133
Views
10
CrossRef citations to date
0
Altmetric
Reviews

The role of progesterone and the progesterone receptor in human reproduction and cancer

&
Pages 469-484 | Published online: 10 Jan 2014

References

  • Corner GW, Allen WM. Physiology of the corpus luteum. 1929. Am. J. Obstet. Gynecol. 193, 1574; discussion 1575 (2005).
  • Jones GS. Some newer aspects of management of infertility. JAMA 141, 1123–1129 (1949).
  • Baulieu EE. Contragestion and other clinical applications of RU-486, an antiprogesterone at the receptor. Science 245, 1351–1357 (1989).
  • Noyes RW, Hertig AI, Rock J. Dating the endometrial biopsy. Fertil. Steril. 1, 3–35 (1950).
  • Meyer WR, Castelbaum AJ, Somkuti Set al. Hydrosalpinges adversely affect markers of endometrial receptivity. Hum. Reprod. 12, 1393–1398 (1997).
  • Savaris RF, Pedrini JL, Flores R, Fabris G, Zettler CG. Expression of alpha 1 and beta 3 integrins subunits in the endometrium of patients with tubal phimosis or hydrosalpinx. Fertil. Steril. 85, 188–192 (2006).
  • Daftary GS, Kayisli U, Seli E, Bukulmez O, Arici A, Taylor HS. Salpingectomy increases peri-implantation endometrial HOXA-10 expression in women with hydrosalpinx. Fertil. Steril. 87, 367–372 (2007).
  • Yashinaga K. Uterine receptivity for blastocyst implantation. Ann. N. Y. Acad. Sci. 541, 424–431 (1988).
  • Lessey BA. Assessment of endometrial receptivity. Fertil. Steril. 96, 522–529 (2011).
  • Aplin JD. MUC-1 glycosylation in endometrium: possible roles of the apical glycocalyx at implantation. Hum. Reprod. 14(Suppl. 2), 17–25 (1999).
  • De Souza MM, Surveyor GA, Price REet al. MUC1/episialin: a critical barrier in the female reproductive tract. J. Reprod. Immunol. 45, 127–158 (1999).
  • Surveryor GA, Gendler SJ, Pemberton Let al. Expression and steroid hormonal control of Muc-1 in the mouse uterus. Endocrinology 136, 3639–3647 (1995).
  • Margarit L, Gonzalez D, Lewis PDet al. L-selectin ligands in human endometrium: comparison of fertile and infertile subjects. Hum. Reprod. 24, 2767–2777 (2009).
  • Wang B, Sheng JZ, He RH, Qian YL, Jin F, Huang HF. High expression of L-selectin ligand in secretory endometrium is associated with better endometrial receptivity and facilitates embryo implantation in human being. Am. J. Reprod. Immunol. 60, 127–134 (2008).
  • Fukuda MN, Sato T, Nakayama Jet al. Trophinin and tastin, a novel cell adhesion molecule complex with potential involvement in embryo implantation. Genes. Dev. 9, 1199–1210 (1995).
  • Albelda SM, Buck CA. Integrins and other cell adhesion molecules. FASEB J. 4, 2868–2880 (1990).
  • Achache H, Revel A. Endometrial receptivity markers, the journey to successful embryo implantation. Hum. Reprod. Update 12, 731–746 (2006).
  • Creus M, Balasch J, Ordi Jet al. Integrin expression in normal and out-of-phase endometria. Hum. Reprod. 13, 3460–3468 (1998).
  • Surrey ES, Lietz AK, Gustofson RL, Minjarez DA, Schoolcraft WB. Does endometrial integrin expression in endometriosis patients predict enhanced in vitro fertilization cycle outcomes after prolonged GnRH agonist therapy. Fertil. Steril. 93, 646–651 (2010).
  • Enders AC, Nelson DM. Pinocytotic activity of the uterus of the rat. Am. J. Anat. 138, 277–299 (1973).
  • Quinn CE, Ryan E, Claessens EAet al. The presence of pinopodes in the human endometrium does not delineate the implantation window. Fertil. Steril. 87, 1015–1021 (2007).
  • Usadi RS, Groll JM, Lessey BAet al. Endometrial development and function in experimentally induced luteal phase deficiency. J. Clin. Endocrinol. Metab. 93, 4058–4064 (2008).
  • Matsuzaki S, Canis M, Darcha C, Pouly JL, Mage G. HOXA-10 expression in the mid-secretory endometrium of infertile patients with either endometriosis, uterine fibromas or unexplained infertility. Hum. Reprod. 24, 3180–3187 (2009).
  • Szczepanska M, Wirstiein P, Luczak M, Jagodzinski PP, Skrzypczak J. Reduced expression of HOXA-10 in the midluteal endometrium from infertile women with minimal endometriosis. Biomed. Pharmacother. 64, 697–705 (2010).
  • Cakmak H, Taylor HS. Molecule mechanisms of treatment resistance in endometriosis: the role of progesterone-hox gene interactions. Semin. Reprod. Med. 28, 69–74 (2010).
  • Young SL, Lessey BA, Balthazar Uet al. Defining the relationship between progesterone dose, endometrial histology and gene expression using an in vivo luteal phase defect model. Reprod. Sci. 18, 273A (2011).
  • Dominguez F, Garrido-Gomez T, Lopez JAet al. Proteomic analysis of the human receptive versus non-receptive endometrium using differential in-gel electrophoresis and MALDI-MS unveils stathmin 1 and annexin A2 as differentially regulated. Hum. Reprod. 24, 2607–2617 (2009).
  • Parmar T, Gadkar-Sable S, Savardekar Iet al. Protein profiling of human endometrial tissues in the midsecretory and proliferative phases of the menstrual cycle. Fertil. Steril. 92, 1091–1103 (2009).
  • DeSouza L, Diehl G, Yang ECet al. Proteomic analysis of the proliferative and secretory phases of the human endometrium: protein identification and differential protein expression. Proteomics 5, 270–281 (2005).
  • Wetendorf M, DeMayo FJ. The progesterone receptor regulates implantation, decidualization, and glandular development via a complex paracrine signaling network. Mol. Cell Endocrinol. 357, 108–118 (2012).
  • Lachmann M, Gelbmann D, Kalman Eet al. PIBF (progesterone-induced blocking factor) is overexpressed in highly proliferating cells and associated with the centrosome. Int. J. Cancer 112, 51–60 (2004).
  • Polgar B, Kispal GY, Lachmann Met al. Molecular cloning and immunological characterization of a novel cDNA coding for PIBF. J. Immunol. 171, 5956–5963 (2003).
  • Szekeres-Bartho J, Hadnagy J, Pacsa AS. The suppressive effect of progesterone on lymphocyte cytotoxicity: unique progesterone sensitivity of pregnancy lymphocytes. J. Reprod. Immunol. 7, 121–128 (1985).
  • Szekeres-Bartho J, Autran B, Debre P, Andreu G, Denver L, Chaouat G. Immunoregulatory effects of a suppressor factor from healthy pregnant women’s lymphocytes after progesterone induction. Cell Immunol. 122, 281–294 (1989).
  • Szekeres-Bartho J, Szekeres GY, Debre P, Autran B, Chaouat G. Reactivity of lymphocytes to a progesterone receptor-specific monoclonal antibody. Cell Immunl. 125, 273–283 (1990).
  • Polgar B, Barakony A, Xinos I, Szekeres-Bartho J. The role of γ/δ TCR positive cells in pregnancy. Am. J. Reprod. Immunol. 41, 239–244 (1999).
  • Szekeres-Bartho J, Barakonyi A, Polgar Bet al. The role of γ/δ T cells in progesterone-mediated immunomodulation during pregnancy: a review. Am. J. Reprod. Immunol. 42, 44–48 (1999).
  • Hansen KA, Opsahl MS, Nieman LK, Baker JR Jr, Klein TA. Natural killer cell activity from the pregnant subjects is modulated by RU 486. Am. J. Obstet. Gynecol. 166, 87–90 (1992).
  • Szekeres-Bartho J, Kilar F, Falkay G, Csernus V, Torok A, Pacsa AS. Progesterone-treated lymphocytes of healthy pregnant women release a factor inhibiting cytotoxicity and prostaglandin synthesis. Am. J. Reprod. Immunol Microbiol. 9, 15–18 (1985).
  • Ivanova-Todorova E, Kyurkchiev DS, Nalbanski A, Timeva T, Shterev A, Kyurkchiev SD. Production and characterization of a novel monoclonal antibody against progesterone-induced blocking factor (PIBF). J. Reprod. Immunol. 78, 94–101 (2008).
  • Szekeres-Bartho J, Weill BJ, Mike G, Houssin D, Chaouat G. Progesterone receptors in lymphocytes of liver-transplanted and transfused patients. Immunol. Lett. 22, 259–261 (1989).
  • Chiu L, Nishimura M, Ishi Yet al. Enhancement of the expression of progesterone receptor on progesterone-treated lymphocytes after immunotherapy in unexplained recurrent spontaneous abortion. Am. J. Reprod. Immunol. 35, 552–557 (1996).
  • Check JH, Arwitz M, Gross J, Peymer M, Szekeres-Bartho J. Lymphocyte immunotherapy (LI) increases serum levels of progesterone-induced blocking factor. Am. J. Reprod. Immunol. 37, 17–20 (1997).
  • Check JH, Szekeres-Bartho J, O'Shaughnessy A. Progesterone-induced blocking factor seen in pregnancy lymphocytes soon after implantation. Am. J. Reprod. Immunol. 35, 277–280 (1996).
  • Check JH, Arwitz M, Gross J, Szekeres-Bartho J, Wu CH. Evidence that the expression of progesterone-induced blocking factor by maternal T-lymphocytes is positively correlated with conception. Am. J. Reprod. Immunol. 38(1), 6–8 (1997).
  • Cohen R, Check JH, DiAntonio A, Srivastava MD. Progesterone-induced blocking factor (PIBF), an immunosuppressive protein that inhibits natural killer (NK) cell cytolytic activity, detected 3 days after embryo transfer (ET). Fertil. Steril. 98(Suppl. 3), S28 (2012).
  • Check JH, Cohen R, Jaffe A, Tran J, Sarumi M. An allogeneic stimulus is not a prerequisite for the expression of the immunomodulatory protein the progesterone-induced blocking factor (PIBF). Am. J. Reprod. Immunol, ISIR, Boston, MA (2013).
  • Check JH, Cohen R, DiAntonio A, Check D. The demonstration that the immunomodulatory protein the progesterone-induced blocking factor significantly rises in males with short term progesterone exposure provides new insights into the immunology of pregnancy. Fertil. Steril. 99(3), S22–S23 (2013).
  • Faust Z, Laskarin G, Rukavina Det al. Progesterone-induced blocking factor inhibits degranulation of NK cells. Am. J. Reprod. Immunol 42, 71 (1999).
  • Kozma N, Halasz M, Polgar Bet al. PIBF activates STAT6 via binding to a novel IL-4 receptor. J. Immunol. 176, 819–826 (2006).
  • Szekeres-Bartho J, Wegmann TG. A progesterone-dependent immuno-modulatory protein alters the Th1/Th2 balance. J. Reprod. Immunol. 31, 81–95 (1996).
  • Wegmann TG, Lin H, Guilbert L, Mosmann TR. Bidirectional cytokine interactions in the maternal-fetal relationship: is successful pregnancy a Th2 phenomenon? Immunol. Today 14, 353–356 (1993).
  • Raghupathy R. Th-1 type immunity is incompatible with successful pregnancy. Immunol. Today 18, 478–482 (1997).
  • Raghupathy R, Al Mutawa E, Makhseed Met al. Modulation of cytokine production by dydrogesterone in lymphocytes from women with recurrent abortion. BJOG 112, 1096–1101 (2005).
  • Raghupathy R, Al Mutawa E, Maksheed M, Azizieh F, Szekeres-Bartho J. Redirection of cytokine production by lymphocytes from women with pre-term delivery by dydrogesterone. Am. J. Reprod. Immunol 58, 31–38 (2007).
  • Raghupathy R, Makhseed M, Azizieh F, Omu A, Gupta M, FArhat R. Cytokine production by maternal lymphocytes during normal human pregnancy and in unexplained recurrent spontaneous abortion. Hum. Reprod. 15, 713–718 (2000).
  • Yao MW, Lim H, Schust DJet al. Gene expression profiling reveals progesterone-mediated cell cycle and immunoregulatory roles of HOXA-10 in the preimplantation uterus. Mol. Endocrol. 17, 610–627 (2003).
  • Sentman CL, Meadows SK, Wira CR, Eriksson M. Recruitment of uterine NK cells. Induction of CXC chemokine ligands 10 and 11 in human endometrium by estradiol and progesterone. J. Immunol. 173, 6760–6766 (2004).
  • Blois S, Ilarregui JM, Tometten Met al. A pivotal role for galectin-1 in fetal tolerance Nat. Med. 13, 1450–1457 (2007).
  • Thomas P. Characteristics of membrane progestin receptor alpha (mPRα) and progesterone membrane receptor component 1 IPGMRC1) and their roles in mediating rapid progestin actions. Front. Neuroendocrinol. 29, 292–312 (2008).
  • Migliaccio A, Piccolo D, Castoria Get al. Activation of the Src/p21ras/Erk pathway by progesterone receptor via cross-talk with estrogen receptor. EMBO J. 17, 2008–1018 (1998).
  • Chien EJ, Liao CF, Chang CPet al. The non-genomic effects on NA(+)/H(+)-exchange 1 by progesterone and 20 alpha-hydroxyprogesterone in human T cells. J. Cell Physiol. 211, 544–550 (2007).
  • Xu X, Weaver Z, Linke SPet al. Centrosome amplification and a defective G2-M cell cycle checkpoint induce genetic instability in BRCA1 exon 11 isoform-deficient cells. Mol. Cell 3, 389–395, (1999).
  • Fukasawa K, Choi T, Kuriyama R, Rulong S, Vande Woude GF. Abnormal centrosome amplification in the absence of p53. Science 271, 1744–1747, (1996).
  • Srivastava MD, Thomas A, Srivastava BI, Check JH. Expression and modulation of progesterone-induced blocking factor (PIBF) and innate immune factors in human leukemia cell lines by progesterone and mifepristone. Leuk. Lymphoma 48, 1610–1617, (2007).
  • Check JH, Nazari P, Goldberg J, Yuen W, Angotti D. A model for potential tumor immunotherapy based on knowledge of immune mechanisms responsible for spontaneous abortion. Med. Hypoth. 57, 337–343 (2001).
  • Check JH, Dix E, Sansoucie L. Support for the hypothesis that successful immunotherapy of various cancers can be achieved by inhibiting a progesterone associated immunomodulatory protein. Med. Hypoth. 72, 87–90 (2009).
  • Check JH, Cohen R, Rosenberg A, DiAntonio A, Sarumi M, Srivastava M. Evidence that the immunosuppressive effect of the progesterone-induced blocking factor (PIBF) inhibiting a host response against cancer cells is probably not by influencing gamma/delta T cells in the tumor microenvironment to secrete PIBF and thus inhibit natural killer cell activity and therefore is dissimilar to the pregnancy state. Presented at: ISIR 2013 Meeting, Boston, MA, USA 28 May-1 June 2013 abstract #P-13. Am. J. Reprod. Immunol. 69, 50–51 (2013).
  • Lange CA, Shen T, Horwicz KB. Phosphorylation of human progesterone receptors at serine-294 by mitogen-activated protein kinase signals their degradation by the 26S proteasome. Proc. Natl Acad. Sci. USA 97, 1032–1037 (2000).
  • Cui X, Schiff R, Arpino G, Osborne CK, Lee AV. Biology of progesterone receptor loss in breast cancer and its implications for endocrine therapy. J. Clin. Oncol. 23, 7721–7735 (2005).
  • Conneely OM, Jericevic BM, Lydon JP. Progesterone receptors in mammary gland development and tumorigenesis. J. Mammary Gland. Biol. Neoplasia. 8, 205–214 (2003).
  • Wei LI, Hawkins P, Baker C, Norris B, Sheridan LP, Quinn PG. An amino-terminal truncated progesterone receptor isoform, PRc, enhances progestin-induced transcriptional activity. Mol. Endocrinol. 10, 1379–1387 (1996).
  • Condon JC, Hardy DB, Kovaric K, Mendelson CR. Upregulation of the progesterone receptor (PR)-C isoform in laboring myometrium by activation of NK-&kappaB may contribute to the onset of labor through inhibition of PR function. Mol. Endocrinol. 20, 764–775 (2006).
  • Daniel AR, Hagan CR, Lange CA. Progesterone receptor action: defining a role in breast cancer. Expert Rev. 6, 359–369 (2011).
  • Friedman LS, Ostermeyer EA, Lynch EDet al. The search for BRCA1. Cancer Res. 54, 6374–6382 (1994).
  • Zheng L, Annab LA, Afshari CA, Lee WH, Boyer tG. BRCA1 mediates ligand-independent transcriptional repression of the estrogen receptor. Proc. Natl Acad. Sci. USA 98, 9587–9592 (2001).
  • Fan S, Wang J, Yuan Ret al. BRCA1 inhibition of estrogen receptor signaling in transfected cells. Science 284, 1354–1356 (1999).
  • Ma Y, Katiyar P, Jones LPet al. The breast cancer susceptibility gene BRCA1 regulates progesterone receptor signaling in mammary epithelial cells. Mol. Endocrinol. 20, 14–34 (2006).
  • Hashizume R, Fukuda M, Maeda Iet al. The RING heterodimer BRCA1-BARD1 is a ubiquitin ligase inactivated by a breast cancer derived mutation. J. Biol. Chem. 276, 14537–14540 (2001).
  • Baer R, Ludwig T. The BRCA1/BARD1 heterodimer a tumor suppressor complex with ubiquitin E3 ligase activity. Curr. Opin. Genet. Dev. 12, 86–91 (2002).
  • Calvo V, Beato M. BRCA1 counteracts progesterone action by ubiquitination leading to progesterone receptor degradation and epigenetic silencing of target promoters. Cancer Res. 71, 3422–3431 (2011).
  • Fan S, Ma YX, Wang Cet al. Role of direct interaction in BRCA1 inhibition of estrogen receptor activity. Oncogene 20, 77–87 (2001).
  • Esteva FJ, Hortobagvi GN. Prognostic molecular markers in early breast cancer. Breast Cancer Res. 6, 109–118 (2004).
  • Payne SJ, Bowen RI, Jones JL, Wells CA. Predictive markers in breast cancer-the present. Histopathology 52, 82–90 (2008).
  • Grann VR, Troxel AB, Zojwalla NJ, Jacobson JS, Hershman D, Neugut AI. Hormone receptor status and survival in a population-based cohort of patients with breast carcinoma. Cancer 103, 2241–2251 (2005).
  • Albert JM, Gonzalez-Angulo AM, Guray Met al. Patients with only 1 positive hormone receptor have increased locoregional recurrence compared with patients with estrogen receptor-positive progesterone receptor-positive disease in very early stage breast cancer. Cancer 15, 1595–1601 (2011).
  • Graham JD, Yeates C, Balleine RIet al. Characterization of progesterone receptor A and B expression in human breast cancer. Cancer Res. 55, 5063–5068 (1995).
  • Mote PA, Bartow S, Tran N, Clarke CL. Loss of co-ordinate expression of progesterone receptors A and B is an early event in breast carcinogenesis. Breast Cancer Res. Treat. 72, 163–173 (2002).
  • Jones GS, Poumand K. An evaluation of etiologic factors and therapy in 555 private patients with primary infertility. Fertil. Steril. 13, 398 (1962).
  • Quagliarello J, Weiss G. Clomiphene citrate in the management of infertility associated with shortened luteal phases. Fertil. Steril. 31, 373 (1979).
  • Downs KA, Gibson M. Clomiphene citrate therapy for luteal phase defect. Fertil. Steril. 39, 34 (1983).
  • Check JH, Nowroozi K, Wu CH, Adelson HG, Lauer C. Ovulation-inducing drugs versus progesterone therapy for infertility in patients with luteal phase defects. Int. J. Fertil. 33, 252–256 (1988).
  • Check JH, Liss J, Check D. The beneficial effect of luteal phase support on pregnancy rates in women with unexplained infertility. Fertil. Steril. 99(3), S23 (2013).
  • Guzick DS, Sullivan MW, Adamson GDet al. Efficacy of treatment for unexplained infertility. Fertil. Steril. 70, 207–213 (1998).
  • Bjuresten K, Landgren BM, Hovatta O, Stavreus-Evers A. Luteal phase progesterone increases live birth rate after frozen embryo transfer. Fertil. Steril. 95, 534–537 (2011).
  • Sonntag B, Ludwig M. An integrated view on the luteal phase: diagnosis and treatment in subfertility. Clin. Endocrinol. 77, 500–507 (2012).
  • Kerin JF, Broom TJ, Ralph MMet al. Human luteal phase function following oocyte cycles. Br. J. Obstet. Gynecol. 88, 1021–1028 (1981).
  • Edwards RG, Steptoe PC, Purdy JM. Establishing full-term human pregnancies using cleaving embryos grown in vitro. Br. J. Obstet. Gynecol. 87, 737–756 (1980).
  • Check JH, Wu CH, Adelson HG. Decreased abortions in hMG-induced pregnancies with prophylactic progesterone therapy. Int. J. Fertil. 30, 45–47 (1985).
  • Check JH, Chase JS, Wu CH, Adelson HG, Teichman M, Rankin A. The efficacy of progesterone in achieving successful pregnancy: I. Prophylactic use during luteal phase in anovulatory women. Int. J. Fertil. 32, 135–138 (1987).
  • van der Linden M, Buckingham K, Farquhar C, Kremer JAM, Metwally M. Luteal phase support for assisted reproduction cycles. Cochrane Database Syst. Rev. (10), CD009154 (2011).
  • McAuley JW, Kroboth FJ, Kroboth PD. Oral administration of micronized progesterone: a review and more experience. Pharmacotherapy 16, 453–457 (1996).
  • Licciardi FL, Kwiatkowski A, Noyes NL, Merkeley AS, Krey LL, Grifo JA. Oral versus intramuscular progesterone for in vitro fertilization: a prospective randomized study. Fertil. Steril. 71, 614–618 (1999).
  • Check JH, Chase JS, Nowroozi K, Wu C. Spontaneous abortion rate in patients with endometriosis treated with progesterone. Int. J. Fertil. 32, 366–368 (1987).
  • Nowroozi K, Chase JS, Check JH, Wu CH. The importance of laparoscopic coagulation of mild endometriosis in infertile women. Int. J. Fertil. 32, 442–444 (1987).
  • Check JH. The association of minimal and mild endometriosis without adhesions and infertility with therapeutic strategies. Clin. Exp. Obst. Gyn. 30(1), 13–8 (2003).
  • Cspo AI, Pukkinen M. Indispensability of the human corpus luteum in the maintenance of early pregnancy: lutectomy evidence. Obstet. Gynecol. Surv. 3, 69 (1978).
  • Herman W, Wyss R, Riondel Aet al. Effects of an anti-progestin steroid in women: interruption of the menstrual cycle or early pregnancy (author’s transl). Contracept. Fertil. Sex. 10, 389–393 (1982).
  • Yeko TR, Gorrill MJ, Hughes MH, Rodi IA, Buster JE, Sauer MV. Timely diagnosis of early ectopic pregnancy using a single blood progesterone measurement. Fertil. Steril. 48, 1048–1050 (1987).
  • Check JH, Winkel CA, Check ML. Abortion rate in progesterone treated women presenting initially with low first trimester serum progesterone levels. Am. J. Gynecol. Health 4, 33–34 (1990).
  • Choe JK, Check JH, Nowroozi K, Benveniste R, Barnea ER. Serum progesterone and 17-hydroxyprogesterone in the diagnosis of ectopic pregnancies and the value of progesterone replacement in intrauterine pregnancies when serum progesterone levels are low. Gynecol. Obstet. Invest. 34, 133–138 (1992).
  • Fazleabas AT. Progesterone resistance in baboon model of endometriosis. Semin Reprod Med 28, 75–80 (2010).
  • Jackson KS, Brudney A, Hastings JM, Mavroginis PA, Kim JJ, Fazleabas AT. The altered distribution of the steroid hormone receptors and the chaperone immunophilin FKBP52 in a baboon model of endometriosis is associated with progesterone resistance during the window of uterine receptivity. Reprod. Sci. 14, 137–150 (2007).
  • Wahabi HA, Fayed AA, Esmaeil SA, Al Zeidan RA. Progestogen for treating threatened miscarriage. Cochrane Database Syst. Rev. (12), CD005943 (2011).
  • Di Renzo GC, Giardina I, Clerici G, Mattei A, Alajmi AH, Gerli S. The role of progesterone in maternal and fetal medicine. Gynecol. Endoc. 28, 925–932 (2012).
  • Check JH, Lee G, Epstein R, Vetter B. Increased rate of preterm deliveries in untreated women with luteal phase deficiencies. Gynecol. Obstet. Invest. 33, 183–184 (1992).
  • Howritz KB. The structure and function of progesterone receptors in breast cancer. J. Steroid Biochem. 27, 447–457 (1987).
  • Horwitz KB, McGuire WL. Predicting response to endocrine therapy in human breast cancer: a hypothesis. Science 189, 726–727 (1975).
  • Ramieu G, Maudelonde T, Ulmann Aet al. The antiprogestin RU486 in advanced breast cancer: preliminary clinical trial. Bull. Cancer 74, 455–461 (1987).
  • Klijn JG, de Jong FH, Bakker GH, Lamberts SW, Rodenburg CJ, Alexieva-Figusch J. Antiprogestins, a new form of endocrine therapy for human breast cancer. Cancer Res. 49, 2851–2856 (1989).
  • Robertson JF, Willsher PC, Winterbottom L, Blamey RW, Thorpe S. Onapristone, a progesterone receptor antagonist, as first-line therapy in primary breast cancer. Eur. J. Cancer 35, 214–218 (1999).
  • Check JH, Sansoucie L, Chern J, Amadi N, Katz Y. Mifepristone treatment improves length and quality of survival of mice with spontaneous leukemia. Anticancer Res. 29(8), 2977–2980 (2009).
  • Check JH, Sansoucie L, Chern J, Dix E. Mifepristone treatment improves length and quality of survival of mice with spontaneous lung cancer. Anticancer Res. 30, 119–122 (2010).
  • Check JH, Dix E, Wilson C, Check D. Progesterone receptor antagonist therapy has therapeutic potential even in cancer restricted to males as evidenced from murine testicular and prostate cancer studies. Anticancer Res. 30, 4921–4923 (2010).
  • Check JH, Dix E, Sansoucie L, Check D. Mifepristone may halt progression of extensively metastatic human adenocarcinoma of the colon–case report. Anticancer Res. 29(5), 1611–1613 (2009).
  • Check JH, Dix E, Cohen R, Check D, Wilson C. Efficacy of the progesterone receptor antagonist mifepristone for palliative therapy of patients with a variety of advanced cancer types. Anticancer Res. 30, 623–628 (2010).
  • Check JH, DiAntonio A, Check Det al. Clinical improvement of symptomatic advancing chronic lymphocytic leukemia following mifepristone therapy despite normal serum levels of the immunomodulatory protein the progesterone-induced blocking factor (PIBF). American Association for Cancer Research Annual Meeting.Washington, DC, 6–10 April 2013.
  • Check JH, Wilson C, Check D. Evidence that mifepristone, a progesterone receptor antagonist, can cross the blood brain barrier and provide palliative benefits for glioblastoma multiforme grade IV. American Association for Cancer Research Annual Meeting.Washington, DC, 6–10 April 2013.
  • Lai JN, Wang OY, Lin VH, Liao CF, Tarng DC, Chien EJ. The non-genomic rapid acidification in peripheral T cells by progesterone depends on intracellular calcium increase and not an Na+/H+-exchange inhibition. Steroids 77, 1017–1024 (2012).
  • Falkenstein E, Norman AW, Wehling M. Mannheim classification of nongenomically initiated (rapid) steroid action(s). J. Clin. Endocrinol. Metab. 85, 2072–2075 (2000).
  • Chien CH, Lai JN, Liao CFet al. Mifepristone acts as progesterone antagonist of non-genomic responses but inhibits phytohemagglutinin-induced proliferation in human T cells. Hum. Reprod. 24, 1968–1975 (2009).
  • Van Voorhis BJ, Anderson DJ, Hill JA. The effects of RU 486 on immune function and steroid-induced immunosuppression in vitro. J. Clin. Endocrinol. Metab. 69, 1195–1199 (1989).
  • Garrido-Gomez T, Ruiz-Alonso M, Blesa D, Diaz-Gimeno P, Vilella F, Simon C. Profiling the gene signature of endometrial receptivity: clinical results. Fertil. Steril. 99, 1078–1085 (2013).
  • Diaz-Gimeno P, Horcajadas JA, Martinez-Conejero JAet al. A genomic diagnostic tool for human endometrial receptivity based on the transcriptomic signature. Fertil. Steril. 95, 50–60 (2011).
  • Diaz-Gimeno P, Ruiz-Alanso M, Blesa Det al. The accuracy and reproducibility of the endometrial receptivity array is superior to histology as a diagnostic method for endometrial receptivity. Fertil. Steril. 99, 508–517 (2013).
  • Evans GE, Martinez-Conejero JA, Phillipson GTMet al. Gene and protein expression signature of endometrial glandular and stromal compartments during the window of implantation. Fertil. Steril. 97, 1365–1373 (2012).
  • Check JH, Choe JK, Katsoff D, Summers-Chase D, Wilson C. Controlled ovarian hyperstimulation adversely affects implantation following in vitro fertilization-embryo transfer. J. Assist. Reprod. Genet. 16, 416–420 (1999).
  • Check JH, Choe JK, Nazari A, Summers-Chase D. Ovarian hyperstimulation can reduce uterine receptivity. A case report. Clin. Exp. Obst. Gyn. 27(2), 89–91 (2000).
  • Check JH, Check ML. A case report demonstrating that follicle maturing drugs may create an adverse uterine environment even when not used for controlled ovarian hyperstimulation. Clin. Exp. Obst. Gyn. 28, 217–218 (2001).
  • Check JH. Minimal and natural stimulations for IVF. In: Advances in Embryo Transfer Chapter 2. Wu B (Ed.). InTech, Rejeka, Croatia, 21–36 (2012).
  • Farhi J, Weissman A, Steinfeld Z, Shorer M, Nahum H, Levran D. Estradiol supplementation during the luteal phase may improve the pregnancy rate in patients undergoing in vitro fertilization-embryo transfer cycles. Fertil. Steril. 73, 73: 761–766 (2000).
  • Lukaszuk K, Liss J, Kukaszuk M, Maj B. Optimization of estradiol supplementation during the luteal phase improves the pregnancy rate in women undergoing in vitro fertilization-embryo transfer cycles. Fertil. Steril. 83, 1372–1376 (2005).
  • Friedler S, Gilboa S, Schachter M, Raziel A, Strassburger D, Ron El R. Luteal phase characteristics following GnRH antagonist or agonist treatment–a comparative study. Reprod. Biomed. Online 12, 27–32 (2006).
  • Smitz J, Bourgain C, Van Waesberghe L, Camus M, Devroey P, Van Steirteghem AC. A prospective randomized study on oestradiol valerate supplementation in addition to intravaginal micronized progesterone in buserelin and HMG induced superovulation. Hum. Reprod. 8, 40–45 (1993).
  • Check JH, Tarquini P, Gandy P, Lauer C. A randomized study comparing the efficacy of reducing the spontaneous abortion rate following lymphocyte immunotherapy and progesterone treatment versus progesterone alone in primary habitual aborters. Gynecol. Obstet. Invest. 39, 257–261 (1995).
  • Cohen R, Check JH. Recurrent aneuploidy–fact of fiction. Clin. Exp. Obst. Gyn. 37, 175–177 (2010).
  • Check JH. The importance of up and down regulation of receptors for FSH, progesterone and the gonadotropin releasing hormone (GnRH) in the ovary, endometrium and lymphocytes in correcting various ovulatory disorders and achieving live deliveries. In: Ovulation: Detection Sign/Symptoms and Outcomes. De Lange K, Deguire E (Eds). Nova Science Publishers, Inc., Hauppauge, NY, (In press).
  • Stadtmauer L, Silverberg KM, Ginsburg ES, Weiss H, Howard B. Progesterone vaginal ring versus vaginal gel for luteal support with in vitro fertilization: a randomized comparative study. Fertil. Steril. 99, 1543–1549 (2013).
  • Ganesh A, Chakravorty N, Mukherjee R, Goswami S, Chaudhury K, Chakravarty B. Comparison of oral dydrogesterone with progesterone gel and micronized progesterone for luteal support in 1,373 women undergoing in vitro fertilization: a randomized clinical study. Fertil. Steril. 95, 1961–1965 (2011).
  • Lanari C, Wargon V, Rojas P, Molinolo AA. Antiprogestins in breast cancer treatment are we ready? Endo. Rel. Cancer19, R35–R50 (2012).
  • Afhuppe W, Beckman JM, Otto Cet al. in vitro characterization of ZK 230211–a type III progesterone receptor antagonist with enhanced antiproliferative properties. J. Steroid. Biochem. Mol. Biol. 119, 45–55 (2010).
  • Chwalisz K, Brenner RM, Fuhrmann UU, Hess-Stumpp H, Elger W. Antiproliferative effects of progesterone antagonists and progesterone receptor modulators on the endometrium. Steroids 65, 741–751 (2000).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.