Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 handwiki -- 2415 2022-10-24 01:31:22

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
HandWiki. Hormonal Breast Enhancement. Encyclopedia. Available online: https://encyclopedia.pub/entry/30800 (accessed on 27 April 2024).
HandWiki. Hormonal Breast Enhancement. Encyclopedia. Available at: https://encyclopedia.pub/entry/30800. Accessed April 27, 2024.
HandWiki. "Hormonal Breast Enhancement" Encyclopedia, https://encyclopedia.pub/entry/30800 (accessed April 27, 2024).
HandWiki. (2022, October 24). Hormonal Breast Enhancement. In Encyclopedia. https://encyclopedia.pub/entry/30800
HandWiki. "Hormonal Breast Enhancement." Encyclopedia. Web. 24 October, 2022.
Hormonal Breast Enhancement
Edit

Hormonal breast enhancement or augmentation is a highly experimental potential medical treatment for the breasts in which hormones or hormonal agents such as estrogen, progesterone, growth hormone (GH), and insulin-like growth factor 1 (IGF-1) are utilized or manipulated to produce breast enlargement in women. It is a possible alternative or supplement to surgical breast augmentation with breast implants or fat transfer and other means of medical breast enlargement. In addition to pharmaceuticals, some herbal breast enlargement supplements contain phytoestrogens such as 8-prenylnaringenin (found in hops) and miroestrol (a constituent of Pueraria mirifica) and thus may be regarded as a form of hormonal breast enhancement. However, evidence of their effectiveness, as well as safety data, are lacking.

miroestrol breast augmentation medical treatment

1. Breast Growth

At puberty, estrogen, but not progesterone at this time, and GH/IGF-1 are critical in mediating the development of the breasts, and are synergistic in doing so.[1][2][3][4][5] In accordance, hormonal contraception and hormone replacement therapy (HRT) with estrogen (and/or progestogens) have been associated with increased breast growth and breast size.[6][7] Moreover, a trial of hormonal breast enhancement in 45 young women with very high doses (80 mg/injection) of intramuscular, bioidentical estrogen (in the form of estradiol polyphosphate, a slow-releasing estradiol prodrug) for six months found that only the women in whom an increase in IGF-1 levels occurred after four weeks (46.7% of subjects) experienced a significant increase in breast size (824.3 mm to 898.5 mm).[8][9] This is in accordance with the established fact that both estrogen and IGF-1 appear to be essential for breast development, and when present together, are synergistic in mediating it.[10][11]

The administration of estrogen to women with Turner syndrome, who normally do not develop breasts due to hypogonadism, results in normal pubertal breast development.[12] Estrogen and GH are often combined in Turner syndrome.[13][14] Estrogen in combination with GH or IGF-1 has been employed safely and effectively to improve bone density in women with anorexia nervosa.[15][16][17] Trans women who are treated with estrogen experience normal pubertal breast development similarly to the case of girls with Turner syndrome.[18] However, they generally show a smaller final breast size in comparison to their immediate relatives (one cup size less on average).[18] This is perhaps due to the fact that most trans women do not commence HRT until adulthood, which is of relevance because GH/IGF-1 levels significantly and progressively decrease after normal adolescent puberty (from late adolescence/early adulthood and thereafter).[19] As such, synergy of estrogen with GH/IGF-1, and by extension, maximal breast development potential, may be reduced.

Systemic administration of GH or IGF-1 causes mammary hyperplasia (enlargement of the mammary glands) in animals.[20] For example, in a study of aged female rhesus macaques, treatment with GH alone, IGF-1 alone, and the combination of GH and IGF-1, were found to produce mammary gland hyperplasia and increased mammary gland size and epithelial proliferation by 2-fold, 3- to 4-fold, and 4- to 5-fold, respectively, changes that were directly correlated with serum concentrations of GH and IGF-1.[21][22][23] Accordingly, research has found that girls with growth hormone deficiency (GHD) who are treated with GH experience accelerated breast growth[24] and that boys with growth hormone deficiency treated with GH sometimes experience gynecomastia.[25] Moreover, IGF-1 levels and activity have been found to be correlated with breast volume in the female general population.[7]

In women with Laron syndrome, where the growth hormone receptor (GHR) is defective and insensitive to GH and serum IGF-1 levels are very low, puberty, including breast development, is delayed, although full sexual maturity is always eventually reached.[26] Moreover, breast development and size are normal (albeit delayed) in spite of GH/IGF-1 axis insufficiency, and in some the breasts may actually be large in relation to body size (which has been hypothesized to be due to increased secretion of prolactin caused by a drift phenomenon from somatomammotrophic cells in the pituitary gland with a high GH secretion).[26][27] An animal model of Laron syndrome, the GHR knockout mouse, shows severely impaired ductal outgrowth at 11 weeks of age.[28][29][30] However, by 15 weeks, ductal development has caught up with that of normal mice and the ducts have fully distributed throughout the mammary fat pad, although the ducts remain narrower than those of wild-type mice.[28][29][30] In any case, female GHR knockout mice can lactate normally.[28][30] As such, taken together, it is said that the phenotypes of women with Laron syndrome and GHR knockout mice are identical, with diminished body size and delayed sexual maturation accompanied by normal lactation.[28]

An adolescent Vietnamese girl with Laron syndrome who was treated with a high dosage of IGF-1 and a gonadotropin-releasing hormone analogue for 3–4 years paradoxically experienced isolated progression of breast development without any other pubertal changes in spite of estrogen levels in the low prepubertal range.[31] Noting that gynecomastia is a recognized complication of treatment with GH and IGF-1, the authors of the study attributed the breast development to a synergism of her high, supraphysiological IGF-1 levels with the low levels of estrogen derived from peripheral aromatization of adrenal androgens.[31]

Certain long-acting growth hormone secretagogues, such as CJC-1295[32] and ibutamoren (MK-677),[33] are capable of reliably and effectively increasing serum GH and IGF-1 concentrations in humans.[34][35][36] Alternatively, exogenous, pharmaceutical GH and IGF-1 (as mecasermin or mecasermin rinfabate) themselves, or analogues of IGF-1 such as des(1-3)IGF-1 and IGF-1 LR3, may be employed to increase GH/IGF-1 axis function. A number of dietary supplements, including L-arginine, L-ornithine, L-lysine, acetyl-L-carnitine, and creatine, may be able to significantly increase GH levels, although evidence is mixed.[37][38][39][40][41] Vitamin D has been found to increase IGF-1 levels in both healthy subjects and individuals with GHD, and vitamin D deficiency is associated with low IGF-1 levels.[42][43][44] However, there is evidence that vitamin D may also potently inhibit breast growth via activation of the vitamin D receptor.[45][46][47][48]

Oral estrogen treatment suppresses IGF-1 production in the liver, where approximately 80% of serum IGF-1 originates from,[49] and reduces total serum IGF-1 levels (by 15–40%, dependent on dose and type of estrogen administered), as well as increases levels of insulin-like growth factor-binding protein 1 (IGFBP1) (a carrier protein that inhibits IGF-1 binding/activity).[50] This results in a state of functional GH resistance (as GH induces IGF-1 production and secretion in the liver to mediate most of its effects),[49] with combined oral estrogen and GH being less effective in evoking the clinical effects of GH relative to GH alone in clinical studies of individuals with hypopituitarism/GHD.[50] In contrast, treatment with combined GH and transdermal estrogen has been found not to decrease IGF-1 levels or increase IGFBP1 levels.[50] As such, estrogen administered via other routes of administration that bypass the liver, such as transdermal (in the form of estrogen patches), sublingual, intranasal, intramuscular injection, and subcutaneous injection, may be significantly more effective than oral estrogen.[50]

Progesterone and non-androgenic progestins, such as dydrogesterone, do not affect serum IGF-1 levels regardless of route of administration.[51] However, androgenic progestins, such as 19-nortestosterone derivatives like norethisterone and levonorgestrel and others like, to a lesser extent, medroxyprogesterone acetate (MPA), when taken orally, induce IGF-1 production via activation of the androgen receptor (AR) in the liver.[51] However, at the same time, androgens potently inhibit estrogen action on the breast, such as by suppressing ER expression in breast tissue, and this action would be expected to likely cancel out any benefit.[52][53] In accordance, a single small clinical study found that the addition of oral MPA to estrogen in trans women undergoing sex reassignment therapy did not result in increased breast size.[54]

Diet and nutrition have been found to affect serum IGF-1 levels. Specifically, low protein intake, fasting, and malnourishment are associated with low IGF-1 levels, whereas obesity is associated with high or normal IGF-1 levels and lowered IGFBP1 and IGFBP3 levels (resulting in higher free IGF-1 concentrations).[55][56][57] In addition, milk consumption and circulating IGF-1 levels have been found to be positively correlated.[58] Aside from diet and nutrition, exercise has also been found to significantly increase GH levels.[37]

Androgens, such as testosterone and dihydrotestosterone (DHT), powerfully suppress the action of estrogen in the breasts.[52][53][59][60] At least one way that they do this is by reducing the expression of the estrogen receptor in breast tissue.[52][53] In women with complete androgen insensitivity syndrome (CAIS), who are completely insensitive to androgens and have only modest levels of estrogen (50 pg/ml), the relatively low levels of estrogen are capable of mediating significant breast development, and the breast sizes of CAIS women, on average, are in fact actually larger than those of non-CAIS women.[59] In males treated with antiandrogens, gynecomastia (enlargement of the breasts in males) and mastodynia (breast tenderness/pain) commonly occur.[61][62][63] Antiandrogens, for instance spironolactone, are also known to cause breast enlargement and mastodynia in women.[64] Some examples of widely used and highly-potent antiandrogens include cyproterone acetate and bicalutamide.[65][66]

Cyclooxygenase-2 (COX-2) overexpression in mammary gland tissue produces mammary gland hyperplasia as well as precocious mammary gland development in female mice, indicating a strong stimulatory effect of this enzyme on the growth of the mammary glands.[67][68] These effects appear to be downstream actions of increased activation of the prostaglandin EP2, EP3, and EP4 receptors, but not the EP1 receptor, in mammary gland tissue, which in turn results in the potent induction of amphiregulin expression, a critical growth factor involved in normal mammary gland development.[67][68] In addition, agonists of the epidermal growth factor receptor (EGFR), the molecular target of amphiregulin, induce COX-2 expression in mammary gland tissue, potentially resulting in a self-perpetuating cycle of growth amplification by COX-2.[67][68] This mechanism is closely related to formation, growth, and spreading of cancers with poor prognosis, and is in accordance with the fact that long-term administration of aspirin, a COX inhibitor, as well as of other COX-inhibiting nonsteroidal anti-inflammatory drugs (NSAIDs), have been found to slightly reduce the risk of breast cancer in women (it is notable here that breast growth/size and breast cancer risk are positively associated).[69] Taken together, these findings indicate that COX-2 inhibitors, such as aspirin, ibuprofen, naproxen, and celecoxib, may decrease breast cell proliferation.[67][68]

Elevated levels of HGF and, to a lesser extent, IGF-1 (by 5.4-fold and 1.8-fold, respectively), in breast stromal tissue, have been found in macromastia, a very rare condition of extremely and excessively large breast size.[70] Exposure of macromastic breast stromal tissue to non-macromastic breast epithelial tissue was found to cause increased alveolar morphogenesis and epithelial proliferation in the latter.[70] A neutralizing antibody for HGF, but not for IGF-1 or EGF, was found to attenuate the proliferation of breast epithelial tissue caused by exposure to macromastic breast stromal cells, potentially directly implicating HGF in the breast growth and enlargement seen in macromastia.[70] As such, treatment with HGF or agonists of its receptor, c-Met, or potentiators of the HGF-c-Met axis (such as dihexa)[71] might have the potential to induce macromastia-like breast growth in an exposure-dependent manner.[70] However, a genome-wide association study highly implicated HGF and c-Met in breast cancer aggressiveness,[72] and a study of women with macromastia indicated that there may be a significant association between macromastia and increased risk of breast cancer.[73]

2. Possible Increased Risk of Cancer

The risk of breast cancer in women is approximately 100-fold that of men.[74] As such, the development of female-typical breasts is associated with a dramatic increase in the risk of breast cancer.[74] Moreover, breast size and breast cancer are positively correlated,[75][76] and macromastia, a condition of excessively large breast size, is considered to be a risk factor for breast cancer.[73] In accordance, it has been hypothesized that there could be a further increase in the risk of breast cancer with hormonal breast enhancement.[9]

Long-term treatment with estrogens and/or progestogens in women, specifically in the form of oral contraceptives, appears to be associated with a slightly increased risk of breast cancer.[77][78] The risk appears to be highest in younger women, notably in those who started taking oral contraceptives before 20 years of age.[79] This could be related to increased synergy of estrogens with the higher levels of GH/IGF-1 that are present with younger age.[80]

Research has suggested that the enhancement of growth factor pathways, including that of GH/IGF-1, could potentially increase the risk of various cancers, including breast cancer.[81] Increased proliferation due to increased IGF-1 activity has been suggested to possibly play a key role in the high risk of breast cancer seen in women with the BRCA1 mutation.[82] Multiple large studies have found a correlation in premenopausal women between serum IGF-1 levels in the upper quartile of the normal range and IGFBP-3 levels in the lower quartile (i.e., high circulating IGF-1 levels and low circulating IFGBP-3 levels) and the risk of developing various cancers, including breast cancer.[83][84] However, the increase in breast cancer risk has been found only to be modest (e.g., only more than twice the usual risk).[85] Subsequent studies have found the increase in risk to be even less clear, and it is notable that high-normal range IGF-1 levels have been found to correlate only with premenopausal and not with postmenopausal breast cancer incidence.[86][87] In any case, mice engineered to have lower circulating levels of IGF-1 show a lower risk of developing various cancers, including breast cancer.[88] In contrast to the case of IGF-1, the upper quintile (20%) of postmenopausal women with the highest of both circulating estrogen and androgen levels have been found to have a significantly increased risk of breast cancer (relative to lowest quintile, the risk is 2- to 3-fold higher).[87] A significant positive association with breast cancer risk has also been found with prolactin levels in postmenopausal women.[87]

In acromegaly, a condition caused and maintained by highly elevated GH/IGF-1 levels, overall, there appears to be little or no increased risk of breast cancer nor certain other cancers (e.g., prostate cancer, lung cancer) relative to that of the general population.[89][90] That said, cancer risk does appear to be consistently elevated in individuals specifically with uncontrolled disease.[91] In addition, there appears to be an increased risk of colorectal cancer and pre-malignant tubular adenomas in acromegaly.[92] In any case, individuals with acromegaly appear to show no increased risk of cancer mortality or general mortality post-treatment (i.e., after their GH/IGF-1 levels have been normalized with medical treatment), and this includes breast cancer.[26][90][93] (Larger-scale studies may be needed, however.)[90][92] In contrast to acromegaly, people with Laron syndrome, a condition characterized by insensitivity to GH and very low IGF-1 levels, have a majorly reduced, in fact almost absent, risk of developing cancer, including breast cancer.[94] There has been concern expressed about doping in athletes with GH/IGF-1 and possible increased risk of cancer, including breast cancer.[95]

References

  1. Brisken; Malley (December 2, 2010). "Hormone Action in the Mammary Gland". Cold Spring Harb. Perspect. Biol. (Cold Spring Harb Perspect Biol) 2 (12): a003178. doi:10.1101/cshperspect.a003178. PMID 20739412.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2982168
  2. "Role of IGF-I in normal mammary development". Breast Cancer Res. Treat. 47 (3): 201–8. 1998. doi:10.1023/a:1005998832636. PMID 9516076.  https://dx.doi.org/10.1023%2Fa%3A1005998832636
  3. "Early mammary development: growth hormone and IGF-1". J Mammary Gland Biol Neoplasia 2 (1): 49–57. 1997. PMID 10887519.  http://www.ncbi.nlm.nih.gov/pubmed/10887519
  4. "Insulin-like growth factor I is essential for terminal end bud formation and ductal morphogenesis during mammary development". Endocrinology 140 (11): 5075–81. 1999. doi:10.1210/endo.140.11.7095. PMID 10537134. http://press.endocrine.org/doi/10.1210/endo.140.11.7095?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%3dpubmed. 
  5. "IGF-I: an essential factor in terminal end bud formation and ductal morphogenesis". J Mammary Gland Biol Neoplasia 5 (1): 7–17. 2000. PMID 10791764.  http://www.ncbi.nlm.nih.gov/pubmed/10791764
  6. Isaksson, Erika; Sahlin, Lena; Söderqvist, Gunnar; von Schoultz, Eva; Masironi, Britt; Wickman, Marie; Wilking, Nils; von Schoultz, Bo et al. (1999). "Expression of sex steroid receptors and IGF-1 mRNA in breast tissue — effects of hormonal treatment". J. Steroid Biochem. Mol. Biol. 70 (4–6): 257–262. doi:10.1016/S0960-0760(99)00115-6. ISSN 0960-0760.  https://dx.doi.org/10.1016%2FS0960-0760%2899%2900115-6
  7. Jernström, H; Sandberg, T; Bågeman, E; Borg, Å; Olsson, H (2005). "Insulin-like growth factor-1 (IGF1) genotype predicts breast volume after pregnancy and hormonal contraception and is associated with circulating IGF-1 levels: implications for risk of early-onset breast cancer in young women from hereditary breast cancer families". Br. J. Cancer 92 (5): 857–866. doi:10.1038/sj.bjc.6602389. ISSN 0007-0920. PMID 15756256.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2361904
  8. R.E. Mansel; Oystein Fodstad; Wen G. Jiang (14 June 2007). Metastasis of Breast Cancer. Springer Science & Business Media. pp. 217–. ISBN 978-1-4020-5866-0. https://books.google.com/books?id=14pb5b6gT-oC&pg=PA217. 
  9. "Hormonal breast augmentation: prognostic relevance of insulin-like growth factor-I". Gynecol. Endocrinol. 12 (2): 123–7. 1998. doi:10.3109/09513599809024960. PMID 9610425.  https://dx.doi.org/10.3109%2F09513599809024960
  10. Derek Leroith; Walter Zumkeller; Robert C. Baxter (31 July 2003). Insulin-like Growth Factor Receptor Signalling. Springer Science & Business Media. pp. 189–. ISBN 978-0-306-47846-8. https://books.google.com/books?id=2pJS5CeoUYIC&pg=PA189. 
  11. Kleinberg, David L.; Ruan, Weifeng (2008). "IGF-I, GH, and Sex Steroid Effects in Normal Mammary Gland Development". J. Mammary Gland Biol. Neoplasia 13 (4): 353–360. doi:10.1007/s10911-008-9103-7. ISSN 1083-3021. PMID 19034633.  https://dx.doi.org/10.1007%2Fs10911-008-9103-7
  12. Ismail Jatoi; Manfred Kaufmann (11 February 2010). Management of Breast Diseases. Springer Science & Business Media. pp. 19–. ISBN 978-3-540-69743-5. https://books.google.com/books?id=nsUBW3-qJ9MC&pg=PA19. 
  13. Suzanne B. Cassidy; Judith E. Allanson (20 September 2011). Management of Genetic Syndromes. John Wiley & Sons. pp. 1330–. ISBN 978-1-118-21067-3. https://books.google.com/books?id=E8sHMf96uuoC&pg=PT1330. 
  14. "The patient with Turner syndrome: puberty and medical management concerns". Fertil. Steril. 98 (4): 780–6. 2012. doi:10.1016/j.fertnstert.2012.07.1104. PMID 22884020.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=3760009
  15. "Endocrine consequences of anorexia nervosa". Lancet Diabetes Endocrinol. 2 (7): 581–92. 2014. doi:10.1016/S2213-8587(13)70180-3. PMID 24731664.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=4133106
  16. "Effects of recombinant human insulin-like growth factor (IGF)-I and estrogen administration on IGF-I, IGF binding protein (IGFBP)-2, and IGFBP-3 in anorexia nervosa: a randomized-controlled study". J. Clin. Endocrinol. Metab. 88 (3): 1142–9. 2003. doi:10.1210/jc.2002-021402. PMID 12629097.  https://dx.doi.org/10.1210%2Fjc.2002-021402
  17. "Effects of recombinant human IGF-I and oral contraceptive administration on bone density in anorexia nervosa". J. Clin. Endocrinol. Metab. 87 (6): 2883–91. 2002. doi:10.1210/jcem.87.6.8574. PMID 12050268.  https://dx.doi.org/10.1210%2Fjcem.87.6.8574
  18. James Barrett (2007). Transsexual and Other Disorders of Gender Identity: A Practical Guide to Management. Radcliffe Publishing. pp. 172–. ISBN 978-1-85775-719-4. https://books.google.com/books?id=I-8qZlGIpnQC&pg=PA172. 
  19. "The potential clinical applications of insulin-like growth factor-1 ligand in human breast cancer". Anticancer Res. 27 (3B): 1617–24. 2007. PMID 17595785. http://ar.iiarjournals.org/cgi/pmidlookup?view=long&pmid=17595785. 
  20. "Growth hormone and insulin-like growth factor-I in the transition from normal mammary development to preneoplastic mammary lesions". Endocr. Rev. 30 (1): 51–74. 2009. doi:10.1210/er.2008-0022. PMID 19075184.  https://dx.doi.org/10.1210%2Fer.2008-0022
  21. Alice C. Levine (3 October 2011). Hormones and Cancer: Breast and Prostate, An Issue of Endocrinology and Metabolism Clinics of North America,. Elsevier Health Sciences. pp. 26–. ISBN 1-4557-1239-6. https://books.google.com/books?id=ZabQAQAAQBAJ&pg=PT26. 
  22. Douglas Yee (2004). Insulin-like Growth Factors. IOS Press. pp. 33–. ISBN 978-1-58603-409-2. https://books.google.com/books?id=y-tZ989CulEC&pg=PA33. 
  23. "Growth hormone treatment induces mammary gland hyperplasia in aging primates". Nat. Med. 3 (10): 1141–4. 1997. doi:10.1038/nm1097-1141. PMID 9334728.  https://dx.doi.org/10.1038%2Fnm1097-1141
  24. Sat Dharam Kaur (2003). The Complete Natural Medicine Guide to Breast Cancer: A Practical Manual for Understanding, Prevention & Care. R. Rose. p. 79. ISBN 978-0-7788-0083-5. https://books.google.com/books?id=_MAhAQAAMAAJ. 
  25. Souza, Flavio Moutinho; Collett-Solberg, Paulo Ferrez (2011). "Adverse effects of growth hormone replacement therapy in children". Arq. Bras. Endocrinol. Metab. 55 (8): 559–565. doi:10.1590/S0004-27302011000800009. ISSN 0004-2730.  https://dx.doi.org/10.1590%2FS0004-27302011000800009
  26. Zvi Laron; J. Kopchick (25 November 2010). Laron Syndrome - From Man to Mouse: Lessons from Clinical and Experimental Experience. Springer Science & Business Media. pp. 113, 498. ISBN 978-3-642-11183-9. https://books.google.com/books?id=THLPdLG4000C&pg=PA113. 
  27. Laron, Zvi (2004). "Laron Syndrome (Primary Growth Hormone Resistance or Insensitivity): The Personal Experience 1958–2003". J. Clin. Endocrinol. Metab. 89 (3): 1031–1044. doi:10.1210/jc.2003-031033. ISSN 0021-972X. PMID 15001582.  https://dx.doi.org/10.1210%2Fjc.2003-031033
  28. Brisken, Cathrin (2002). "Hormonal Control of Alveolar Development and Its Implications for Breast Carcinogenesis". J. Mammary Gland Biol. Neoplasia 7 (1): 39–48. doi:10.1023/A:1015718406329. ISSN 1083-3021.  https://dx.doi.org/10.1023%2FA%3A1015718406329
  29. McNally, Sara; Martin, Finian (2011). "Molecular regulators of pubertal mammary gland development". Ann. Med. 43 (3): 212–234. doi:10.3109/07853890.2011.554425. ISSN 0785-3890.  https://dx.doi.org/10.3109%2F07853890.2011.554425
  30. "A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone receptor/binding protein gene (the Laron mouse)". Proc. Natl. Acad. Sci. U.S.A. 94 (24): 13215–20. 1997. doi:10.1073/pnas.94.24.13215. PMID 9371826.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=24289
  31. Walker, J. L.; Crock, P. A.; Behncken, S. N.; Rowlinson, S. W.; Nicholson, L. M.; Boulton, T. J. C.; Waters, M. J. (1998). "A Novel Mutation Affecting the Interdomain Link Region of the Growth Hormone Receptor in a Vietnamese Girl, and Response to Long-Term Treatment with Recombinant Human Insulin-Like Growth Factor-I and Luteinizing Hormone-Releasing Hormone Analogue1". J. Clin. Endocrinol. Metab. 83 (7): 2554–2561. doi:10.1210/jcem.83.7.4954. ISSN 0021-972X.  https://dx.doi.org/10.1210%2Fjcem.83.7.4954
  32. Teichman, Sam L.; Neale, Ann; Lawrence, Betty; Gagnon, Catherine; Castaigne, Jean-Paul; Frohman, Lawrence A. (2006). "Prolonged Stimulation of Growth Hormone (GH) and Insulin-Like Growth Factor I Secretion by CJC-1295, a Long-Acting Analog of GH-Releasing Hormone, in Healthy Adults". J. Clin. Endocrinol. Metab. 91 (3): 799–805. doi:10.1210/jc.2005-1536. ISSN 0021-972X. PMID 16352683.  https://dx.doi.org/10.1210%2Fjc.2005-1536
  33. Copinschi, G; Van Onderbergen, A; L'Hermite-Balériaux, M; Mendel, C M; Caufriez, A; Leproult, R; Bolognese, J A; De Smet, M et al. (1996). "Effects of a 7-day treatment with a novel, orally active, growth hormone (GH) secretagogue, MK-677, on 24-hour GH profiles, insulin-like growth factor I, and adrenocortical function in normal young men.". J. Clin. Endocrinol. Metab. 81 (8): 2776–2782. doi:10.1210/jcem.81.8.8768828. ISSN 0021-972X.  https://dx.doi.org/10.1210%2Fjcem.81.8.8768828
  34. "The growth hormone secretagogue receptor". Vitam. Horm. 77: 47–88. 2008. doi:10.1016/S0083-6729(06)77004-2. PMID 17983853.  https://dx.doi.org/10.1016%2FS0083-6729%2806%2977004-2
  35. "Novel mechanisms of growth hormone regulation: growth hormone-releasing peptides and ghrelin". Braz. J. Med. Biol. Res. 39 (8): 1003–11. 2006. doi:10.1590/s0100-879x2006000800002. PMID 16906274.  https://dx.doi.org/10.1590%2Fs0100-879x2006000800002
  36. "Growth hormone-releasing hormone and growth hormone secretagogue-receptor ligands: focus on reproductive system". Endocrine 14 (1): 35–43. 2001. doi:10.1385/endo:14:1:035. PMID 11322500.  https://dx.doi.org/10.1385%2Fendo%3A14%3A1%3A035
  37. Marie Dunford; J. Doyle (3 February 2014). Nutrition for Sport and Exercise. Cengage Learning. pp. 190–. ISBN 978-1-285-75249-5. https://books.google.com/books?id=xRAeCgAAQBAJ&pg=PA190. 
  38. Ira Wolinsky; Judy A. Driskell (25 June 2004). Nutritional Ergogenic Aids. CRC Press. pp. 27–28. ISBN 978-0-203-50770-4. https://books.google.com/books?id=Ix8l0yAHqvUC&pg=PA27. 
  39. Fred Brouns; Cerestar-Cargill (7 February 2003). Essentials of Sports Nutrition. John Wiley & Sons. pp. 141–. ISBN 978-0-470-85536-2. https://books.google.com/books?id=QolubvuTbowC&pg=PA141. 
  40. Joseph F. Audette; Allison Bailey (26 February 2008). Integrative Pain Medicine: The Science and Practice of Complementary and Alternative Medicine in Pain Management. Springer Science & Business Media. pp. 159–. ISBN 978-1-59745-344-8. https://books.google.com/books?id=GWQr7FJhBe0C&pg=PA159. 
  41. "Acute creatine loading enhances human growth hormone secretion". J. Sports Med. Phys. Fit. 40 (4): 336–42. 2000. PMID 11297004.  http://www.ncbi.nlm.nih.gov/pubmed/11297004
  42. Template:Vcite2 journal
  43. Template:Vcite2 journal
  44. Template:Vcite2 journal
  45. Wanda M. Haschek; Colin G. Rousseaux; Matthew A. Wallig (1 May 2013). Haschek and Rousseaux's Handbook of Toxicologic Pathology. Elsevier Science. pp. 2675–. ISBN 978-0-12-415765-1. https://books.google.com/books?id=RXsdAAAAQBAJ&pg=PA2675. 
  46. "Vitamin D and the mammary gland: a review on its role in normal development and breast cancer". Breast Cancer Res. 14 (3): 211. 2012. doi:10.1186/bcr3178. PMID 22676419.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=3446331
  47. "Targets of vitamin D receptor signaling in the mammary gland". J. Bone Miner. Res. 22 Suppl 2: V86–90. 2007. doi:10.1359/jbmr.07s204. PMID 18290729.  https://dx.doi.org/10.1359%2Fjbmr.07s204
  48. "Vitamin D metabolism in mammary gland and breast cancer". Mol. Cell. Endocrinol. 347 (1–2): 55–60. 2011. doi:10.1016/j.mce.2011.05.020. PMID 21669251.  https://dx.doi.org/10.1016%2Fj.mce.2011.05.020
  49. Pauline M. Camacho (26 September 2012). Evidence-Based Endocrinology. Lippincott Williams & Wilkins. pp. 20–. ISBN 978-1-4511-7146-4. https://books.google.com/books?id=s06wXkPAnfcC&pg=PA20. 
  50. Isotton, A. L.; Wender, M. C. O.; Casagrande, A.; Rollin, G.; Czepielewski, M. A. (2011). "Effects of oral and transdermal estrogen on IGF1, IGFBP3, IGFBP1, serum lipids, and glucose in patients with hypopituitarism during GH treatment: a randomized study". Eur. J. Endocrinol. 166 (2): 207–213. doi:10.1530/EJE-11-0560. ISSN 0804-4643. PMID 22108915. http://www.eje-online.org/content/166/2/207.full.pdf. 
  51. Sonnet, Emmanuel; Lacut, Karine; Roudaut, Nathalie; Mottier, Dominique; Kerlan, Véronique; Oger, Emmanuel (2007). "Effects of the route of oestrogen administration on IGF-1 and IGFBP-3 in healthy postmenopausal women: results from a randomized placebo-controlled study". Clin. Endocrinol. 66 (5): 626–631. doi:10.1111/j.1365-2265.2007.02783.x. ISSN 0300-0664. PMID 17492948.  https://dx.doi.org/10.1111%2Fj.1365-2265.2007.02783.x
  52. "Breast size in relation to endogenous hormone levels, body constitution, and oral contraceptive use in healthy nulligravid women aged 19-25 years". Am. J. Epidemiol. 145 (7): 571–80. 1997. doi:10.1093/oxfordjournals.aje.a009153. PMID 9098173. http://aje.oxfordjournals.org/cgi/pmidlookup?view=long&pmid=9098173. 
  53. "Testosterone inhibits estrogen-induced mammary epithelial proliferation and suppresses estrogen receptor expression". FASEB J. 14 (12): 1725–30. 2000. doi:10.1096/fj.99-0863com. PMID 10973921. http://www.fasebj.org/cgi/pmidlookup?view=long&pmid=10973921. 
  54. "Physical and hormonal evaluation of transsexual patients: a longitudinal study". Arch. Sex. Behav. 15 (2): 121–38. 1986. doi:10.1007/bf01542220. PMID 3013122.  https://dx.doi.org/10.1007%2Fbf01542220
  55. Lawrence F. Ditmier (1 January 2006). New Developments in Obesity Research. Nova Publishers. pp. 20–21. ISBN 978-1-60021-296-3. https://books.google.com/books?id=BfeTsTAiq6MC&pg=PA20. 
  56. Peter G. Kopelman; Ian D. Caterson; William H. Dietz (15 April 2008). Clinical Obesity in Adults and Children. John Wiley & Sons. pp. 202–. ISBN 978-1-4051-4366-0. https://books.google.com/books?id=qGVnItPoPCYC&pg=PA202. 
  57. Robert H. Eckel (2003). Obesity: Mechanisms and Clinical Management. Lippincott Williams & Wilkins. pp. 157–. ISBN 978-0-7817-2844-7. https://books.google.com/books?id=v_NXqwsljYYC&pg=PA157. 
  58. "Milk consumption and circulating insulin-like growth factor-I level: a systematic literature review". Int. J. Food Sci. Nutr. 60 Suppl 7: 330–40. 2009. doi:10.1080/09637480903150114. PMID 19746296.  https://dx.doi.org/10.1080%2F09637480903150114
  59. Jerome F. Strauss, III; Robert L. Barbieri (13 September 2013). Yen and Jaffe's Reproductive Endocrinology. Elsevier Health Sciences. pp. 236–. ISBN 978-1-4557-2758-2. https://books.google.com/books?id=KZ95AAAAQBAJ&pg=PA236. 
  60. Christopher B. Wilson; Victor Nizet; Yvonne Maldonado; Jack S. Remington; Jerome O. Klein (24 February 2015). Remington and Klein's Infectious Diseases of the Fetus and Newborn Infant. Elsevier Health Sciences. pp. 190–. ISBN 978-0-323-24147-2. https://books.google.com/books?id=VuZ1BwAAQBAJ&pg=PA190. 
  61. Jeanne Held-Warmkessel (2006). Contemporary Issues in Prostate Cancer: A Nursing Perspective. Jones & Bartlett Learning. pp. 256–. ISBN 978-0-7637-3075-8. https://books.google.com/books?id=dZe4ZSVDdBsC&pg=PA256. 
  62. "Drug-induced gynecomastia". Pharmacotherapy 32 (12): 1123–40. 2012. doi:10.1002/phar.1138. PMID 23165798.  https://dx.doi.org/10.1002%2Fphar.1138
  63. "Gynaecomastia--pathophysiology, diagnosis and treatment". Nat. Rev. Endocrinol. 10 (11): 684–98. 2014. doi:10.1038/nrendo.2014.139. PMID 25112235.  https://dx.doi.org/10.1038%2Fnrendo.2014.139
  64. Jeffrey K. Aronson (2 March 2009). Meyler's Side Effects of Cardiovascular Drugs. Elsevier. pp. 255–. ISBN 978-0-08-093289-7. https://books.google.com/books?id=oeBgU3UwgZkC&pg=PA255. 
  65. Kenneth L. Becker (2001). Principles and Practice of Endocrinology and Metabolism. Lippincott Williams & Wilkins. pp. 1196–. ISBN 978-0-7817-1750-2. https://books.google.com/books?id=FVfzRvaucq8C&pg=PA1196. 
  66. Sarah H. Wakelin; Howard I. Maibach; Clive B. Archer (21 May 2015). Handbook of Systemic Drug Treatment in Dermatology, Second Edition. CRC Press. pp. 29–. ISBN 978-1-4822-2286-9. https://books.google.com/books?id=fCysCQAAQBAJ&pg=PA29. 
  67. "The prostaglandin E2 receptor EP2 is required for cyclooxygenase 2-mediated mammary hyperplasia". Cancer Res. 65 (11): 4496–9. 2005. doi:10.1158/0008-5472.CAN-05-0129. PMID 15930264.  https://dx.doi.org/10.1158%2F0008-5472.CAN-05-0129
  68. "Cyclooxygenase-2 transactivates the epidermal growth factor receptor through specific E-prostanoid receptors and tumor necrosis factor-alpha converting enzyme". Cell. Signal. 19 (9): 1956–63. 2007. doi:10.1016/j.cellsig.2007.05.003. PMID 17572069.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2681182
  69. "Effect of COX-2 inhibitors and other non-steroidal inflammatory drugs on breast cancer risk: a meta-analysis". Breast Cancer Res. Treat. 149 (2): 525–36. 2015. doi:10.1007/s10549-015-3267-9. PMID 25589172.  https://dx.doi.org/10.1007%2Fs10549-015-3267-9
  70. Zhong, Aimei; Wang, Guohua; Yang, Jie; Xu, Qijun; Yuan, Quan; Yang, Yanqing; Xia, Yun; Guo, Ke et al. (2014). "Stromal-epithelial cell interactions and alteration of branching morphogenesis in macromastic mammary glands". J Cell Mol Med 18 (7): 1257–1266. doi:10.1111/jcmm.12275. ISSN 1582-1838. PMID 24720804.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=4124011
  71. Benoist, C. C.; Kawas, L. H.; Zhu, M.; Tyson, K. A.; Stillmaker, L.; Appleyard, S. M.; Wright, J. W.; Wayman, G. A. et al. (2014). "The Procognitive and Synaptogenic Effects of Angiotensin IV-Derived Peptides Are Dependent on Activation of the Hepatocyte Growth Factor/c-Met System". J. Pharmacol. Exp. Ther. 351 (2): 390–402. doi:10.1124/jpet.114.218735. ISSN 1521-0103. PMID 25187433.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=4201273
  72. "Pathway analysis of breast cancer genome-wide association study highlights three pathways and one canonical signaling cascade". Cancer Res. 70 (11): 4453–9. 2010. doi:10.1158/0008-5472.CAN-09-4502. PMID 20460509.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2907250
  73. Talghini, Shahla (2013). "Is Macromastia a Risk Factor for Breast Cancer? A Study on 198 Patients". Pak. J. Biol. Sci. 16 (21): 1348–1352. doi:10.3923/pjbs.2013.1348.1352. PMID 24511745.  https://dx.doi.org/10.3923%2Fpjbs.2013.1348.1352
  74. Pathobiology of Human Disease: A Dynamic Encyclopedia of Disease Mechanisms. Elsevier Science. 1 August 2014. pp. 946–. ISBN 978-0-12-386457-4. https://books.google.com/books?id=uQB0AwAAQBAJ&pg=PA946. 
  75. Jansen, L.A.; Backstein, R.M.; Brown, M.H. (2014). "Breast size and breast cancer: A systematic review". J. Plast. Reconstr. Aesthetic Surg. 67 (12): 1615–1623. doi:10.1016/j.bjps.2014.10.001. ISSN 1748-6815. PMID 25456291.  https://dx.doi.org/10.1016%2Fj.bjps.2014.10.001
  76. Eriksson, Nicholas; Benton, Geoffrey M; Do, Chuong B; Kiefer, Amy K; Mountain, Joanna L; Hinds, David A; Francke, Uta; Tung, Joyce Y (2012). "Genetic variants associated with breast size also influence breast cancer risk". BMC Medical Genetics 13 (1): 53. doi:10.1186/1471-2350-13-53. ISSN 1471-2350. PMID 22747683.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=3483246
  77. "Oral contraceptives and breast cancer risk overall and by molecular subtype among young women". Cancer Epidemiol. Biomarkers Prev. 23 (5): 755–64. 2014. doi:10.1158/1055-9965.EPI-13-0944. PMID 24633144.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=4032363
  78. "Oral contraceptive use and breast cancer: a prospective study of young women". Cancer Epidemiol. Biomarkers Prev. 19 (10): 2496–502. 2010. doi:10.1158/1055-9965.EPI-10-0747. PMID 20802021.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=3055790
  79. Monica Morrow; Virgil Craig Jordan (2003). Managing Breast Cancer Risk. PMPH-USA. pp. 50–. ISBN 978-1-55009-260-8. https://books.google.com/books?id=HXKibhaF5lMC&pg=PA50. 
  80. Lukanova, A.; Toniolo, P.; Zeleniuch-Jacquotte, A.; Grankvist, K.; Wulff, M.; Arslan, A. A.; Afanasyeva, Y.; Johansson, R. et al. (2006). "Insulin-like Growth Factor I in Pregnancy and Maternal Risk of Breast Cancer". Cancer Epidemiol. Biomarkers Prev. 15 (12): 2489–2493. doi:10.1158/1055-9965.EPI-06-0625. ISSN 1055-9965.  https://dx.doi.org/10.1158%2F1055-9965.EPI-06-0625
  81. "Insulin-like growth factor-I and cancer risk". Growth Horm. IGF Res. 14 (4): 261–9. 2004. doi:10.1016/j.ghir.2004.01.005. PMID 15231294.  https://dx.doi.org/10.1016%2Fj.ghir.2004.01.005
  82. "Breast cancer risk in BRCA1 mutation carriers: insight from mouse models". Ann. Oncol. 24 Suppl 8: viii8–viii12. 2013. doi:10.1093/annonc/mdt305. PMID 24131977.  https://dx.doi.org/10.1093%2Fannonc%2Fmdt305
  83. "The insulin-like growth factor system and cancer". Cancer Lett. 195 (2): 127–37. 2003. doi:10.1016/s0304-3835(03)00159-9. PMID 12767520.  https://dx.doi.org/10.1016%2Fs0304-3835%2803%2900159-9
  84. "Mechanisms by which IGF-I may promote cancer". Cancer Biol. Ther. 2 (6): 630–5. 2003. doi:10.4161/cbt.2.6.678. PMID 14688466.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=4164051
  85. "Polymorphisms and circulating levels in the insulin-like growth factor system and risk of breast cancer: a systematic review". Cancer Epidemiol. Biomarkers Prev. 14 (1): 2–19. 2005. PMID 15668470.  http://www.ncbi.nlm.nih.gov/pubmed/15668470
  86. "Insulin-like growth factor (IGF)-I, IGF binding protein-3, and breast cancer risk: eight years on". Endocr. Relat. Cancer 13 (2): 273–8. 2006. doi:10.1677/erc.1.01219. PMID 16728563.  https://dx.doi.org/10.1677%2Ferc.1.01219
  87. "Endogenous hormones and risk of breast cancer in postmenopausal women". Breast Dis. 24: 3–15. 2005. doi:10.3233/bd-2006-24102. PMID 16917136.  https://dx.doi.org/10.3233%2Fbd-2006-24102
  88. "Circulating IGF-1 and its role in cancer: lessons from the IGF-1 gene deletion (LID) mouse". Novartis Found. Symp. 262: 3–9; discussion 9–18, 265–8. 2004. PMID 15562820.  http://www.ncbi.nlm.nih.gov/pubmed/15562820
  89. "IGFs and human cancer: implications regarding the risk of growth hormone therapy". Horm. Res. 51 Suppl 3: 42–51. 1999. doi:10.1159/000053161. PMID 10592443.  https://dx.doi.org/10.1159%2F000053161
  90. "Oncological complications of excess GH in acromegaly". Pituitary 5 (1): 21–5. 2002. PMID 12638722.  http://www.ncbi.nlm.nih.gov/pubmed/12638722
  91. "Acromegaly: re-thinking the cancer risk". Rev. Endocr. Metab. Disord. 9 (1): 41–58. 2008. doi:10.1007/s11154-007-9063-z. PMID 18157698.  https://dx.doi.org/10.1007%2Fs11154-007-9063-z
  92. "Acromegaly and cancer". Horm. Res. 62 Suppl 1: 108–15. 2004. doi:10.1159/000080768. PMID 15761242.  https://dx.doi.org/10.1159%2F000080768
  93. Nathan A. Berger (19 June 2015). Murine Models, Energy Balance, and Cancer. Springer. pp. 76–. ISBN 978-3-319-16733-6. https://books.google.com/books?id=qdzyCQAAQBAJ&pg=PA76. 
  94. Gallagher, Emily Jane; LeRoith, Derek (2011). "Is Growth Hormone Resistance/IGF-1 Reduction Good for You?". Cell Metab. 13 (4): 355–356. doi:10.1016/j.cmet.2011.03.003. ISSN 1550-4131. PMID 21459318.  https://dx.doi.org/10.1016%2Fj.cmet.2011.03.003
  95. "Doping with growth hormone/IGF-1, anabolic steroids or erythropoietin: is there a cancer risk?". Pharmacol. Res. 55 (5): 359–69. 2007. doi:10.1016/j.phrs.2007.01.020. PMID 17349798.  https://dx.doi.org/10.1016%2Fj.phrs.2007.01.020
More
Information
Subjects: Others
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 9.6K
Entry Collection: HandWiki
Revision: 1 time (View History)
Update Date: 24 Oct 2022
1000/1000