert]. https://www.allergan.com/assets/pdf/crinone_pi
  • 11.Endometrin® (progesterone) Vaginal Insert [package insert]. https://www.drugs.com/pro/endometrin.html
  • 12.Thind KK, Goldsmith PC. Expression of estrogen and progesterone receptors in glutamate and GABA neurons of the pubertal female monkey hypothalamus. Neuroendocrinology 1997;65:314–24 [DOI] [PubMed] [Google Scholar]
  • 13.Szabo M, Kilen SM, Nho SJ, et al. Progesterone receptor A and B messenger ribonucleic acid levels in the anterior pituitary of rats are regulated by estrogen. Biol Reprod 2000;62:95–102 [DOI] [PubMed] [Google Scholar]
  • 14.Heikinheimo O, Mahony MC, Gordon K, et al. Estrogen and progesterone receptor mRNA are expressed in distinct pattern in male primate reproductive organs. J Assist Reprod Genet 1995;12:198–204 [DOI] [PubMed] [Google Scholar]
  • 15.Luetjens CM, Didolkar A, Kliesch S, et al. Tissue expression of the nuclear progesterone receptor in male non-human primates and men. J Endocrinol 2006;189:529–39 [DOI] [PubMed] [Google Scholar]
  • 16.Wadsworth PF, Heywood R, Allen DG, et al. Treatment of rhesus monkeys (Macaca mulatta) with intravaginal rings impregnated with either progesterone or norethisterone. Contraception 1979;20:339–51 [DOI] [PubMed] [Google Scholar]
  • 17.Mannino CA, South SM, Inturrisi CE, et al. Pharmacokinetics and effects of 17beta-estradiol and progesterone implants in ovariectomized rats. J Pain 2005;6:809–16 [DOI] [PubMed] [Google Scholar]
  • 18.Hudson R, Hempill P, Tillson SA. Preclinical evaluation of intrauterine progesterone as a contraceptive agent. I. Local contraceptive effects and their reversal. Contraception 1978;17:465–74 [DOI] [PubMed] [Google Scholar]
  • 19.Coyle IR, Anker R, Cragg B. Behavioral, biochemical and histological effects of prenatal administration of progesterone in the rat. Pharmacol Biochem Behav 1976;5:587–90 [DOI] [PubMed] [Google Scholar]
  • 20.Fulper LD, Cleary RW, Harland EC, et al. Comparison of serum progesterone levels in dogs after administration of progesterone by vaginal tablet and vaginal suppositories. Am J Obstet Gynecol 1987;156:253–6 [DOI] [PubMed] [Google Scholar]
  • 21.Scott-Moncrieff JC, Nelson RW, Bill RL, et al. Serum disposition of exogenous progesterone after intramuscular administration in bitches. Am J Vet Res 1990;51:893–5 [PubMed] [Google Scholar]
  • 22.Gangrade NK, Boudinot FD, Price JC. Pharmacokinetics of progesterone in ovariectomized rats after single dose intravenous administration. Biopharm Drug Dispos 1992;13:703–9 [DOI] [PubMed] [Google Scholar]
  • 23.Fang SM, Lin CS, Lyon V. Progesterone retention by rat uterus. I. Pharmacokinetics after uterine intraluminal instillation. J Pharm Sci 1977;66:1744–8 [DOI] [PubMed] [Google Scholar]
  • 24.Kowalski WB, Chatterton RT, Jr, Kazer RR, et al. Disappearance and unexpected reappearance of progesterone in the circulation of the monkey: novel hormone kinetics. J Physiol 1996;493:877–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sitruk-Ware R, Ramarao S, Merkatz R, et al. Risk of pregnancy in breastfeeding mothers: role of the progesterone vaginal ring on birth spacing. EMJ Reprod Health 2016;2:66–72 [Google Scholar]
  • 26.Zegers-Hochschild F, Balmaceda JP, Fabres C, et al. Prospective randomized trial to evaluate the efficacy of a vaginal ring releasing progesterone for IVF and oocyte donation. Hum Reprod 2000;15:2093–7 [DOI] [PubMed] [Google Scholar]
  • 27.International Agency for Research on Cancer (IARC). IARC Monographs on the Evaluations of Carcinogenic Risk of Chemicals to Man, Supplement 7, Overall evaluation of carcinogenicity: an update of IARC monographs volumes 1 to 42: progestins, combined oral contraceptives. Report No: Supplement 7. International Agency for Research on Cancer (IARC): 1987, 289 [Google Scholar]
  • 28.National Toxicology Program. Report on Carcinogens. Report No: Eleventh Edition United States Department of Health and Human Services: 2005 [Google Scholar]
  • 29.Progestational drug products for human use; requirements for labeling directed to the patient. Fed Regist 1999;64:62110–12 [PubMed] [Google Scholar]
  • 30.Golub MS, Kaufman FL, Campbell MA, et al. Evidence on the developmental and reproductive toxicity of progesterone Draft. Reproductive and Cancer Hazard Assessment Section. Office of Environmental Health Hazard Assessment, California: Environmental Protection Agency: August 2004 [Google Scholar]
  • 31.International Agency for Research on Cancer (IARC). IARC Monographs on the Evaluations of Carcinogenic Risk of Chemicals to Man, Progesterone: Summary of data reported and evaluation. Report No: 21. International Agency for Research on Cancer (IARC): 1979, 135 [Google Scholar]
  • 32.International Agency for Research on Cancer (IARC ). IARC Monographs on the Evaluations of Carcinogenic Risk of Chemicals to Man, Progesterone: Summary of data reported and evaluation. Report No: 6. International Agency for Research on Cancer (IARC): 1974, 135 [Google Scholar]
  • 33.Russo IH, Russo J. Mammary gland neoplasia in long-term rodent studies. Environ Health Perspect 1996;104:938–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kordon EC, Molinolo AA, Pasqualini CD, et al. Progesterone induction of mammary carcinomas in BALB/c female mice. Correlation between progestin dependence and morphology. Breast Cancer Res Treat 1993;28:29–39 [DOI] [PubMed] [Google Scholar]
  • 35.Rajkumar L, Guzman RC, Yang J, et al. Prevention of mammary carcinogenesis by short-term estrogen and progestin treatments. Breast Cancer Res 2004;6:R31–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Guzman RC, Yang J, Rajkumar L, et al. Hormonal prevention of breast cancer: mimicking the protective effect of pregnancy. Proc Natl Acad Sci U S A 1999;96:2520–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Dunkel VC, Zeiger E, Brusick D, et al. Reproducibility of microbial mutagenicity assays. II. Testing of carcinogens and noncarcinogens in Salmonella typhimurium and Escherichia coli. Environ Mutagen 1985;7(Suppl 5):1–248 [DOI] [PubMed] [Google Scholar]
  • 38.Myhr BC, Caspary WJ. Evaluation of the L5178Y mouse lymphoma cell mutagenesis assay: intra-laboratory results for sixty-three coded chemicals tested at Litton Bionetics, Inc. Environ Mol Mutagen 1988;12(Suppl 13):103–94 [DOI] [PubMed] [Google Scholar]
  • 39.Coyle IR, Anker R, Cragg B. Behavioral, biochemical and histological effects of prenatal administration of progesterone in the rat. Pharmacol Biochem Behav 1976;5:587–90 [DOI] [PubMed] [Google Scholar]
  • 40.Piotrowski J The effect of progesterone on the foetal development of rats on the Wistar strain. Part 3. Folia Biol (Krakow) 1968;16:343–53 [PubMed] [Google Scholar]
  • 41.Piotrowski J Experimental investigations on the effect of progesterone on embryonal development. Part II. Investigations carried out on rabbits. Folia Biol (Krakow) 1968;16:335–42 [PubMed] [Google Scholar]
  • 42.Wagner CK, Kinsley C, Svare B. Mice: postpartum aggression is elevated following prenatal progesterone exposure. Horm Behav 1986;20:212–21 [DOI] [PubMed] [Google Scholar]
  • 43.Hendrickx AG, Korte R, Leuschner F, et al. Embryotoxicity of sex steroidal hormone combinations in nonhuman primates. I. Norethisterone acetate + ethinylestradiol and progesterone + estradiol benzoate (Macaca mulatta, Macaca fascicularis, and Papio cynocephalus). Teratology 1987;35:119–27 [DOI] [PubMed] [Google Scholar]
  • 44.Hendrickx AG, Korte R, Leuschner F, et al. Embryotoxicity of sex steroidal hormones in nonhuman primates. II. Hydroxyprogesterone caproate, estradiol valerate. Teratology 1987;35:129–36 [DOI] [PubMed] [Google Scholar]
  • 45.Foote WD, Foote WC, Foote LH. Influence of certain natural and synthetic steroids on genital development in guinea pigs. Fertil Steril 1968;19:606–15 [DOI] [PubMed] [Google Scholar]
  • 46.Pointis G, Latreille MT, Richard MO, et al. Effect of natural progesterone treatment during pregnancy on fetal testosterone and sexual behavior of the male offspring in the mouse. Dev Pharmacol Ther 1987;10:385–92 [DOI] [PubMed] [Google Scholar]
  • 47.MacLusky NJ and McEwen BS. Oestrogen modulates progestin receptor concentrations in some rat brain regions but not in others. Nature 1978;274:276–8 [DOI] [PubMed] [Google Scholar]
  • 48.Sandstrom NJ, Williams CL. Memory retention is modulated by acute estradiol and progesterone replacement. Behav Neurosci 2001;115:384–93 [PubMed] [Google Scholar]
  • 49.Robertson CL, Puskar A, Hoffman GE, et al. Physiologic progesterone reduces mitochondrial dysfunction and hippocampal cell loss after traumatic brain injury in female rats. Exp Neurol 2006;197:235–43 [DOI] [PubMed] [Google Scholar]
  • 50.Howard RB, Sayeed I, Stein DG. Suboptimal dosing parameters as possible factors in the negative phase III clinical trials of progesterone for traumatic brain injury. J Neurotrauma 2017;34:1915–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Woolley CS, McEwen BS. Roles of estradiol and progesterone in regulation of hippocampal dendritic spine density during the estrous cycle in the rat. J Comp Neurol 1993;336:293–306 [DOI] [PubMed] [Google Scholar]
  • 52.Edwards HE, Epps T, Carlen PL, et al. Progestin receptors mediate progesterone suppression of epileptiform activity in tetanized hippocampal slices in vitro. Neuroscience 2000;101:895–906 [DOI] [PubMed] [Google Scholar]
  • 53.Frye CA, Rhodes ME. Estrogen-priming can enhance progesterone’s anti-seizure effects in part by increasing hippocampal levels of allopregnanolone. Pharmacol Biochem Behav 2005;81:907–16 [DOI] [PubMed] [Google Scholar]
  • 54.Schumacher M, Guennoun R, Ghoumari A, et al. Novel perspectives for progesterone in hormone replacement therapy, with special reference to the nervous system. Endocr Rev 2007;28:387–439 [DOI] [PubMed] [Google Scholar]
  • 55.Hussain R, El-Etr M, Gaci O, et al. Progesterone and Nestorone facilitate axon remyelination: a role for progesterone receptors. Endocrinology 2011;152:3820–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.El-Etr M, Rame M, Boucher C, et al. Progesterone and Nestorone promote myelin regeneration in chronic demyelinating lesions of corpus callosum and cerebral cortex. Glia 2015;63:104–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Garay L, Gonzalez Deniselle MC, Sitruk-Ware R, et al. Efficacy of the selective progesterone receptor agonist Nestorone for chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 2014;276:89–97 [DOI] [PubMed] [Google Scholar]
  • 58.Liu L, Zhao L, She H, et al. Clinical relevant progestins regulate neurogenic and neuroprotective responses in vitro and in vivo. Endocrinology 2010;151:5782–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Gibson CL, Murphy SP. Progesterone enhances functional recovery after middle cerebral artery occlusion in male mice. J Cereb Blood Flow Metab 2004;24:805–13 [DOI] [PubMed] [Google Scholar]
  • 60.Sayeed I, Wali B, Stein DG. Progesterone inhibits ischemic brain injury in a rat model of permanent middle cerebral artery occlusion. Restor Neurol Neurosci 2007;25:15–9 [PubMed] [Google Scholar]
  • 61.Gibson CL, Coomber B, Murphy SP. Progesterone is neuroprotective following cerebral ischaemia in reproductively ageing female mice. Brain 2011;134:2125–33 [DOI] [PubMed] [Google Scholar]
  • 62.Sayeed I, Guo Q, Hoffman SW, et al. Allopregnanolone, a progesterone metabolite, is more effective than progesterone in reducing cortical infarct volume after transient middle cerebral artery occlusion. Ann Emerg Med 2006;47:381–9 [DOI] [PubMed] [Google Scholar]
  • 63.Liu A, Margaill I, Zhang S, et al. Progesterone receptors: a key for neuroprotection in experimental stroke. Endocrinology 2012;153:3747–57 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Irwin RW, Solinsky CM, Loya CM, et al. Allopregnanolone preclinical acute pharmacokinetic and pharmacodynamic studies to predict tolerability and efficacy for Alzheimer’s disease. PLoS One 2015;10:1–31 - e0128313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Irwin RW, Solinsky CM, Brinton RD. Frontiers in therapeutic development of allopregnanolone for Alzheimer’s disease and other neurological disorders. Front Cell Neurosci 2014;8:2–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Irwin RW, Brinton RD. Allopregnanolone as regenerative therapeutic for Alzheimer’s disease: translational development and clinical promise. Prog Neurobiol 2014;113:40–55 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES