Hormone Replacement for Breast Cancer Survivors Part Two by Jeffrey Dach MD
Michelle is a 52 year old CEO of a high tech company with severe menopausal symptoms of hot flashes, night sweats, insomnia, osteoarthritis in her finger joints, brain fog, chronic fatigue, and recurrent urinary tract infections. Three years ago, Michelle found a lump in her breast, and had a lumpectomy for breast cancer (infiltrating ductal). Since then, her oncologist has been treating her with the estrogen blocking drug, anastrozole. Michelle is now in my office seeking relief from her menopausal symptoms asking for bioidentical hormone replacement therapy (BHRT). The mainstream medical dogma is that hormone replacement is contraindicated for the breast cancer survivor (BCS) for fear of inducing recurrent cancer or a new cancer. In part one of this series, the opposite argument was presented. We now have 25 studies showing hormone replacement is beneficial for the BCS’s quality of life, without increased breast cancer recurrence compared to controls. Perhaps the strongest advocate for treating the BCS with BHRT is Avrum Bluming, MD, now retired after 50 years practicing oncology in Encino, California. Dr. Bluming was a clinical professor of medical oncology at the University of Southern California and spent four years as a senior investigator for the National Cancer Institute. He is author of the book, Estrogen Matters. (1) (47-48)
Header Image: Red Ellipse and Arrow shows the peak in HRT use at 35 million in 2002. Decline started after the publication of the 2002 WHI study. Green arrow shows decline in use to 10 million in 2011. Vertical axis: numbers of users, and horizontal axis: Date. MHT=menopausal hormone therapy. Courtesy of Fig. 1 (Lancet 2019) Collaborative Group on Hormonal Factors in Breast Cancer. “Type and timing of menopausal hormone therapy and breast cancer risk: individual participant meta-analysis of the worldwide epidemiological evidence.” Lancet (London, England) 394.10204 (2019): 1159.
The above header chart is a graphic illustration of the dramatic decline in HRT use after publication of the WHI 2002 study. Another major shift after 2002, not shown in this chart, is the change in the predominant form of HRT from (oral estrogen, and oral medroxyprogesterone) to topical estrogen combined with natural progesterone as discussed in 2011 by Dr. Agnes Fournier from France using E3N and GAZEL, two French prospective cohort studies. The predominant estrogen in the U.S. was Premarin (CEE) while in France, the predominant estrogen was estradiol. (49)
Dr. Bluming’s 26 Studies
Credit and thanks goes to Dr. Bluming for his work on hormone therapy for breast cancer survivors, and his meticulous collection of 26 relevant medical studies. Of the 26 studies, 25 showed no evidence of increased cancer recurrence in the hormone treated group (HRT). The only study to show increased recurrence with HRT is the HABITS study from Sweden. This study used estradiol combined with norethisterone, a highly carcinogenic progestin derived from a testosterone backbone. (1)
In addition, all patients with recurrence in the HRT group were also taking tamoxifen which blocks estrogen. In 2020, Dr. Lindsey Berkson writes in the Townsend Letter:
Of the 20 studies between 1980 and 2008 that showed estrogen was not only safe
for breast cancer patients but was also protective, only the HABITS study found an increased risk of recurrence in breast cancer patients on HRT. As previously stated, this risk only occurred if the women were on tamoxifen, which “blocked” the action of estrogen. (55-56)
In a recent podcast September 5, 2024, Dr. Bluming says:
there are now 26 studies in the medical literature that I could find in languages that I can understand, and I reviewed all of those studies. Of the 26 studies, 25 show no increased risk of recurrence of breast cancer among women who take hormone replacement therapy (HRT) of any kind. And that’s independent of whether they had estrogen receptor positive or estrogen receptor negative breast cancer. The one study that suggested an increased risk of recurrence is a study that was done in Sweden called the HABITS Study. HABITS stands for Hormones After Breast Cancer: Is it Safe? And that study reported an increased risk of contra, or local breast cancer, not an increased risk of distant recurrence. And there was no increased risk of death from breast cancer. (4)
in 2022 , Drs. Abbie Laing and Louise Newson reviewed recurrence of breast cancer in Survivors taking HRT, writing:
Of all the data, there is currently only 1 study [HABITS] that describes an increased risk of recurrence in women taking hormonal replacement therapy after a history of breast cancer, whereas 3 studies demonstrate a decreased risk [John Eden (1995), Ellen O’Meara (2001) and, Meurer (2002)], and most studies demonstrate no difference whatsoever. (101) (10) (20) (31)
In 2022, Dr Avrum Bluming, writing in the Cancer Journal reviewed hormone replacement therapy (HRT) after breast cancer. This is his list of 6 studies showing less recurrence in the hormone treated group (36):
Palshof et al. 1980 Prospective randomized trial: Reduced recurrence (102)
Palshof et al. 1985 Updates of the original study: Reduced recurrence, Reduced mortality.(Note: the abstract does not have full the information on estrogen treated group) (103)
Eden et al. 1995 Retrospective case-control: Reduced recurrence. (20)
Guidozzi 1999 Prospective single-arm: No recurrence. (28)
O’Meara et al. 2001 Retrospective case-control: Reduced recurrence, Reduced mortality. (10)
Durna et al. 2002 Retrospective observational: Reduced recurrence Reduced mortality (20)
Bluming 2008 Prospective cohort: Reduced recurrence. (34)
In this newsletter, we will take a deep dive into these 26 studies compiled by Dr. Bluming through the lens of estrogen receptors, protective effects of pregnancy, prior estrogen exposure, carcinogenic progestins, various hormone preparations that increase or decrease the risk of breast cancer, and the effects of LTED, long term estrogen deprivation. And of course, the first and most important question is: what is the exact hormone formula used in each study?
Estrogen is a Growth Factor which Stimulates Cancer Cells
Laboratory studies on breast cancer cells show that estrogen is a growth factor that stimulates growth and breast cancer cell division. In the 1970’s, many in-vitro studies of breast cancer cells such as this 1976 study by Dr. Marc Lippman who worked with MCF-7 breast cancer cells. Dr. Marc Lippman found that estrogen at physiologic levels is a growth signal and stimulates cell division. Anti-estrogen drugs which block estrogen receptors serve as growth inhibitors. However, the growth and cell division stimulation by estrogen is biphasic, with a cancer cell killing effect seen at high estrogen concentrations. This cancer killing effect of estrogen can be amplified by submitting the cells to long term estrogen deprivation (LTED). (57)
Two other obvious facts which question a causative role of estrogen in breast cancer are:
1) the greater incidence of breast cancer in women than males by a factor of 100.
Ly, Diana, et al. “An international comparison of male and female breast cancer incidence rates.” International journal of cancer 132.8 (2013): 1918-1926.
2) the use of estrogen blocking drugs as standard of care for women at increased risk for breast cancer.
Owens, Douglas K., et al. “Medication use to reduce risk of breast cancer: US preventive services task force recommendation statement.” Jama 322.9 (2019): 857-867.
Shete, Nivida, Jordan Calabrese, and Debra A. Tonetti. “Revisiting Estrogen for the treatment of endocrine-resistant breast cancer: novel therapeutic approaches.” Cancers 15.14 (2023): 3647.
Because of the above considerations, the mainstream medical dogma is to contra-indicate the use of estrogen hormone replacement for breast cancer survivors thinking this would increase risk for breast cancer recurrence. This has produced a “fear of estrogen” among the general population and also among the medical profession. The fear of estrogen was amplified by the media in 2002 after the publication of the 2002 first arm WHI study showing 26 percent increased breast cancer in the HRT group (Premarin/MPA). As Dr. Avrum Bluming says in a recent interview, the data from published studies and actual clinical practice does not support dogmatic denial of HRT for the breast cancer survivor. (4)
Cancer Treatments Cause Long Term Estrogen Deficiency
The other factor to consider is that current mainstream treatments for breast cancer survivors are estrogen blocking drugs, tamoxifen, aromatase inhibitors (anastrazole) and LH blockers (Zoladex). These drugs cause severe climacteric symptoms making women miserable who seek relief, and embark on a futile search for a doctor willing to provide hormone replacement. Especially distressing are symptoms related to genito-urinary syndrome affecting 70% of menopausal women, yet only 7% receive treatment, as discussed in 2022 by Drs. Abbie Laing and Louise Newson, writing:
Symptoms [of genito-urinary syndrome] are very common, affecting approximately 70% of all menopausal women. Symptoms include genital changes (dryness, itching, burning, pain, and irritation), sexual concerns (dyspareunia, decreased arousal, reduced lubrication, postcoital bleeding, reduced or absent orgasm), and urinary problems (urgency, nocturia, dysuria, recurrent UTIs, and urinary incontinence)… Collectively, these symptoms can cause significant distress, reduced quality of life, depression, anxiety, and other mood disorders and unlike vasomotor symptoms do not usually improve with time; instead, they are chronic and progressive. A response to hormonal treatments is usually rapid and sustained with most women obtaining substantial relief even after 3 weeks…Thus, it remains very concerning that only 7% of women with GSM [genito-urinary-syndrome of menopause] receive treatment. (101)
All the above symptoms can be completely alleviated by the use of intra-vaginal estrogen. In our office we use vaginal estriol combined with testosterone for less severe cases, and vaginal Bi-est (estriol/estradiol 4:1 with progesterone) and topical testosterone for more severe cases. In Europe, vaginal estriol (E3) and estradiol (E2) are widely available at conventional pharmacies. However in the U.S. vaginal estriol (E3) is available only from compounding pharmacies. The “narrative” controlled by Big Pharma in medical journals has vilified compounding pharmacies. The result is symptomatic women in the US rarely receive vaginal estriol, while this is more commonly done in Europe. Withholding vaginal HRT from symptomatic menopausal women denies them the health benefits for urogenital system (genito-urinary syndrome), bones, joints, heart and cognitive function. So the decision to withhold HRT from breast cancer survivors is not to be taken lightly. We will review the published data and make a few observations.
High Dose Estrogen Equal to Tamoxifen for Metastatic Breast Cancer
If estrogen is a growth factor for breast cancer cells, then how is it possible that estrogen has been used to treat breast cancer? Back in the 1950’s and 1960’s, high dose synthetic estrogen was the mainstay treatment for metastatic breast cancer. The drug was DES, 50 times more potent than estradiol, and response rate was about 30 percent. In 1944, Dr. Alexander Haddow was the first to demonstrate high dose estrogen therapy using synthetic DES Di-Ethyl-Stilbestrol showing efficacy for treatment of metastatic breast cancer, and DES was mainstay treatment for this disease until the 1970’s when DES was replaced by tamoxifen (TAM). In 2009, Dr. Matthew J Ellis showed that daily dose of 6 mg of oral estradiol may serve instead of DES, similarly providing clinical benefit in 30 percent of metastatic breast cancer patients who had failed aromatase inhibitors after 3-5 years of long term estrogen deprivation (LTED). (58-61)
Shete, Nivida, Jordan Calabrese, and Debra A. Tonetti. “Revisiting Estrogen for the treatment of endocrine-resistant breast cancer: novel therapeutic approaches.” Cancers 15.14 (2023): 3647.
Studies comparing efficacy of DES (high dose estrogen) with that of tamoxifen (TAM) in treating progressive metastatic breast cancer showed high dose estrogen was more effective at improving survival than TAM. And, the duration of response was about the same for both. Because TAM was easier to tolerate and had less toxicity, TAM replaced DES, and became standard of care in 1974. (6-8)
LTED Long Term Estrogen Deprivation
Dr. V. Craig Jordan, whose name became synonymous with tamoxifen, spent his career in the laboratory trying to understand breast cancer. One of his discoveries is the concept of long term estrogen deprivation (LTED). Dr. Jordan discovered that LTED transforms estrogen from a growth signal to a death signal, and reintroduction of estrogen will induce mitochondrial apoptosis, programmed cell death in the cancer cell. LTED up-regulates ER-alpha, so when estrogen is reintroduced, ER alpha sends excess signals which stimulate mitochondria to induce apoptosis, programmed cell death. Think of it this way. If you plug your 110 volt lamp into a 220 volt socket for the dryer or washing machine, what happens? the light bulb “pops” as it burns out, same as the cancer cells undergoing apoptosis. The voltage is too high for the little fragile light bulb, and the estrogen is too much for the cancer cells after LTED. Note: Sadly, Dr. V. Craig Jordan passed away on June 9, 2024 at age 76 of renal cell carcinoma. (6-8) (94)
Estrogen Receptor Alpha and Beta, and How Hormone Components Increase or Decrease Breast Cancer Risk
The discovery of estrogen receptor (ER) alpha by Elwood Jensen in 1958 and cloned in 1985, and ER beta by Jan-Åke Gustafsson in 1995, gives us greater understanding why various hormone preparations increase or decrease breast cancer risk. It all depends on preferential binding to ER alpha or ER beta. ER alpha is the pro-carcinogenic, proliferative receptor while ER beta is the tumor suppressor receptor, so best results are obtained with hormones or natural agents that bind preferntially to ER-beta and down regluate ER-alpha. Hormones which preferentially target ER-beta or down-regulate ER-alpha are breast cancer preventive. Specifically, E3 (estriol), Premarin (B-ring unsaturated steroids), testosterone (3-beta-diol) and progesterone are breast cancer protective. Hormones or hormone disrupting chemicals (HDCs) which preferentially target ER-alpha are proliferative and pro-carcinogenic. This explains why estradiol is more proliferative than Premarin (CEE). Estradiol targets ER-alpha and ER-beta equally, (50%:50%), while Premarin contains B-ring unsaturated steroids which preferentially bind to ER-beta. Progesterone is breast cancer protective acting as a proliferative brake on ER alpha. However progestins do the exact opposite, activating ER-alpha and down stream oncogenes, cyclin D1 and cMYC. In 2012, Sebastián Giulianelli studied a mouse breast cancer model showing the progestin, medroxyprogesterone activates ER-alpha leading to oncogene activation of cMYC and Cyclin D1. This behavior is the exact opposite to natural progesterone which acts as a proliferative brake on ER-alpha based on the 2015 study by Hisham Mohammed. Dr. Sebastián Giulianelli writes:
In this study, we used a murine progestin-dependent tumor to investigate the role of ER alpha in progestin-induced tumor cell proliferation. We found that treatment with the progestin medroxyprogesterone acetate (MPA) induced the expression and activation of ER alpha, as well as rapid nuclear colocalization of activated ER alpha with PR. Treatment with the pure antiestrogen fulvestrant to block ER alpha disrupted the interaction of ER alpha and PR in vitro and induced the regression of MPA-dependent tumor growth in vivo. ER alpha blockade also prevented an MPA-induced increase in CYCLIN D1 (CCND1) and MYC expression [oncogenes]. Chromatin immunoprecipitation studies showed that MPA triggered binding of ER alpha and PR to the CCND1 and MYC promoters [oncogenes]. Interestingly, blockade or RNAi-mediated silencing of ER alpha inhibited ER alpha, but not PR binding to both regulatory sequences, indicating that an interaction between ER alpha and PR at these sites is necessary for MPA-induced gene expression and cell proliferation. We confirmed that nuclear colocalization of both receptors also occurred in human breast cancer samples. Together, our findings argued that ER alpha–PR association on target gene promoters is essential for progestin-induced cell proliferation. (62-65)
In 2015, Dr. Hisham Mohammed progesterone acting as a proliferative brake to ER alpha, writing:
In this scenario, under estrogenic conditions, an activated PR [progesterone receptor] functions as a proliferative brake in ER-alpha+ breast tumours by re-directing ER alpha chromatin binding and altering the expression of target genes that induce a switch from a proliferative to a more differentiated state. (66)
Another mechanism for synthetic progestins carcinogenicity arises from its androgenic properties which interference with the androgen signalling, thus progestins abrogate ER-beta signalling by androgens. Carcinogenicity of the many synthetic progestins will vary based on androgenic activity. The more androgenicity, the greater the carcinogencicity. This explains why norethisterone made from a testosterone backbone is more carcinogenic than medroxyprogesterone (MPA), made from a progesterone backbone. The carcinogenicity of progestins is dose dependent. At lower doses used clinically, they compete with androgen receptors and are carcinogenic. At higher doses, they become actual androgens and are used for breast cancer treatment with efficacy similar to tamoxifen and testosterone itself. (95-97)
Exogenous Estrogen vs Tumor Produced Estrogen
Even though estrogen is a growth factor for breast cancer, exogenous estrogens may not be a significant factor because breast tumors independently make their own estrogen, thus adding in exogenous estrogen may have little effect on tumor growth. In 2001, Dr. Ellen O’Meara writes:
Breast tumors can regulate and maintain internal levels of estradiol independent of levels outside the tumor, so exogenous estrogens may have relatively little effect on tumor growth. (10)
Actually things are more nuanced than this because one must also consider the differing effects of the two receptors, ER-alpha, which is proliferative, and ER-beta which is a tumor suppressor. LTED upregulates estrogen receptor-alpha (ER alpha) four to ten-fold, so that estrogen re-introduction causes mitochondrial apoptosis of the breast cancer cells via ER alpha. One must consider the beneficial effect of LTED upon starting HRT which will induce apoptosis within pre-existing microscopic nests of breast cancer cells in the post menopausal breast cancer survivor who has been estrogen deprived for a few years. (67)
Prior Exposure to HRT or High Hormone Levels of Pregnancy Are Protective
Another factor is the protective effect of high hormone levels of pregnancy, estradiol, estriol and progesterone, confering a 70 percent decrease in breast cancer risk. The high endogenous hormone levels reprogram the epigenome of breast cells making them less responsive to oncogene activation. Prior exposure to exogenous estrogen has a similar protective effect. In 2020, Dr. Lindsey Berkson discusses the breast cancer effects of pregnancy writing:
In fact, there is a 70% decrease in breast cancer risk associated with a full-term pregnancy before the age of 18. It’s also been shown that pregnancy is safe after treatment of breast cancer, even among estrogen receptor–positive women patients (ER+ means pathologists identify estrogen receptors in the tumor). (55-56) (68-70)
When doing HRT studies, prior HRT use and number of pregnancies should be included in the data. In 2002, Dr. Durna studies HRT in breast cancer survivors, and recorded the total number of pregnancies (parity) and prior use of HRT, two factors which can effect outcome. The median parity was 2.2 children for both HRT users and non-users, so this was not a factor in the study. However, prior use of HRT (before breast cancer diagnosis) was greater in the HRT user group of breast cancer survivors (67% vs. 19% ). This factor, by itself, could skew the data in favor of lower recurrence for the HRT-user group. Remember, high hormone levels of pregnancy confer protection from breast cancer by reprogramming the epigenome to be more resistant to oncogene activation, as discussed by Dr. Mary Feigman (2020). This same mechanism is thought to be in play for women with prior HRT exposure. Prior HRT exposure protection from breast cancer was demonstrated by Howard Hodis in his 2018 re-analysis of the WHI study. Upon removal of women with prior HRT exposure from the placebo group, there was no longer increased cancer risk in the HRT group using Premarin and MPA, 2004, first arm WHI. (68) (70)
In 2002, Dr. Eva Durna from Australia study results showed the HRT group had 38 percent reduction in recurrence, 66 percent reduction in both all-cause and breast cancer mortality compared to non-users. Dr. Durna made two pertinent observations to explain the excellent results of her study. Her first comment is a recognition of the beneficial effect of LTED, and the second comment is a recognition of increased risk of breast cancer with synthetic progestins, known to be carcinogenic, writing:
HRT users had reduced risk of cancer recurrence (adjusted relative risk [RR], 0.62; 95% CI,0.43–0.87), all-cause mortality (RR, 0.34; 95% CI, 0.19–0.59) and death from primary tumour (RR, 0.40; 95% CI, 0.22–0.72)…A possible explanation of the [good] results is that women with estrogen deficiency tend to have better outcomes after breast cancer. [This is the beneficial effect of LTED] …Other recent observational studies also suggest that the use of sequential or cyclic progestins in HRT may increase the risk of breast cancer. [This is the recognition of progestins as carcinogenic]. (20)
Estrogen Metabolism Studies?
We have previously discussed SNP’s or mutations in estrogen metabolism playing an important role in the genesis of breast cancer. In 2021, Dr. Ercole Cavalier showed specific estrogen metabolites cause breast cancer. The 4-hydroxy-estrogen metabolites lead to cardinogenic 4-hydroxy-quinones which are DNA adducts. The 2-hydroxy estrogen metabolites lead to 2-methoxy-estradiol, an endogenous anti-cancer drug. However, none of these 26 studies evaluated estrogen metabolites or genetic studies pertaining to COMT, CYP1B1, CYP1B2, 4-hydroxy-estrogens, etc. Future studies should incorporate either metabolic or genetic testing, so this type of data can be recorded and analyzed. For more on estrogen metabolism and breast cancer see this previous newsletter: Estrogen Metabolism, Iodine, 2MEO Part Three. (71)
Did the Study HRT Include Testosterone ?
I was very impressed with Drs. Rebecca Glaser and Gary Donowitz studies showing subcutaneous testosterone pellets inserted into menopausal women reduce the incidence of breast cancer by about 40%.(38-40)
Testosterone Opposes Carcinogenic Progestins
In 2007, Dr. Marie Hofling from Stockholm Sweden asked the question, could testosterone oppose the carcinogenic effects of synthetic progestins such as the highly carcinogenic noresthisterone used in the HABITS trial? Dr. Hofling studied 88 postmenopausal women over 6-month in a prospective, randomized, double-blind, placebo-controlled study looking at estrogen/progestin HRT induced breast proliferation with breast biopsy specimens. All patients were given continuous combined estradiol 2 mg and norethisterone acetate 1 mg. Half were randomized to a testosterone skin patch or placebo. Dr. Marie Hofling found, yes, the addition of testosterone opposed the proliferative effects of the estradiol/norethisterone combination, writing:
Addition of testosterone may counteract breast cell proliferation as induced by
estrogen/progestogen therapy in postmenopausal women. (72)
Androgen Receptor Potently Inhibits ER-alpha Stimulation
In 2009, Dr. Amelia Peters studied androgen receptors in 215 invasive ductal breast carcinomas, followed with in-vitro studies on these same breast cancer cells showing the androgen receptor (AR) binds to estrogen response elements (ERE), thus potently inhibiting ER-alpha transactional activity and estradiol induced proliferation, writing:
There is emerging evidence that the balance between estrogen receptor-alpha (ER(alpha)) and androgen receptor (AR) signaling is a critical determinant of growth in the normal and malignant breast. In this study, we assessed AR status in a cohort of 215 invasive ductal breast carcinomas. AR and (ER alpha) were coexpressed in the majority (80-90%) of breast tumor cells.,, AR potently inhibited (ER alpha) transactivation activity and 17beta-estradiol-stimulated growth of breast cancer cells. Transfection of MDA-MB-231 breast cancer cells with either functionally impaired AR variants or the DNA-binding domain of the AR indicated that the latter is both necessary and sufficient for inhibition of (ER(alpha)) signaling. Consistent with molecular modeling, electrophoretic mobility shift assays showed binding of the AR to an estrogen-responsive element (ERE). Evidence for a functional interaction of the AR with an ERE in vivo was provided by chromatin immunoprecipitation data, revealing recruitment of the AR to the progesterone receptor promoter in T-47D breast cancer cells. We conclude that, by binding to a subset of EREs, the AR can prevent activation of target genes that mediate the stimulatory effects of 17beta-estradiol on breast cancer cells. (98)
DHT Inhibits E2-induced Expression of Cyclin D1 mRNA
in 2012, Dr. Natalija Eigeliene from Finland studied the effect of testosterone on human breast tissue obtained from reduction mammoplasty operations of postmenopausal women (explants). These tissue samples were then cultured and studied in-vitro with or without testosterone (T) and its more potent metabolite 5α-dihydrotestosterone (DHT) or in combination with 17 beta-estradiol (E2) for 7 days and 14 days. Dr. Natalija Eigeliene found the testosterone and its more active metabolite, DHT, inhibited proliferation of the human breast tissue cells grown in culture. When estradiol (E2) was added to the culture media, testosterone opposed the estradiol (E2) stimulated proliferation, and DHT inhibited E2-induced expression of cyclin D1 mRNA. Note: cyclin D1 is an oncogene, causing breast cancer, so inhibiting mRNA expression of cyclin D1 is highly cancer preventive. Eigeliene writes:
T and DHT reduced proliferation and increased apoptosis in breast epithelium, the effects of which were reversed by bicalutamide [androgen blocker]. In combination with E2, they [T and DHT] suppressed E2-stimulated proliferation and cell survival. DHT also inhibited basal (P < 0.05) and E2-induced expression of cyclin-D1 mRNA (P < 0.05). (104)
Testosterone Treats Progressive Metastatic Breast Cancer
In 2014, Dr. Corrado Boni from Emilia, Italy did a retrospective study of 53 breast cancer patients treated with intramuscular injections of testosterone propionate for progessive metastic disease non-responsive to multiple chemotherapy, and endocrine treatments. About 60 percent of the patients had a favorable outcome with regression in 10 patients and stabilization in 22 patients. About 40% of patients did not respond to testosterone with progressive disease. Median survival after starting testosterone was about a year. (105)
Does the HRT Contain Testosterone?
So, getting back to the 26 studies collated by Dr. Bluming the next logical question is: Does the HRT formula include testosterone? One such study in 1999 by Dr. Puthgraman Natrajan from Augusta, Georgia included testosterone pellets in 38 of 50 patients on HRT after a diagnosis of breast cancer. His study showed an 81% reduction in mortality in HRT users. Testosterone metabolite 3-beta-diol targets ER-beta and is strongly cancer preventive. I would expect similar excellent outcomes for any study including testosterone. On a practical basis, it is imperative that all HRT formulas include testosterone for its superb ability to prevent breast cancer. (13) (38-40)
Vaginal Estrogen Associated with Exceptionally Good Outcomes
In 2002, Dr. Eva Durna from Australia noticed the excellent outcomes with vaginal estradiol-alone in 32 breast cancer survivors. These results were much better than all other hormone formulas and all other types of hormone delivery (oral and transdermal). This group of 32 patients showed the lowest relative risk (RR for recurrence, 0.18 compared to non-users. The RR of 0.18 means HRT users had 82 percent less recurrence than HRT non-users. This is astounding! Dr. Durna writes:
those who used vaginal estrogen alone [32 patients] had a significantly lower risk of recurrence or new breast cancer (adjusted RR, 0.18; 95% CI, 0.04–0.75). (20)
In 2001, Dr. George Peters studied HRT use in breast cancer survivors, finding eight vaginal estrogen patients in his study group that had results so outstanding, they were reported separately, so as not to skew results of the main study. In these eight patients followed for 11.4 years, there were no cancer recurrences and no deaths. Dr. Peters writes:
Of the eight patients who have used only vaginal cream ERT, median follow-up from diagnosis was 11.4 years, and median time on ERT since diagnosis was 4.0
years [LTED]. There have been no contralateral breast cancers; no local, regional, or distant recurrences; and no cancer deaths in this group. (16)
More on Vaginal Estrogen
A 2019 study by the Collaborative Group in Lancet was a meta-analysis of 58 epidemiological studies from 21 countries including 143,887 postmenopausal women with invasive breast cancer and 424,972 without breast cancer serving as controls. All women in this 2019 Lancet study had been using post-menopausal HRT when the breast cancer diagnosis was made. This 2019, Lancet study reports the vaginal delivery of estrogen is the only form of menopausal hormone replacement NOT associated with increased breast cancer risk. Note: this 2019 Lancet study is not a breast cancer survivor study. It includes only every-day post menopausal HRT users. This study also found greater breast cancer risk when synthetic progestins are used. The authors write:
Every MHT [menopausal hormone therapy] type, except vaginal estrogens, was associated with excess breast cancer risks, which increased steadily with duration of use and were greater for estrogen-progestagen than estrogen-only preparations. (21)
A 2006 Finnish study of 110,000 women by Dr. H. Lyytinen found vaginal use of any type of estrogen-alone without progestin was not associated with increased breast cancer risk, writing:
The use of estradiol [without progestin] was associated with an increased risk of breast cancer after 5 years of use (incidence ratio=1.3-1.4). Neither an oral estriol regimen nor vaginal use of any estrogen formulations were accompanied by a significantly increased risk of breast cancer. (73)
Going back to the HABITS trial in which HRT users had 3 fold increase in recurrence compared to non-users, it was stated that all the patients with recurence were also taking tamoxifen, suggesting tamoxifen was a factor for the increased recurrence. What about the use of vaginal estrogen (LHT local hormone therapy) in patients also taking tamoxifen or aromatase inhibitors? Has this been studied? Yes it has. In 2012, Dr. Isabelle Le Ray from Dijon, France used a large UK database to do a cohort study with nested case-control analysis. Recurrence rate in controls was 2.6 percent. Vaginal estrogen users had reduced recurrence (RR: 0.78). Tamoxifen treated vaginal estrogen users also had less recurrence tamoxifen-treated patients (RR: 0.83). However for estrogen users on aromatase inhibitors using vaginal estrogen, no patient had a recurrence. It would be nice to know the vaginal estrogen formula, whether Premarin, estradiol or estriol was used and at what dosage. We do not have this information, and should. Dr. Isabelle Le Ray writes:
A total of 13,479 women were included in the study, of which 2,673 received AIs, 10,806 received tamoxifen, and 271 received LHT [local hormone therapy]. Mean age at cohort entry was 63.7 years, and mean follow-up was 3.5 years. The crude recurrence rate 25.9 per 1,000 per year. Overall, the use of LHT was not associated with an increased risk of recurrence (RR: 0.78, 95 % CI 0.48-1.25) compared with non-use. In stratified analyses, LHT did not increase the risk of recurrence among tamoxifen-treated patients (RR: 0.83, 95 % CI 0.51-1.34), while the risk was not estimable among AI-treated patients since no patients receiving LHT experienced a recurrence. The use of LHT is not associated with an increase in breast cancer recurrence among women receiving a hormone therapy. (100).
What is the explanation for these outstanding results for vaginal estrogen use? We still do not know. My thinking on this goes as follows: Firstly, in the studies using estrogen-alone via transvaginal delivery, there was no added carcinogenic progestin. The second explanation is that many vaginal preparations contain estriol (E3) which preferentially binds to ER-beta, the tumor suppressor. Thirdly, vaginal delivery has much better absorption rate compared to transdermal, reaching higher blood levels without first pass through the liver. Thus vaginal estrogen (estradiol E2 and estriol E3), used at systemic doses, closely mimics the high hormone levels of pregnancy which is known for centuries to confer protection from breast cancer. These factors account for the excellent outcomes with vaginal estrogen alone. (74-79)
The Value of LTED, Long Term Estrogen Deprivation
As mentioned above, long term estrogen deprivation (LTED) transforms estrogen from a growth signal to a death signal, which causes mitochondrial apoptosis in the breast cancer cell. For the typical breast cancer survivor treated for 3-5 years with estrogen blocking drugs such as aromatase inhibitors (Letrozole) and LH blocking drugs (Zoladex), these women have been living through 3-5 years of estrogen deficiency and are generally miserable with climacteric symptoms. When started on estrogen hormone replacement after LTED, these women enjoy the benefits of estrogen induced apoptosis rendering an excellent prognosis. Of the 26 studies compiled by Dr. Bluming, the one study which best illustrates the value of LTED is a small study of 25 breast cancer survivors by Dr. Alan G. Wile from Orange County California. In this study, patients were divided into two groups based on HRT free interval after diagnosis of breast cancer. For Group one, the LTED was less than 2 years (average 8 months), and for Group II, the LTED was greater than 2 years (average 64.5 months). All three patients with breast cancer recurrence were in Group I with the shorter length of estrogen deprivation, less than 2 years. Dr. Wiles writes:
The HRT-free interval for group I patients averaged 7.9 months and for group II patients averaged 64.5 months…Three of 25 patients have had a recurrence, all in group I. (27)
Increased Cancer Risk with Progestins, Medroxyprogesterone, Levonorgestrol and Northisterone
In 2021, Dr. Irene Lambrinoudaki reviewed the 2020 study by Dr. Vinogradova, a case controlled study using two large UK general practice databases. Dr. Vinogradova’s study revealed over 9 years of use, estrogen alone (84% CEE, equine estrogen, 16% estradiol) therapy showed only a marginal increase in breast cancer (OR=1.14) , while the combination of estrogen with a progestin showed a more pronounced increase in breast cancer (OR=1.87), writing:
Estrogen-only therapy for up to 9 years increased only marginally the risk of breast cancer (OR 1.14, CI 1.08–1.21), whereas estrogen–progestin combination therapy for the same duration was associated with a more pronounced increase in breast cancer risk (OR 1.70, CI 1.64 to 1.76). The risk differed according to the progestin used, being higher with medroxyprogesterone acetate [OR=1.87], levonorgestrel [OR=1.79] and norethisterone [1.88] …and lower with dydrogesterone [OR=1.24] for more than 5 years of therapy. The excess risk dissipated in past users…At the time of our study, two types of oestrogen (conjugated equine oestrogen and estradiol) and four types of progestogen (norethisterone acetate, levonorgestrel, medroxyprogesterone, and dydrogesterone) were commonly prescribed in the UK and were included in our analyses. (80-81)
The HABITS Study
Above image: HABITS study chart shows 3-fold higher recurrence rate in HRT users. Red Arrow HRT. Green Arrow NON-HRT. Cumulative incidence of breast cancer recurrence (vertical axis). Chart is Fig 1.courtesy of Dr. Lars Holmberg HABITS 2008. (31)
HABITS by Dr. Lars Holmberg of Sweden
This brings us to the next obvious question, which is why on earth would anyone give a carcinogenic progestin, norethisterone, for menopausal hormone replacement when cancer preventive natural progesterone is available? This is the HABITS study by Dr. Lars Holmberg of Sweden which used estradiol combined with norethisterone, a highly carcinogenic progestin, resulting in three times greater recurrence in the HRT group after 2.1 years of use. One should take note that there was no increased recurrence in HABITS trial in these subgroups: women using estrogen alone, or in women using Premarin (CEE). We have previously discussed how Premarin (CEE) has breast cancer protective properties conferred by the unsaturated B-ring steroids which preferentially bind to ER-beta, the tumor suppressor. The binding affinity of unsaturated B-ring steroids in Premarin (CEE) to ER-beta is 5-6 times greater than the binding affinity to ER-alpha.The use of Premarin -alone could explain the favorable results in the WHI second arm 2004 in which HRT users had a 23 percent reduction in breast cancer compared to non-users. The 18 year follow up showed the Premarin-alone users had a 45 percent reduction in mortality from breast cancer. Mainstream medicine mindlessly lumps together estradiol and Premarin (CEE) calling them both estrogens, and giving them rough equality. They are not equal. Whenever the medical literature reports a study in which estrogen was used, we need to know what type of estrogen? Did the study use Premarin (CEE) or estradiol? The cancer preventive properties of Premarin (CEE) vs. the proliferative properties of estradiol were recognized in 2019 by Richard J. Santen, writing:
Notably, pre-clinical studies demonstrated that conjugated equine estrogen [Premarin,CEE} , as used in the WHI [ Women’s Health Initiative, second arm, 2004 ], has unique, pro-apoptotic properties compared to the anti-apoptotic effects of estradiol, a finding providing an explanation for the reduction in breast cancer with conjugated equine estrogen. (99)
The dismal outcome of the main study of the HABITS trial obscures the improved outcomes within subgroups. Notice there was no increased recurrence in the estrogen-alone sub-group compared with the control group of non-users. This raises an accusing finger at the carcinogenicity of norethisterone, the synthetic progestin. All patients with breast cancer recurrence were also taking Tamoxofen, an estrogen blocking drug. In 2022, Dr. Avrum Bluming discusses the HABITS trial, writing:
The HABITS trial was prematurely terminated on December 17, 2003, after only 2 years of median follow-up and after only 434 women of the proposed 1300 had been enrolled. The reason for the sudden termination, according to the initial paper, was the disproportionate number of women randomized to HRT who developed another breast cancer (26 of 174 = 15%), compared with only 7 of the 171 (5%) randomized to no HRT. The increase was seen only as local recurrences or contralateral tumors. There was no increase in the development of distant metastases, nor was there an increase in the risk of death. Further, there was no increase among women randomized to estrogen alone; there was no increase when Premarin [CEE] (conjugated estrogens) was used as the source of estrogen; there was no increase among women who had been initially diagnosed with lymph node involvement, and the increase was noted only among women who were taking tamoxifen in conjunction with HRT. (1) (30-31)
In the HABITS trial the increased recurrence was noted only in women on HRT (estradiol plus norethisterone) taking tamoxifen. There was no increased recurrence in women taking estrogen alone (without a progestin) and no increase when the estrogen was Premarin (with or without a progestin). This tells us a number of things. Firstly, never give estradiol and noresthisterone to women on Tamoxifen. This is a bad combination. But if you have to use a progestin, use medroxyprogesterone with Premarin. The unsaturated B ring steroids bind to ER beta, a tumor suppressor, so Premarin tends to counteract the carcinogenicity of the progestin. Estrogen alone, without the progestin is OK with no recurrence reported in the HABITS trial for estrogen-alone. (82-84)
The Stockholm Study
The 2005, Stockholm Study by Dr. Eva von Schoultz was a parallel study to the HABITS trial by Lars Holberg from Sweden. The main difference between these two studies was that Dr. Eva von Schoultz of the Stockholm study recognized the carcinogenic effect of progestins, and modified the Stockholm study design to minimize the use of the progestin, medroxyprogesteone (MPA) thought to be less carcinogenic than the norethisterone used in the HABITS study. A second study modification was reduction in exposure time to the progestin. Instead of continuous progestin use, this was changed to sequential use for 10 days per month, every third month in 73 percent of the women. As a result, the Stockholm study showed much better results with an RR value of 0.82 for recurrence in the hormone treated group, meaning there was 18 percent less recurrence for HRT users compared to non-HRT users in the Stockholm Study. I am rather perplexed as to why the Stockholm study designers could not bring themselves to using natural progesterone rather than medroxyprogesterone. Study results would have been much better. In any event, by replacing norethisterone with medroxyprogesterone (MPA), a less carcinogenic progestin, and changing continuous use (30 days a month ) to sequential use (10 days a month) and a “spacing out regimen” to reduce its use even further, the Stockholm study resulted in less recurrence for the HRT users (RR=0.82), and both total mortality and breast cancer mortality was reduced in half compared to non-users.
The author, Dr. Eva von Schoultz stated in her discussion section that changing the progestin, and reducing its use was the main difference between the two studies, HABITS and Stockholm, responsible for yielding the better results. In his 2022 review article, Dr. Avrum Bluming quite thoroughly discussed both studies, mentioning the larger percentage of women in the HRT group of the Stockholm study also taking tamoxifen (52% vs 34%), and criticizing the HABITS study for not doing baseline mammograms. Yet, Dr. Bluming was silent about much larger issue, the carcinogenicity of synthetic progestins. This is surprising since the stated goal of the Stockholm study was to minimize the use of synthetic progestins, a goal not shared by the HABITS trial. The rationale for minimizing the use of progestins was a 1996 monkey study (cynomolgus macaques) by Dr. J. Mark Cline at Wake Forest, North Carolina in which menopausal monkeys were treated with either conjugated equine estrogens (Premarin, CEE) and medroxyprogesterone acetate (MPA), or Premarin-alone (CEE), in doses equivalent to human HRT. Dr.J. Mark Cline found greater breast cell proliferation in the combined treatment (CEE plus MPA) compared to estrogen alone (Premarin, CEE). Dr.J. Mark Cline concludes the clinical implications of his monkey study is that women receiving combined therapy (CEE plus MPA) may have a greater risk for breast cancer than women receiving CEE alone. Dr.J. Mark Cline writes:
These results [of Dr. Cline’s monkey study] indicate a proliferative response of mammary gland epithelium to therapy with conjugated equine estrogens plus medroxyprogesterone acetate in postmenopausal macaques. The clinical implication of this finding may be a greater risk for development of breast neoplasms in women receiving combined hormone replacement therapy. (41-46)
Apparently, Dr. Eva von Schoultz took this to heart and made it the rationale for minimizing exposure to MPA in the Stockholm study, and then stated this reduced exposure to progestins is the reason for improved results in the Stockholm Study (less recurrence RR=0.82) compared to the HABITS study (more recurrence RH, Relative Hazard=3.3). In 2005, the first author of the Stockholm study, Dr. Eva von Schoultz, writes:
In 1997 two independent randomized clinical trials, Hormonal Replacement Therapy After Breast Cancer–Is It Safe? (HABITS; 434 patients) and the Stockholm trial (378 patients), were initiated in Sweden to compare menopausal hormone therapy with no menopausal hormone therapy after diagnosis of early-stage breast cancer. Much of the design of both studies was similar; however, a goal of the Stockholm protocol, not shared with the HABITS trial, was to minimize the use of progestogen combined with estrogen. The HABITS trial was prematurely stopped in December 2003, because, at a median follow-up of 2.1 years, the risk for recurrence of breast cancer among patients receiving menopausal hormone therapy was statistically significantly higher (relative hazard [RH] = 3.3, 95% confidence interval [CI] = 1.5 to 7.4) than among those receiving no treatment. In the Stockholm trial, however, at a median follow-up of 4.1 years, the risk of breast cancer recurrence was not associated with menopausal hormone therapy (RH = 0.82, 95% CI = 0.35 to 1.9)…Patients who had had a hysterectomy in the menopausal hormone therapy group were given continuous treatment with 2 mg of estradiol valerate daily… In addition, the Stockholm protocol attempted to minimize the use of progestogen in combination with estrogen. In contrast to the HABITS trial, the Stockholm trial recommended that patients avoid continuous combined treatment with estrogen and progestogen and use regimens that incorporated 1 week of no treatment every 1 (cyclic regimen) or 3 (spacing out regimen) months…These treatment recommendations were based on the following results, available when the trial was initiated, that indicated differential effects on the breast when treatment with estrogen alone was compared with combined treatment with estrogen and progestogen. First, breast cell proliferation is increased only during the luteal phase of the menstrual cycle when levels of both estrogen and progesterone are high. Second, using a relevant prospective monkey model for menopausal hormone therapy, we reported statistically significantly higher proliferation in the breast during continuous combined estrogen and progestogen treatment than during estrogen-only treatment (3) . Finally, cyclic discontinuation of hormonal treatment was hypothesized to decrease the expression of local growth factors in breast tissue and to initiate and stimulate apoptosis (4)…In the Stockholm trial, 73% of the women were first assigned to menopausal hormone therapy containing either estrogen alone or the spacing out regimen, in which progestogen was given for only 14 days at 3-month intervals. This protocol could provide one explanation for the lack of difference in breast cancer recurrence between the menopausal hormone therapy group and the no treatment group in the Stockholm trial. (1) (32-33)
In view of the above, I find it astounding that Dr. Bluming was silent on the carcinogenicity of progestins in the HABITS and Stockholm studies. What could be the possible explanation for this? Remember, Dr. Bluming’s area of specialty was oncologist, and as a genreal rule oncologists have no knowledge or interest in prescribing hormone replacement to menopausal women. Most likely, all of his breast cancer patients on hormone replacement were being treated by colleagues in the surrounding community, most of whom were prescribing oral Premarin with medroxyprogesterone (MPA), a synthetic progestin, as this was the standard of care in southern California and still is.
Big Pharma Has Captured the Narrative
Big Pharma has “captured” the regulatory agencies, the media, the medical societies, the medical journals and controls “the narrative”, thus maximizing and protecting profits. In the U.S. Big Pharma has more control, and it is not a good idea for mainstream medical doctors to mention carcinogenicity of progestins in a publication, as this will decrease market share for synthetic progestins. Meanwhile, market share will increase to their main competitor, natural progesterone, resulting in economic loss to Big Pharma. However, in Sweden, outside and distant from U.S. there is more freedom to express an opposing view, without fear of retribution. This could explain the ease with which Dr. Eva von Schoultz discusses the carcinogenicity of synthetic progestins, and Dr. Avrum Bluming’s silence about it. (85-87)
Error in HABITS Study
I noticed a possible error in the report on the 2008 HABITS study by Dr. Lars Holmberg in JCI. The author mistakenly says medroxyprogesterone “resembles natural progesterone”. Perhaps he is referring to the fact that synthesis of medroxyprogestreone uses a progesterone backbone, and the resulting progestin is less androgenic (and less carcinogenic) than the other two, norethisterone and levenorgestrol which are synthesized from a testosterone backbone. Dr. Lars Holmberg writes:
The progestin preferred in the USA is medroxyprogesterone-acetate (MPA), which resembles natural progesterone and is administered either cyclically or continuously. In Europe the predominant regimen prescribed is 17-b-estradiaol opposed by testosterone-derived progestins, mainly norethistosteroneacetate (NETA) or levonorgestrel (LNG); while the less androgenic progestin MPA is used to a lesser extent. (31)
How Not to Do a Hormone Replacement Study
Let’s do a thought experiment on a hypothetical hormone replacement study done by Dr. Evil who wants the results to show HRT increases breast cancer recurrence. This is easy, just use a highly carcinogenic progestin such as norethisterone with a proliferative estrogen, estradiol. This was used in the HABITS trial. What else can Dr. Evil do to increase breast cancer recurrence? Make sure the hormone treated group includes more advanced, more aggressive cancers, as these are associated with a greater recurrence rate. Make sure the HRT group has no long term estrogen deprivation. Make sure most of the HRT group are women who have never been exposed to estrogen, and never been pregnant (nulliporous). Make sure you do not use any agents that target ER-beta or otherwise prevent breast cancer such as Premarin (CEE), estriol (E3) Testosterone (3-beta-Diol), natural progesterone, DIM, and Iodoral. I think you are getting the idea.
Ranking Hormone Formulas as Carcinogenic vs. Breast Cancer Preventive
Most carcinogenic: oral estradiol combined with oral noresthisterone while on tamoxifen. This is from the HABITS trial in which 3-fold greater breast cancer recurrence in the HRT group was found, and all were using tamoxifen, an estrogen blocker. Note: If only the HABITs trial had added testosterone to the hormone cocktail ! The addition of testosterone reverses the proliferation induced by this hormone combination, as shown by Marie Hofling, 2007.
Less Carcinogenic: Oral estradiol plus oral medroxyprogesterone (Dr. Agnes Fournier French Cohort Study showed increased risk of breast cancer (RR=1.69)
Neutral: Oral Premarin plus oral medroxyprogestreone (The corrected 2002 first arm WHI study shows no increased breast cancer, once prior estrogen exposure is removed from placebo group as discussed by Howard Hodis, 2018.)
Preventive: Vaginal Estradiol-Alone (Estradiol is more proliferative than Premarin, binds to ER-alpha and ER-beta with equal affinity)
Preventive: Vaginal Premarin-alone (Unsaturaled B ring steroids bind preferentially to ER-beta making Premarin (CEE) breast cancer preventive, with less proliferative effect than estradiol.)
Most Preventive: Vaginal Biest/Progesterone with topical Testosterone. Biest contains 20% estradiol (E2) and 80% Estriol (E3). The E3 is a pregnancy hormone which has preferential ER-beat activity.) Both progesterone and Testosterone are strongly breast cancer prtective as sicussed above.
Now it has become obvious to the reader that lumping together all hormone formulations into one category, and calling it HRT, is not a good idea, as this blurs the important distinctions between formulations. When reviewing the 26 studies of HRT compiled by Dr. Bluming and even the entire medical literature on hormone replacement, one is struck by the failure to understand the differences between hormone formulations. Instead what we find is a tendency to lump together natural progesterone with synthetic progestins, and to lump together diferent estrogens, Premarin (CEE, horse estrogen), estradiol (E2) and Estriol (E3). Many of the authors of the above 26 studies compiled by Dr. Bluming simply describe the HRT used as estrogen and progestogen, words which lump together different estrogens, and lump together natural progeterone with synthetic progestins. This is so wrong, it makes me very upset. As you have seen with the above discussion, there is considerable variations in breast cancer outcomes depending on exact hormone formulations. (49-54)
Future Studies of Vaginal Estrogen in Breast Cancer Survivors
If any future studies of HRT use in breast cancer survivors are planned, I would suggest a prospective controlled study of a large number of patients, to reach statistical significance This study should use vaginal estriol (E3)/ estradiol (E2) in a 4:1 ratio. This is called Bi-est, first pioneered by Dr. Jonathan Wright. The estriol (E3) binds preferentially to ER-beta, the tumor suppressor receptor. The Bi-est should be combined with natural progesterone also breast cancer preventive, rather than synthetic progestins known to be carcinogenic. (88-90)
The final hormone to add is testosterone which selectively binds to ER-beta and is highly breast cancer preventive. What about providing supplements to the HRT users? Both Iodine and DIM divert estrogen metabolism towards beneficial pathways, and these should be used. Low vitamin D, and low selenium levels are associated with increased cancer risk. Why not check these levels and make sure all patients are given supplements if deficient? This is what we do in our office,
The number of patients should be large enough to reach statistical significance with the data. It is imperative that natural progesterone rather than synthetic progestins are used for all patients regardless of with or without a uterus. Synthetic progestins are known to be carcinogenic and should not be used, especially when natural progesterone is widely available. A prospective controlled trial is suggested because a randomized controlled trial (RCT) may not be feasible because “one cannot offer a placebo for a symptomatic climacteric patient wishing to receive ERT (estrogen replacement therapy)” as stated in 2001 by Dr. Merja Marttunen. (23)
Conclusion: Credit and thanks goes to Dr. Avrum Bluming for compiling the 26 studies of HRT in breast cancer survivors. By reviewing these studies, we have gained a greater understanding of how various HRT components affect breast cancer risk. The first lesson from HABITS and Stockholm studies is to avoid synthetic progestins which are carcinogenic. Use natural progesterone for endometrial protection, instead. Progesterone acts as a proliferative brake on ER-alpha, and is breast cancer preventive. Progestins do the exact opposite. They activate ER alpha and its downstream oncogenes. The second lesson is testosterone selectively binds to ER-beta and is strongly breast cancer preventive. Testosterone is capable of reversing the breast proliferation induced by oral estradiol/norethisteone, the ill-fated combination causing 3-fold increase in breast recurrence in the HABITS trial. The third lesson is the unique safety profile of vaginal estrogen. The fourth lesson is the value of LTED, long term estrogen deprivation. The fifth lesson is the breast cancer protection conferred by high hormone levels of pregnancy or previous estrogen exposure. The sixth lesson is the importance of using agents that bind preferentially to ER-beta, the tumor suppressor, such as Premarin, CEE with its unsaturated B-ring steroids, Estriol, and Testosterone. The seventh lesson is to add DIM and Iodine to divert estrogen metabolism towards beneficial pathways, away from the 4-hydroxy-quinones, and towards 2-methoxy-estradiol. Credit and thanks goes to Dr. Lindsey Berkson for her many HRT insights which were invaluable in writing this article. (47-48) (55-56) (66) (91-93)
Articles with Related Interest:
Hormone Replacement for Breast Cancer Survivvors Part One
Estrogen Metabolism, Iodine, 2MEO Part Three
Testosterone for Breast Cancer Prevention and Treatment
All Bioidentical Hormone Articles
Jeffrey Dach MD
7450 Griffin Road, Suite 190
Davie, Fl 33314
954-792-4663
====================================
References
avrum bluming does HRT cause breast cancer ?
1) Bluming, Avrum Zvi. “Hormone replacement therapy after breast cancer: it is time.” The Cancer Journal 28.3 (2022): 183-190.
2) Fillon, Mike. “The association between menopausal hormone therapy and breast cancer remains unsettled.” CA: A Cancer Journal for Clinicians 74.3 (2024).
In response, a review published in the journal sought to rectify the association between breast cancer and HT—both CEE alone and CEE in combination with MPA, a large source of the misinterpretation (doi:10.1097/GME.0000000000002267).
One of the authors, Avrum Z. Bluming, MD, an oncologist at the Keck School of Medicine at the University of Southern California in Los Angeles, explains it this way:
“According to WHI’s own data, estrogen alone significantly decreases the risk of breast cancer development (by 23%) and the risk of breast cancer death (by 40%)—crucial information for women who have had hysterectomies.” In addition, “when started within 10 years of a woman’s final period (the ‘window of opportunity’), the WHI now agrees,” says Dr Bluming, that “it significantly decreases the risk for coronary heart disease, improves longevity, is the best and safest treatment for menopausal symptoms, and does not increase the risk of stroke. Further, it decreases the risk of osteoporotic hip fracture, colon cancer, and diabetes mellitus.” The sole issue at play is the association between combined HT (CEE plus MPA) and the risk of breast cancer.
Further, even if one were to accept that the WHI’s claims of an increased risk were accurate, that increase would amount to one additional case of breast cancer for every 1,000 women treated per year but no increase in the risk of dying from breast cancer.” In addition, they argue that the assertion from WHI investigators that there is an association between the declining incidence of breast cancer and the reduction in HT prescriptions is not supported by several lines of data, including the fact that the decline in breast cancer incidence in the United States actually predated the release of the WHI’s results.
3) Bluming, Avrum Z., Howard N. Hodis, and Robert D. Langer. “‘Tis but a scratch: a critical review of the Women’s Health Initiative evidence associating menopausal hormone therapy with the risk of breast cancer.” Menopause (2023): 10-1097.
Use of menopausal hormone therapy (HT) fell precipitously after 2002, largely as a result of the Women’s Health Initiative’s report claiming that the combination of conjugated equine estrogen (CEE) and medroxyprogesterone acetate increased breast cancer risk and did not improve quality of life. More recently, Women’s Health Initiative (WHI) publications acknowledge HT as the most effective treatment for managing menopausal vasomotor symptoms and report that CEE alone reduces the risk of breast cancer by 23% while reducing breast cancer death by 40%. Their sole remaining concern is a small increase in breast cancer incidence with CEE and medroxyprogesterone acetate (1 per 1,000 women per year) but with no increased risk of breast cancer mortality. This article closely examines evidence that calls even this claim of breast cancer risk into serious question, including the WHI’s reporting of nonsignificant results as if they were meaningful, a misinterpretation of its own data, and the misleading assertion that the WHI’s findings have reduced the incidence of breast cancer in the United States.
4) Menopause, Breast Cancer, and What Comes Next-a Conversation With Dr. Avrum Bluming. Alloy Staff September 5, 2024 Dr. Corrine Menn, Monica Molinarr
We now know that women who take estrogen have a median increase in longevity of about three and a half years
During the early part of the 2000s, about 44% of menopausal women in the United States were taking hormones. In July of 2002, there was a press release, and then an article published in the Journal of the American Medical Association that claimed that hormone replacement therapy did many, many bad things. Just about every one of those claims has now been walked back. But one of the claims was breast cancer, and even though at the time estrogen was not associated with any significant increased risk of breast cancer, that was the headline in the New York Times. And the rate of prescriptions for hormone replacement therapy fell to 5%, and it remains there today.
there are now 26 studies in the medical literature that I could find in languages that I can understand, and I reviewed all of those studies. Of the 26 studies, 25 show no increased risk of recurrence of breast cancer among women who take hormone replacement therapy of any kind. And that’s independent of whether they had estrogen receptor positive or estrogen receptor negative breast cancer. The one study that suggested an increased risk of recurrence is a study that was done in Sweden called the HABITS Study. HABITS stands for Hormones After Breast Cancer: Is it Safe? And that study reported an increased risk of contra, or local breast cancer, not an increased risk of distant recurrence. And there was no increased risk of death from breast cancer.
26 studies:
4 of 26 show decreased recurrence
1 (Habits) shows increased recurence
the other 21 studies no no increased recurrence.
19 studies looked at ER postive breast or progesterone receptor pos cancers. Thses had no increased risk of recurrence with HRT.No subcategory had increased risk of recurrence.
Pregnancy or IVF (in vitro fertilization) after breast cancer, massive hormone increase – no increased recurrence.
Article: Ann Partridge Harvard 6 year study, women on tamoxifen becasue of ER pos Breast cancer, had permission to stop tamoxifen for two years to get pregnant. 60 percent did get pregnant, and followed for 6 years. These women had no increased risk of recurrence.
About 60 % of breast cancers are ER positive.
Tam was marketed as ER blocker. Compared to estrogen, studies in 1960’s TAM was compared to high dose estrogen, estrogen actually had a better effect on the breast cancer than TAM did. To prevent breast cancer from recurring, estrogen was better than TAM, but is carried adverse effects of fluid retention and bloating, so it was stopped..
TAM works in about 50% of ER pos cases. It will shrink measurable breast cancer.
TAM works in 10 others ways.
When women’s breast cancer progress despite the TAM treatment, giving them estrogen often causes the breast cancer to shrink. (LTED)
When you give TAM to a premenopausal woman, the level of circulating estrogen goes up as much as 5-fold, so it was marketed primarily for post-menopausal women. BUT is is more effective in pre-menopausal women even though it increases estrogen levels 5-fold.
Aromatase Inhibitors.
Women have more testosterone than estrogen. Aromatase converts circulating testosterone into estrogen. Aromatase inhibitors cause estrogen levels to fall. AIs are treatment of choice in post menopausal women where AI’s have a small advantage of TAM in post menopause group..AI’s are maybe 2-6% more effective in Post menopausal women than TAM.
Adding TAM to a woman with suppressed ovarian function (the Shot) improves treastment by about 2-6 percent.
V. Craig Jordan: In pre-menopausal women who get TAM, estrogen levels go up dramatically, with no negative effect on efficacy of TAM to control the breast cancer. Why would it be harmful to give low levels of replacement estrogen to women on TAM?
I know of NO EVidence that transvaginal estrogen compromises anybody’s health while it improves genito-urinary symptoms.
Oncology community has incredible resistance to even vaginal estrogen. Dr Bluming: I cant explain it.
How to talk to your oncologist if not tolerating treatment?:
Who is the best doctor at end of AI therapy ? The responsible person is going to be the oncologist. She would have to go to the oncologist.
The data that estrogen either causes or stimulalates breast cancer growth is not proven, and as far as cause is concerned, is no longer considered valid, and the data don’t support that.
So the thought that even early breast cancer should be treated with TAM for 5 or 10 years, has to be challenged by the patients saying to the doctor, OKAY doctor, I had my lump taken out with or without radiotherapy, or I had my breast taken off, depending on how the primary treatment was done, YOU are now advising me to take TAM or an AI. DOCTOR, I need you to tell me, what is my risk of the cancer coming back if I take nothing, and if I take TAM or AI for 5 or 10 years? And, DOCTOR, what do you think of the new trial NOT TAM 20 mg/d for 5-10 years, but TAM 5 mg/day for 3 years, which at 6 years currently, appears to be equivalent TAM 20 for 5 years?
Oncologists have 2 reasons for refusing HRT to the BC survivor.
1) oncologist is looking out for pt best interest.
2) I may have a recurrence and the doctor is looking out for his best interest. Doctor wants to avoid a lawsuit. Patient can sign an informed consent form to relieve the doctor of legal culpability.
Dr. Bluming has published a prototype consent form.
ASCO says vaginal estrogen is acceptable for the breast cancer survivor.
Premature menopause, early mortality, cardiac disease.
=====================================================
Tamoxifen studies 2023
5) Howell, Anthony, and Sacha J. Howell. “Tamoxifen evolution.” British Journal of Cancer 128.3 (2023): 421-425.
LTED
6) Jordan, V. Craig. “The new biology of estrogen-induced apoptosis applied to treat and prevent breast cancer.” Endocrine-related cancer 22.1 (2015): R1.
Alexander Haddow (2) reported that high doses of synthetic estrogens were able to produce a 30% response rate in women with metastatic breast cancer. High dose estrogen therapy was to remain the standard of care until the introduction of tamoxifen a non-steroidal anti-estrogen, for the treatment of breast cancer in the 1970s (18). But here was a paradox. All the laboratory and clinical evidence suggested that breast cancer was dependent on estrogen for growth but Haddow taught us that estrogen causes tumor regression!
The first clinical trial published in 1944(2), is summarized in Table 1. Responses were consistent at about 30% but for less than a year. These preliminary data showed that both breast and prostate cancer were responsive but no other tumor types responded.
The results were clear; a period of 5 years post menopause was necessary for optimal antitumor action with high dose estrogen for breast cancer treatment. All clinical results were, therefore, consistent – a period of time after the menopause was necessary to expose the effectiveness of high dose estrogen as an anticancer agent for metastatic breast cancer.
Early clinical trials of the treatment of metastatic breast cancer in postmenopausal women showed similar response rates and durations of responsiveness as DES, but with fewer side effects than high doses of estrogen (3, 40). However, it was noted that reanalysis of a randomized trial of DES vs. tamoxifen (3) demonstrated that patients treated with DES had a more prolonged survival when compared to those treated with tamoxifen (41).
The successful use of high dose synthetic estrogens to treat post-menopausal metastatic breast cancer, is the first effective “chemical therapy” proven in clinical trial to treat any cancer. This review documents the clinical use of estrogen for breast cancer treatment or estrogen replacement therapy (ERT) for postmenopausal hysterectomized women which can either result in breast cancer cell growth or breast cancer regression. This has remained a paradox since the 1950s until the discovery of the new biology of estrogen induced apoptosis at the end of the 20th century. The key to triggering apoptosis with estrogen is the selection of breast cancer cell populations that are resistant to long term estrogen deprivation.
7) Ingle, James N., et al. “Randomized clinical trial of diethylstilbestrol versus tamoxifen in postmenopausal women with advanced breast cancer.” New England Journal of Medicine 304.1 (1981): 16-21.
Before the introduction of tamoxifen, diethylstilbestrol (DES) was widely considered to be the hormonal treatment of choice in postmenopausal women with advanced breast cancer. We performed a randomized clinical trial of these two agents to determine their relative efficacy and toxicity. The trial involved 143 evaluable patients, of whom 99 had received no prior systemic therapy and 44 had received previous chemotherapy. The regression rates (complete plus partial) were higher in patients receiving DES (41 per cent) than in those receiving tamoxifen (33 per cent), but not significantly so (P = 0.37). (note P value of .05 is needed) In patients who had had no prior systemic therapy, the rates were 44 per cent and 38 per cent, respectively (P = 0.55), and in those who had had previous chemotherapy, 32 per cent vs. 23 per cent (P = 0.50). Analysis of the time until treatment failure for the two treatment groups showed no significant difference (medians: DES, 142 days; tamoxifen, 171 days). Toxicity was greater in patients receiving DES; nine of 74 patients (12 per cent) discontinued therapy solely because of adverse reactions. Since there was no statistically significant difference in efficacy and since tamoxifen was less toxic, tamoxifen appears to be the preferred agent.
143 pts. RCT w/ progressive metastatic breast cancer RX DES vs. TAM
Survival was significantly better for women on DES than for women on TAM (adjusted p = 0.039).
Duration of response and progression-free survival were not found to be significantly different between DES and TAM (p = 0.32 and 0.65, respectively).
8) Peethambaram, Prema P., et al. “Randomized trial of diethylstilbestrol vs. tamoxifen in postmenopausal women with metastatic breast cancer. An updated analysis.” Breast Cancer Research and Treatment 54 (1999): 117-122.
One hundred fifty-one postmenopausal women with progressive metastatic breast cancer and no prior hormonal therapy were treated with either diethylstilbestrol (DES) or tamoxifen (TAM). One hundred forty-three eligible patients were followed until death or for a minimum of 14.1 years on the DES arm or 16.7 years on the TAM arm. The overall objective response was 42% for DES and 33% for TAM (p = 0.31) and the median duration of response was 11.8 months for DES and 9.9 months for TAM (p = 0.38). Duration of response and progression-free survival were not found to be significantly different between DES and TAM (p = 0.32 and 0.65, respectively). The median survival was 3.0 years for DES vs. 2.4 years for TAM. The 5-year survival was 35% for the DES arm and 16% for the TAM arm. Survival was significantly better for women on DES than for women on TAM (adjusted p = 0.039).
Review of records did not show any difference in pattern of treatment failure or subsequent treatments in the DES and TAM arms. Treatment with DES was more commonly associated with toxicity such as nausea, edema, vaginal bleeding, and cardiac problems, whereas hot flashes were commonly seen with TAM therapy. The initial treatment with DES is associated with increased survival. The basis of this survival advantage is not known. TAM still is the preferred agent in the treatment of metastatic breast cancer, but this trial underscores the fact that estrogens have activity and remain in the armamentarium for treatment of selected patients with metastatic breast cancer.
======================================
26 Studies of HRT in Breast Cancer Survivors
======================================
Premarin CEE Only 39 pts.
LTED 40 months
Recurrence rate for HRT half that of non-user controls.
9) Vassilopoulou-Sellin, Rena, et al. “Estrogen replacement therapy after localized breast cancer: clinical outcome of 319 women followed prospectively.” Journal of clinical oncology 17.5 (1999): 1482-1482.
Median DFI at entry was 114 months (range, 24 to 234 months), and median observation duration was 40 months (range, 24 to 99 months).
Therefore, a total of 39 women were administered ERT (ERT group), consisting of conjugated estrogens (0.625 mg on days 1 to 25 of each month) without progesterone. The other 280 women (control group) did not take hormones.
1/39 (2.6%) recurrence Premarin-alone
14/280 (5.0%) recurrence NO HRT COntrols.
observed prospectively for at least 2 years whether they enrolled onto the randomized trial or not.
Therefore, a total of 39 women were administered ERT (ERT group), consisting of conjugated estrogens (0.625 mg on days 1 to 25 of each month) without progesterone. The other 280 women (control group) did not take hormones.
In the present study, there were no excess events among the women taking ERT. One patient (2.6%) in the ERT group developed a new breast cancer whereas 14 patients (5.0%) in the control group developed new or recurrent breast cancer. Given the small number of patients (n 5 319) and the small number of all events in both groups (n 5 20 in the control group and n 5 1 in the ERT group),
=================================================
full pdf
50 reduction in Breast Cancer Recurrence and Total Mortality Reduced in HRT Users
62.5 % Reduction in Mortality from breast cancer for HRT users.
Five HRT users (3%) and 59 nonusers (8%) died of breast cancer during followup
175 HRT users who appeared to be free of breast cancer disease. half 51% had hysterectomy. 68% had used HRT before Dx of breast cancer.
695 non-users
79% unopposed estrogen-alone (62% oral, 38% vaginal) x 15 months
(50% Premarin CEE 50% estradiol)
21 % progestogen added
Women were followed for a median of 3.7 years for recurrence and 4.6 years for mortality.
The median duration of oral HRT use during follow-up was 15 months.
LTED :Time from diagnosis to reference, years
years users nonusers
<1 47 (27) 188 (27)
1–2 46 (26) 184 (26)
3–7 44 (25) 175 (25)
8 37 (21) 148 (21)
About half under 2 years, half over 2 years
Breast cancer recurrence was diagnosed in 16 HRT users (9%) and in 101 nonusers (15%) (Table 4). 40% Less recurrence in users.
The rate of recurrence was 17 per 1000 person-years in users (95% CI 11 to 29) and 30 per
1000 person-years in nonusers (95% CI 25 to 37). (43% less for users)
10) O’Meara, Ellen S., et al. “Hormone replacement therapy after a diagnosis of breast cancer in relation to recurrence and mortality.” Journal of the National Cancer Institute 93.10 (2001): 754-761. Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle;
Retrospective records study. excluded: in-situ cancer and metastatic cancer. Recurrence was defined as invasive disease confined to the ipsilateral breast (22% of diagnosed recurrences) or metastasis. New primary contralateral breast cancers were not included
in this definition.
The cohort comprised 2755 women who met these criteria.
This record-based study 174 users and 695 nonusers of hormone replacement therapy (HRT) after a diagnosis of breast cancer
The median duration of oral HRT use during follow-up was 15 months. Estrogens were unopposed by progestogens for 79% of the users; the rest combined a progestogen with estrogen for at least one monthly cycle.
The rate of breast cancer recurrence was 17 per 1000 person-years in women who used HRT after diagnosis and 30 per 1000 person-years in nonusers (adjusted relative risk for users compared with nonusers = 0.50; 95% confidence interval [CI] = 0.30 to 0.85).
Comparison of rates yielded an unadjusted relative risk associated with ever use of HRT after breast cancer of 0.58 (95% CI 0.34 to 0.98). Adjusting for bilateral oophorectomy, hysterectomy, mastectomy, tamoxifen, and the matching variables in a Cox model resulted in a relative risk of 0.50 (95% CI 0.30 to 0.85). Further adjustments did not appreciably
change the results. The relative risk was similar when restricting to users of unopposed estrogens only.
Breast cancer mortality rates were five per 1000 person-years in HRT users and 15 per 1000 person-years in nonusers (adjusted relative risk = 0.34; 95% CI = 0.13 to 0.91).
Total mortality rates were 16 per 1000 person-years in HRT users and 30 per 1000 person-years in nonusers (adjusted relative risk = 0.48; 95% CI = 0.29 to 0.78).
Route of HRT administration Estrogen type No. of prescriptions* (% of total)
Oral 1182 ( 62)
Conjugated 562
Esterified 543 Estratab and Menest
Ethinyl estradiol 77
Vaginal 728 (38)
Conjugated 442
Dienestrol 286
Topical Estradiol 3 (<1)
Total 1913 (100)
Estrogens can stimulate the growth of breast cancer cells in tissue culture at low doses
but can inhibit growth at high doses (23).
Breast tumors can regulate and maintain internal levels of estradiol independent of levels outside the tumor (24), so exogenous estrogens may have relatively little effect on tumor growth (10).
============
Why did not Dr Bluming mark this one along with HABITS as increased recurrence?
BAD OUTCOME (estrdiol p[lus progestin) I would assume the progestin was highly carcinogenic (norethisterone, like the HABITS)
Abstract only
ALL 21 PATIENTS USED ESTRADIOL. plus progestin (was it norethisterone?)..
LTED OF 5 YEARS
ALMOST DOUBLE (1.7X) RELAPSE RATE for HRT GROUP at 28 months
11) Uršič-Vrščaj, Marjetka, and Sonja Bebar. “A case-control study of hormone replacement therapy after primary surgical breast cancer treatment.” European Journal of Surgical Oncology (EJSO) 25.2 (1999): 146-151.
Department of Gynecological Oncology, Institute of Oncology, Ljubljana, Slovenia
HRT was started on average 62 months (range 1-180 months) after diagnosis, and lasted an average of 28 months (range 3-72 months).
All 21 patients used estradiol as HRT, i.e. a non-conjugated estrogen. Combined hormonal therapy (estrogens + progestagens) was given to 95% of patients with median age of 47 years (range 41-59 years) at the beginning of HRT.
Relapse was observed in four patients (19%) of the HRT group; of these, one had a carcinoma of the contralateral breast. In the control group, relapse was observed in five patients (11%); one of these five patients had a carcinoma of the contralateral breast. In the HRT group, there were no deaths among the patients with confirmed relapse, while one patient died in the control group.
The estimated risk (OR= 1.74, 95%S CI 0.34-8.88) of relapse of breast cancer was calculated by comparing data from HRT users, who had received HRT for 28 months (range 3-72 months) on average, with data from the control group.
The estimated risk of breast cancer relapse in HRT users who had been receiving HRT for less than 24 months was 0.65 (OR = 0.65, 95% CI 0.02-7.85).
All 21 patients used estradiol as HRT, i.e. a non-conjugated oestrogen. Combined hormonal therapy (estrogens + progestagens) was given to 95% of patients with median age of 47 years (range 41–59 years) at the beginning of HRT.
Relapse was observed in four patients (19%) of the HRT group; of these, one had a carcinoma of the contralateral breast.
In the control group, relapse was observed in five patients (11%); one of these five patients had a carcinoma of the contralateral breast. In the HRT group, there were no deaths among the patients with confirmed relapse, while one patient died in the control group. The estimated risk (OR=1.74, 95% CI 0.34–8.88) of relapse of breast cancer was calculated by comparing data from HRT users, who had received HRT for 28 months (range 3–72 months) on average, with data from the control group. The estimated risk of breast cancer relapse in HRT users who had been receiving HRT for less than 24 months was 0.65 (OR=0.65, 95% CI 0.02–
====================================
Poor Outcome OR=1.7 for recurrence for HRT USERS (Premarin+Medroxyprogesterone)
LTED?
Prior Pregnancy?
Very Careful In Person Interview to determine type of HRT used.
CEE with medroxyprogesterone – OR=1.7 for recurrence in HRT Users!!!!
Cee alone- 30% reduction in Recurrence of advanced disease stage, later stage disease ? only the tumors of patients who used ERT retained a substantive reduced relative odds of advanced disease stage [OR 0.7 (95% CI, 0.6–0.9)].
12) Daling, Janet R., et al. “Association of regimens of hormone replacement therapy to prognostic factors among women diagnosed with breast cancer aged 50–64 years.” Cancer Epidemiology Biomarkers & Prevention 12.11 (2003): 1175-1181.
Fred Hutchinson Cancer Research Center, Division of Public Health Sciences,
Seattle, Washington; 2
Women’s CARE Study: multicenter population-based case-control study of invasive breast cancer was conducted. five geographic areas: Atlanta; Detroit; Los Angeles; Philadelphia; and Seattle.
In-person interviewscollected adetailed history of all episodes of hormone use.
2346 women with diagnosed with invasive breast cancer.
In this study, 84.0% of estrogen use was conjugated estrogen, and 94.0% of progestin use was medroxyprogesterone acetate
The tumors of cases who used each regimen of HRT were smaller and of earlier stage than those of non-HRT users.
and only cases who used estrogen alone(estrogen replacement therapy) had reduced odds of being diagnosed with later-stage disease (regional or distant) than cases who never used HRT (OR, 0.7; 95% CI, 0.6–0.9).
Accumulating evidence indicates a more favorable tumor biology
among women who develop breast cancer who are either current or past users of HRT.11
11.Chen, C-L., Weiss, N. S., Newcomb, P., Barlow, W., and White, E. Hormone
replacement therapy in relation to breast cancer. J. Am. Med. Assoc., 287:
734–747, 2002.
70 % Greater Recurrence Using C-CHRT
Among women with breast cancer who had used C-CHRT [Continuous Combined HRT],
20.5% developed a tumor of either lobular or mixed lobular/ductal histology compared with 12.1% of women who had never used any HRT (OR, 1.7; 95% CI, 1.2–2.4; Tables 1 and
2).
ERT users were at 30% reduced odds of being diagnosed with their cancer at later stage (regional or distant) relative to nonusers. Ross et al. (19), whose study included in situ cancers, found that ERT was inversely related to the risk of developing these late-stage
tumors but found no effect of the regimen of CHRT on stage of disease. Magnusson et al. (5) found CHRT was related to reduced risk of developing larger and later stage tumors; however, they made no adjustment for screening practices.
The pooled analysis of 51 epidemiological studies (1) and a number of recent studies indicate CHRT is related to an increased risk of developing breast cancer (9, 11, 12, 15, 17–21).
====================================================
TESTOSTERONE ADDED !!! Much Better Results
Mortality of estrogen (estradiol) HRT users was (3 of 50) 6% (81% reduction in Mortality)
One died of MI.
Mortality of Non-users of HRT (6 of18) women 33%
76 pts, 50 on HRT, of the 50, 40 (80%) were also given testosterone, 50% pellets
LTED of the 6 deaths in HRT :
1 pt = 2 years
2 pts 5 years
Prior Pregnancy or Prior Hormone USe ? There were 3 nulliparous women (6%) in this group (on HRT after Breast Cancer), and the mean parity was 2.2 ± 1.17.
Non-Hormone Users: There were 3 nulliparous women (16.7%) in this group,
and the mean parity was 1.7 ± 1.02.
In the 50 hormone users, 45 were also given progestogens, mostly megestrol acetate.
Willemse, P. H., et al. “A randomized comparison of megestrol acetate (MA) and medroxyprogesterone acetate (MPA) in patients with advanced breast cancer.” European journal of cancer (Oxford, England: 1990) 26.3 (1990): 337-343.
13) Natrajan, Puthgraman K., Kostas Soumakis, and R. Don Gambrell Jr. “Estrogen replacement therapy in women with previous breast cancer.” American journal of obstetrics and gynecology 181.2 (1999): 288-295.
Augusta, Georgia
The study group consisted of 76 patients with breast cancer, including 50 using estrogen
replacement for up to 32 years, 8 using nonestrogenic hormone replacement for up to 6 years and followed for up to 11 years, and 18 using no hormones for up to 10 years. In addition to estrogen use, 40 of the 50 hormone users were treated with androgens, usually in the form of implantation of testosterone pellets. Forty-five subjects were also given progestogens, usually megestrol acetate 20 to 40 mg for 10 to 25 days each month. The 8 nonestrogen hormone users were treated with various combinations of testosterone pellets, tamoxifen, and progestogens.
Therapy No. of patients Range Mean ± SEM (y)
Testosterone 150 mg, estradiol 25 mg, progestogen 19 6 mo–24 y 5.7 ± 2.36
Testosterone 150 mg, estradiol 50 mg, progestogen 8 2.5-7 y 3.7 ± 1.16
Oral estradiol, progestogen 4 6 mo–2 y 1.0 ± 0.78
Testosterone 75 mg, estradiol 25 mg, progestogen 3 1.5-9 y 4.5 ± 1.80
Oral estrogen, testosterone, progestogen 3 1.5-3.5 y 2.3 ± 0.92
Testosterone 225 mg, estradiol 50 mg, progestogen 2 3-4.5 y 3.5 ± 0.71
Transdermal estrogen, progestogen 4 2-5 y 3.2 ± 1.14
Testosterone 75 mg, estradiol 50 mg 2 16-17 y 16.5 ± 0.71
Testosterone 75 mg, estradiol 25 mg 1 32 y —
Testosterone 150 mg, estradiol 50 mg 1 21.5 y —
Estradiol 75 mg, progestogen 1 2 y —
Testosterone 225 mg, estradiol 75 mg, progestogen 1 1.5 y —
Oral estradiol 1 5 y —
TOTAL 50 6 mo–32 y 5.5 ± 2.53
The mortality rate among the estrogen users was 6%. Of the 3 deaths, 2 patients died of breast cancer. mean duration of follow-up of 83.3 ± 8.81 months.
Of the nonestrogenic HRT group, 1 patient died of breast cancer,for a mortality of 12.5%.
In the group not receiving hormonal replacement, 33% have died. Five of the 6 died of breast cancer
Testosterone and estradiol pellets were implanted every 4.5 to 6 months in 38 of the 50 women (76%), and 45 of the 50 women (90%) were given oral progestogen. The
most frequently used progestogen was megestrol acetate 20 to 40 mg given from 10 to 25 days each month to 29 of the 45 progestogen users (64%). Medroxyprogesterone
acetate 10 mg from 10 to 13 days was given to 8 patients, and norethindrone acetate 2.5 to 5 mg was given to the other 8 patients along with the estrogen.
during the 1990s, megestrol acetate was increasingly used as the progestogen of choice
because it was found to be more acceptable to the oncologists than either medroxyprogesterone acetate or norethindrone acetate.
===============================================
abstract only
HRT: Premarin/MPA same as WHI 2002 first arm.
No Control Group for Comparison
LTED: 41 months…41 percent started HRT within 3 year of diagnosis of Br CA
HRT: premarin/MPA , 9 percent recurrence rate.
14) Brewster, Wendy R., et al. “An experience with estrogen replacement therapy in breast cancer survivors.” International journal of fertility and women’s medicine 44.4 (1999): 186-192. University of California Irvine Medical Center, Orange 92868,
Breast cancer survivors who elected ERT received the preferred regimen of conjugated estrogen 0.625 mg/day with medroxyprogesterone acetate 2.5 mg/day.
145 patients received ERT for at least 3 months. Thirteen recurrences (9%) were identified; 10 are alive with disease, 3 are dead of disease. The median interval between diagnosis and commencement of ERT was 41 months. Forty-one percent of the study group initiated ERT within 3 years of their breast cancer diagnosis. The median duration of follow-up on ERT was 30 months.
==========================================================
abstract only
277 pts on HRT.
LTED 3.6 years,
recurrence rate same for HRT vs non-HRT
HRT Mortality half that of control group.
HRT formula? not mentioned
There was no significant difference between the ERT and control groups in ipsilateral primary/recurrence (5/155 v 5/143; P = 0.85), contralateral breast cancers (10/258 v 9/260; P = 0.99), or systemic metastasis (8/277 v 15/277; P = 0.13). Noncause-specific deaths in the control group numbered 15 (of 277), and in the ERT group, 7 (of 277) (P = 0.03). Overall survival favored the ERT group (P = 0.02).
Type of estrogen ? US study- probably premarin ???
Department of Medicine, William Beaumont Hospital, Royal Oak-Troy, MI
15) Decker, David A., et al. “Estrogen replacement therapy in breast cancer survivors: a matched-controlled series.” Menopause 10.4 (2003): 277-285.
Design: Two hundred seventy-seven disease-free survivors received ERT. Controls were matched for exact stage, a recurrence-free period similar to the period to ERT initiation in the ERT group, approximate age, and duration of follow-up. The mean time from breast cancer diagnosis to initiation of ERT was 3.61 (+/- 0.25) years, with a median of 1.88 years. The mean duration of ERT was 3.7 (+/- 3.01) years, with a median of 3.05 years.
Results: Hot flashes were relieved in 206 of 223 women (92%), dyspareunia/vaginal dryness in 149 of 167 women (89%), and reactive depression/anxiety/mood change in 111 of 126 women (88%). Univariate analysis demonstrated no statistical differences between the groups for age, stage, pathology at diagnosis, progesterone receptor status, local therapy, breast at risk, prior chemotherapy, and duration of follow-up. The ERT group was more likely to be estrogen receptor negative (P = 0.01), to have received prior ERT (P < 0.001), and to have received no adjuvant tamoxifen (P < 0.001).
There was no significant difference between the ERT and control groups in ipsilateral primary/recurrence (5/155 v 5/143; P = 0.85), contralateral breast cancers (10/258 v 9/260; P = 0.99), or systemic metastasis (8/277 v 15/277; P = 0.13). Noncause-specific deaths in the control group numbered 15 (of 277), and in the ERT group, 7 (of 277) (P = 0.03). Overall survival favored the ERT group (P = 0.02).
Conclusions: In these selected patients, ERT relieved estrogen deficiency symptoms and did not increase the rate or time to an ipsilateral recurrence/new primary, contralateral new primary, local-regional recurrence, or systemic metastases.
=====================================================================
full pdf
Mostly CEE (44/56 patients)
Excellent Results: 2 local recurences after 13.5 and 9.6 years, no deaths.
One local recurrence and one contralateral breast cancer occurred during the follow-up period (13.5 and 9.6 years, respectively), with no regional or distant recurrences, for a 15-year actuarial disease-free survival rate of 92.5%. There were no breast cancer deaths.
16) Peters, George N., et al. “Estrogen replacement therapy after breast cancer: a 12-year follow-up.” Annals of surgical oncology 8 (2001): 828-832.
From The Center for Breast Care, The University of Texas Southwestern Medical Center at Dallas (GNP, TF, DE), and Baylor University Medical Center, Dallas, Texas (JS, SJ).
LTED: DFI (DISEASE FREE INTERVAL) at entry (initiation of ERT) 3.9 years ,,,….median
disease-free interval before initiation of ERT was 46.7 months (range, 0–448 months).
607 breast cancer survivors, 64 on HRT
The median follow- up from diagnosis was 12.8 years (range, 4.7–38.9
years), and the median number of years on ERT since breast cancer diagnosis was 6.4 years (range, 1.0–20.9 years) (Table 2).
The remaining 56 received ERT as conjugated estrogens, an estradiol patch, estropipate, or birth control pills.
After initial identification of breast cancer patients using ERT, their clinical outcome was prospectively observed for a minimum of 60 months.
44/56 on HRT
24 RECEIVED CEE ALONE
20 CEE + MPA
10 Estradiol
LTED: 38% of the patients initiated ERT within 2 years of diagnosis.
52% after 2 years.
Half received adjuvant or chemo.
Conjugated estrogens 24
Conjugated estrogen and medroxyprogesterone acetate 20
Estradiol 10
Estropipate 1
Birth control pills 1
Methods: From February to August 1995, 607 breast cancer survivors were interviewed concerning ERT usage.
Sixty-four patients indicated they received some form of ERT after their breast
cancer diagnosis. Medical records for these patients were analyzed for disease stage, surgical treatment, adjuvant treatment, estrogen and progesterone receptor status, date of initiation of ERT, type of ERT, recurrence, and final outcome. Patients receiving ERT were followed prospectively.
Results: Eight patients were excluded because they had used only vaginal cream ERT.
The remaining 56 received ERT as conjugated estrogens, an estradiol patch, estropipate, or birth control pills. The median follow-up from diagnosis was 12.8 years (range, 4.7–38.9 years). The median time on ERT since diagnosis was 6.4 years (range, 1.0 –20.9 years); 38% of the patients initiated ERT within 2 years of diagnosis. Estrogen receptors were positive in 28 (74%) of the 38 cases with available information. Pathological disease stage at time of diagnosis and treatment was 0 in 15 cases (27%), I in 27 (48%), and II in 14 (25%).
Twenty-six patients (47%) received adjuvant chemotherapy or hormonal therapy.
One local recurrence 13.7 years after initial diagnosis of her breast cancer. She had been on ERT for 3.8 years at the time of local recurrence. She is now disease-free at 3 years after treatment of her local recurrence and not on ERT.
and one contralateral breast cancer (DCIS) occurred during the follow-up period (13.5 and 9.6 years, respectively), with no regional or distant recurrences, for a 15-year actuarial disease-free survival rate of 92.5%. There were no breast cancer deaths.
Only two patients (4%) developed either recurrent or new disease. To date, our study provides the longest follow-up of breast cancer survivors on ERT with the longest duration of ERT usage.
Comparison Group
Twelve-year follow-up data of the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-06 protocol provides the only comparable results for our ERT
group. The estimate of overall survival at 12 years is 63%; disease-free survival is 50%; and local recurrence for the group treated by lumpectomy and breast irradiation
is 10%.32
For comparison, disease-free survival was calculated for a cohort of 74 stage 0-II breast cancer patients from our practice who were diagnosed and treated in 1996.
These women ranged in age from 36 to 82 years (mean, 61 years). There were 9 stage 0 (12%), 40 stage I (54%), and 25 stage II (34%) patients. After a median follow-up
of 37 months, disease-free survival was 67% for the 1996 cohort as compared to 100% for the ERT cohort (P .029, Fig. 1). Because half of the patients in our group
started ERT nearly 4 years after diagnosis, our ERT patients are likely to be self-selected for good-prognosis tumors, because patients with poor-prognosis tumors would have died of breast cancer earlier, in the first few years after diagnosis.
USE of Vaginal Creams
Of the eight patients who have used only vaginal cream ERT, median follow-up from diagnosis was 11.4 years, and median time on ERT since diagnosis was 4.0
years. There have been no contralateral breast cancers; no local, regional, or distant recurrences; and no cancer deaths in this group.
Conclusions: Use of ERT in a cohort of breast cancer survivors with tumors of generally good prognosis was not associated with increased breast cancer events compared with non-ERT users, even over a long follow-up period.
Two centuries ago, less than 30% of women lived long enough to
reach menopause, but today 90% of women reach theclimacteric. In the United States, more than 30 million women have an average postmenopausal life expectancy
of 28 years.1 Marchant DJ. Estrogen-replacement therapy after breast cancer.
Risks versus benefits. Cancer 1993;71(6 Suppl):2169–76.
With an average age at diagnosis of 60 years, coupled with a 25-year expected
survival, it is estimated that the number of breast cancer survivors in the United States is nearly 2.5 million women. The short-term effects of estrogen deficiency (e.g., vasomotor instability and urogenital atrophy) and long-term consequences (e.g., osteoporosis and cardiovascular disease) are important quality-of-life and health issues for breast cancer survivors.10
However, prior studies have documented that up to 10% of breast cancer survivors use ERT for relief of menopausal symptoms.11
=========================================================
abstract only
230 pts prospective study- many of these patients had a premature, artificial, chemo-induced menopause.
LTED at least 2 years
(estradiol + a progestin compound) given either continuously or with a 5-d interruption each month. The mean duration of treatment was 2.5 years.
17 (7 percent) of cases recurrence (relapse) (seven local, six in the contro-lateral breast and four metastases (7%)).
17) Gorins A, Espié M, Bedairia N, et al. Hormone replacement therapy in breast cancer patients: a study of 230 patients, with a case-control study. Gynecol Obstet Fertil. 2003;31:614–619.
Center of Breast Diseases in Saint-Louis hospital in Paris France.
Objectives: After recalling the classical contra-indication of hormone replacement therapy (HRT) concerning patients with a personal history of breast cancer (BC), and arguments that may be opposed, the authors report the present results of a prospective study undertaken in the Center of Breast Diseases in Saint-Louis hospital in Paris since February 1992.
Patients and methods: By April 2001, 230 patients had been included. A free interval of 2 years at least since the treatment of the primary BC has been observed. The reasons for prescribing HRT were vasomotor troubles (flushes, nightly sweats) or a dyspareunia, which were severe and not controlled by non-hormonal treatments. There was also an indication of a major osteoporotic or cardiovascular danger. In fact, many of these patients had a premature, artificial, chemo-induced menopause. The HRT most often used was an estro-progestin association (estradiol + a progestin compound) given either continuously or with a 5-d interruption each month. The mean duration of treatment was 2.5 years.
Results: Results, concerning the improvement of menopause troubles, were remarkable in the great majority of troubles. HRT had to be stopped in 39 cases, reading as follows: 17 cases for relapses (seven local, six in the contro-lateral breast and four metastases (7%)). Also, 22 patients (9%) interrupted their HRT for serious side-effects. A case-control study did not show any significant difference between with and without HRT patients concerning the overall survival without relapse.
Discussion and conclusions: Quality of life of patients was often substantially improved, and a deleterious effect on the cancer disease was not found. Our results are in agreement with the literature from other countries. However, one must be cautious. In such circumstances, HRT must be prescribed with the informed consent of the patients and delivered in appropriate hospital and university centers. It is wished that large randomised prospective studies may be undertaken.
=====================================================================
Abstract only
33 percent reduction in recurrence in HRT users.
15% reduction in progestin -alone users
continuous combined estrogen/progestogen hormonal therapy
167 subjects had used an oral or transdermal estrogen
hazard ratio for the estrogen-progestin users was 0.67 (95% confidence interval (CI) 0.38-1.16) and for the progestin alone users was 0.85(95% CI 0.44-1.65).
152 (91%) had also used a progestin
18) Dew, J., et al. “A cohort study of hormone replacement therapy given to women previously treated for breast cancer.” Climacteric: the journal of the International Menopause Society 1.2 (1998): 137-142.
Women’s Health Institute, Royal Hospital for Women, Barker Street, Randwick, NSW 2031, Australia.
Women who have been previously treated for breast cancer are usually advised to avoid hormone therapy for fear of increasing their risk of tumor recurrence. However, for some women, menopausal symptoms are so severe that their quality of life is poor. Because ethic committees are reticent to permit a double-blind randomized trial, we performed a cohort study of hormone therapy after breast cancer.
Methods: The study group comprised 1472 women with breast cancer. A total of 167 subjects had used an oral or transdermal estrogen after their treatment for breast cancer. Amongst these estrogen users, 152 (91%) had also used a progestin. In total, 106 other women had used a progestin alone as a treatment for menopausal flushes and not as a treatment for breast cancer. Cox regression analysis was performed using estrogen as a time-dependent covariate with disease-free interval as the outcome.
Results: The uncorrected hazard ratio for the estrogen-progestin users was 0.67(95% confidence interval (CI) 0.38-1.16) and for the progestin alone users was 0.85 (95% CI 0.44-1.65).
Conclusions: This study was unable to demonstrate a significant increase in risk of breast cancer recurrence for women who used HRT and suggests that the time is now appropriate for a randomized prospective trial of hormone therapy after breast cancer.
=================================================
abstract only : Same study as Eden (20)
LTED ?
Exact HRT formula ?
Length of follow up?
Most were using combined continuous estrogen-progestin therapy, usually an oral estrogen with a moderate dosage progestin
120 patients, 90 estrogen users
Among the 90 estrogen users, there were no deaths and only 7% developed a recurrence, compared to 17% of the nonusers (using two matched controls);
RR = 0.40 (95% CI 0.17–0.93).
19) Espie, M., A. Gorins, and F. Perret. “Hormone replacement therapy (HRT) in patients treated for breast cancer: analysis of a cohort of 120 patients.” Proc Am Soc Clin Oncol. Vol. 18. 1999.
Ninety had taken estrogen for relief of severe menopausal symptoms after their diagnosis and treatment of breast cancer. Most were using combined continuous estrogen-progestin therapy, usually an oral estrogen with a moderate dosage progestin. Controls were matched subjects from the same database who had not taken sex steroids after their diagnosis of cancer. The main outcome measures were all-cause mortality and recurrence of breast cancer (or new contralateral breast cancer). Relative risks (RR) were then calculated comparing sex-hormone users with matched controls.
Among the 90 estrogen users, there were no deaths and only 7% developed a recurrence, compared to 17% of the nonusers (using two matched controls); RR = 0.40 (95% CI 0.17–0.93).
These results suggest that short-term usage of combined continuous hormone replacement therapy (HRT) by women with a personal history of breast cancer may be safe and might even reduce the risk of recurrence. A formal prospective double-blind controlled study is needed to confirm these results.
==========================
Abstract only / SAME as 19) Espie?
Premarin/MPA (Assumed because of the good results) 90 women
LTED ???? not disclosed in abstract
20) Eden, John A., et al. “A case-control study of combined continuous estrogen—progestin replacement therapy among women with a personal history of breast cancer.” Menopause 2.2 (1995): 67-72. New South Wales, Australia;
combined continuous estrogen-progestin therapy, usually an oral estrogen with a moderate dosage progestin.
continuous estrogen-progestin therapy given to symptomatic menopausal women with a personal history of breast cancer.
901 women with surgically confirmed breast cancer attending one of three teaching hospitals in south-eastern Sydney, Australia. Ninety had taken estrogen for relief of severe menopausal symptoms after their diagnosis and treatment of breast cancer. Most were using combined continuous estrogen-progestin therapy, usually an oral estrogen with a moderate dosage progestin.
Among the 90 estrogen users, there were no deaths and only 7% developed a recurrence, compared to 17% of the nonusers (using two matched controls); RR = 0.40 (95% CI 0.17–0.93).
===================================
FULL PDF
This Durna study is continuation of study group data from Eden.
retrospective observational study of breast cancer recurrence and mortality among postmenopausal women with histopathologically confirmed breast cancer
Only study so far to include vaginal estriol cream. !!!!
Parity the same for HRT and NON-HRT (N=2.2)
HRT use before diagnosis
Prior Use of HRT:
Number of women NO HRT = 683 127 (19%)
HRT = 240 161 (67%) <0.001
those who used vaginal estrogen alone [32 patients] had a significantly lower risk of recurrence or new breast cancer (adjusted RR, 0.18; 95% CI, 0.04–0.75)
Of the 1122 women in the study, 286 used HRT after breast cancer diagnosis to treat menopausal symptoms.
A total of 1122 women met the entry criteria and were followed up for a median of 6.08 years (range, 0 to 36 /years).
half used Premarin plus Progestin
LTED = one year. Years from diagnosis to HRT start
(median, range) n=258 1.0 years (0–23)
20) Durna, Eva M., et al. “Hormone replacement therapy after a diagnosis of breast cancer: Cancer recurrence and mortality.” Medical Journal of Australia 177.7 (2002): 347-351.
Sydney, New South Wales Australia
retrospective observational study of breast cancer recurrence and mortality among postmenopausal women with histopathologically confirmed breast cancer.
three surgeons and two gynaecologists at three tertiary hospitals in Sydney, New South Wales
286 used HRT
Types of HRT used after breast cancer diagnosis were
continuous combined oestrogen plus progestin (138; 48%),
oral progestin alone (78; 27%),
vaginal oestrogen alone (32; 11%),
vaginal oestrogen plus oral progestin (21; 7%), and
oral or transdermal oestrogen alone (17; 6%).
Tumour size was significant to all outcomes, with larger tumours associated
with higher risk of recurrence and death.
Those who used HRT after diagnosis were significantly younger when diagnosed with breast cancer than nonusers (P <0.001). They were also significantly more likely to have used HRT before diagnosis (P <0.001), had smaller tumours (P <0.001) and fewer positive axillar y lymph nodes (P =0.006) and were followed up for longer (P <0.001). There were no significant differences between the two groups in ages at menarche and menopause, parity and gravidity. Oestrogen receptor status of the tumour was known in only 282 patients and was not included in the analysis. Most women in both groups took tamoxifen (58% and 60%, respectively).
Results: 1122 women were followed up for 0–36 years (median, 6.08 years); 154 were lost to follow-up. 286 women used HRT for menopausal symptoms for up to 26 years (median, 1.75 years).Compared with non-users,
HRT users had reduced risk of cancer recurrence (adjusted relative risk [RR], 0.62; 95% CI,0.43–0.87), all-cause mortality (RR, 0.34; 95% CI, 0.19–0.59) and death from primary tumour (RR, 0.40; 95% CI, 0.22–0.72).
Continuous combined HRT was associated with a reduced risk of death from primary tumour (RR, 0.32; 95% CI, 0.12–0.88) and all-cause mortality (RR, 0.27; 95% CI, 0.10–0.73).
Median daily dose of oestrogen used by women taking continuous combined HRT was 0.625 mg conjugated equine oestrogen (range, 0.3–0.625 mg) or equivalent (1.25 mg oestrone sulfate, 2 mg oestradiol valerate or 50 g transdermal oestradiol). Median daily dose of progestin in combined HRT was 50 mg medroxyprogesterone acetate
(range, 10–500 mg) or 5 mg norethisterone (range, 1–5 mg). Similar doses of oestrogen and progestin, respectively, were used by women who took oestrogen or progestin alone. Vaginal oestrogens included estriol cream (0.5 g) and estradiol vaginal tablets (25 g) used twice weekly, either alone or combined with oral progestins in similar doses to those in combined HRT.
the group that used HRT after diagnosis had a significantly lower risk of recurrence or new breast cancer compared with the no-HRT group (adjusted relative risk [RR], 0.62;95% CI, 0.43–0.87).
The HRT group also had significantly lower risks of death from all causes (adjusted RR, 0.34; 95% CI, 0.19–0.59) and from the primary tumour (adjusted RR, 0.40; 95% CI, 0.22–0.72).
Women who used HRT also had significantly lower risks of death from all
causes (RR, 0.34) and from the primary tumour (RR, 0.40).
3: Rates of recurrence and death and adjusted relative risks in women who used hormone replacement therapy (HRT) after diagnosis versus non-users
Recurrence or new breast cancer Death from all causes Death from primary tumour
Type of HRT Number of events Rate per 1000 person-years Adjusted relative risk (95% CI) Number of events
Rate per 1000 person-years Adjusted relative risk (95% CI) Number of events Rate per 1000 person-years
Adjusted relative risk (95% CI)
No HRT (n=836) 247 58.2 1 (referent) 199 29.4 1 (referent) 122 18.0 1 (referent)
HRT (all types) (n=286) 44 24.0 0.62 (0.43–0.87) 16 8.3 0.34 (0.19–0.59) 13 6.7 0.40 (0.22–0.72)
Combined HRT* (n=138) 23 23.6 0.81 (0.52–1.27) 5 4.9 0.27 (0.10–0.73) 4 3.9 0.32 (0.12–0.88)
Progestin alone (n=78) 12 32.5 0.59 (0.32–1.09) 4 9.6 0.34 (0.12–0.93) 4 9.6 0.33 (0.12–0.91)
Vaginal oestrogen (n=32) 4 14.9 0.18 (0.04–0.75) 4 14.3 0.30 (0.07–1.30) 2 7.1 0.35 (0.07–1.68)
Vaginal oestrogen plus progestin (n=21)
1 8.6 0.00 (0.00–) 0 0.0 0.00 (0.00–) 0 0.0 0.00 (0.00–)
Oestrogen alone (n=17) 4 39.2 0.81 (0.30–2.21) 3 28.8 0.85 (0.26–2.75) 3 28.8 1.11 (0.35–3.60)
*Combined=continuous combined oestrogen plus progestin.
All HRT regimens were associated with smaller risks of recurrence and death than no HRT use, with the exception of unopposed oestrogen for risk of death from primary tumour. ?????????
A possible explanation of the results is that women with oestrogen deficiency tend to have better outcomes after breast cancer. (LTED effect)
A 1997 meta-analysis of 51 studies from 21 countries reported a per annum increased risk of breast cancer of 1.7% with use of oestrogen alone, 1.8% with oestrogen plus continuous progestins, and 7.6% with oestrogen plus cyclic progestins.17
17. see imediately below this
Other recent observational studies also suggest that the use of sequential or cyclic progestins in HRT may increase the risk of breast cancer.18-20
==============================================================
2019 Version of study
21) Collaborative Group on Hormonal Factors in Breast Cancer. “Type and timing of menopausal hormone therapy and breast cancer risk: individual participant meta-analysis of the worldwide epidemiological evidence.” The Lancet 394.10204 (2019): 1159-1168.
Every MHT type, except vaginal oestrogens, was associated with excess breast cancer risks, which increased steadily with duration of use and were greater for oestrogen-progestagen than oestrogen-only preparations.
If these associations are largely causal, then for women of average weight in developed countries, 5 years of MHT, starting at age 50 years, would increase breast cancer incidence at ages 50–69 years by about
one in every 50 users of oestrogen plus daily progestagen preparations;
one in every 70 users of oestrogen plus intermittent progestagen preparations; and
one in every 200 users of oestrogen-only preparations.
The corresponding excesses from 10 years of MHT would be about twice as great.
Older Version of study:
(See above 17.)
Collaborative Group on Hormonal Factors in Breast Cancer. “Breast cancer and hormone replacement therapy: collaborative reanalysis of data from 51 epidemiological studies of 52 705 women with breast cancer and 108 411 women without breast cancer.” The Lancet 350.9084 (1997): 1047-1059.
In North America and Europe the cumulative incidence of breast cancer between the ages of 50 and 70 in never-users of HRT is about 45 per 1000 women. The cumulative excess numbers of breast cancers diagnosed between these ages per 1000 women who began use of HRT at age 50 and used it for 5, 10, and 15 years, respectively, are estimated to be 2 (95% CI 1–3), 6 (3–9), and 12 (5–20). Whether HRT affects mortality from breast cancer is not known.
==================================
abstract only
185 BC patients, 64 with and 121 without HRT
?LTED probably
? premarin ? Germany, probably not.
Progestin? probably
5 year follow up
Dramatic benefits for bone density in HRT group.
22) Beckmann, Matthias W., et al. “Hormone replacement therapy after treatment of breast cancer: effects on postmenopausal symptoms, bone mineral density and recurrence rates.” Oncology 60.3 (2001): 199-206.
Department of Obstetrics and Gynecology, Friedrich Alexander University, Erlangen, Germany.
In a retrospective study (total n = 185 BC patients, 64 with and 121 without HRT), the effect of HRT during or after adjuvant therapy [chemotherapy and/ or (anti-) hormonotherapy] has been investigated. The surveillance period was up to 60 months. Evaluated were HRT effects on (1) PMS measured by a comprehensive life quality questionnaire, (2) bone mineral density (BMD) measured by osteodensitometry and (3) morbidity as well as mortality rates. Results: Both groups did not differ with regard to tumor stage, lymph node involvement, metastasis, grading, and steroid hormone receptor status. A reduction in PMS was significant in women taking HRT (p < 0.001), especially in the subgroup of women ≤50 years (p < 0.0001).
For both age groups, the median reduction in BMD (z-score) was less in women receiving HRT (≤50 years: without HRT –1.99 vs. with HRT –0.95, p < 0.05; >50 years: without HRT –2.29 vs. with HRT –1.19, p < 0.01).
There were no statistically significant differences regarding morbidity and mortality (p = 0.29). Conclusion: In this study of BC patients, the use of HRT shows positive effects on PMS and BMD. There was no significant influence on morbidity or mortality. However, a reevaluation of HRT in the routine management of BC patients should await the results of prospective randomized trials
==================================================
FULL PDF !!!!
LTED 4.2 years
88 pts
54 pts estradiol and 10 days of MPA
34 pts hyx oral E2 only ESTRADIOL (Helsinki Finland)
43 pts controls
With HRT Without HRT
Nulliparous 14 (16%) 10 (23%) NS
Parous 74 (84%) 33 (77%) NS
Recurrence Rate
7/216 woman-years (3% per year). HRT group.
5/112 woman-years, (4% per year). Non-HRT Group
23) Marttunen, Merja B., et al. “A prospective study on women with a history of breast cancer and with or without estrogen replacement therapy.” Maturitas 39.3 (2001): 217-225.
Department of Obstetrics and Gynaecology, Helsinki University Central Hospital, PO Box 140, FIN-00029 HUS, Helsinki, Finland.
We recruited 131 patients who had been treated for breast cancer for a mean of 4.2 years (range 1 month to 20 years) before. Eighty-eight decided to use ERT, whereas 43 refused or had no need for ERT. At recruitment, the patients were carefully examined for breast and gynaecologic findings. Non-hysterectomized patients wishing to receive ERT (n=54) then started using estradiol as oral tablets (2 mg/day) (n=44) or as transdermal gel (1.5 mg/day) (n=10) in combination with 10-day courses of oral medroxyprogesterone acetate at 4-week intervals, whereas hysterectomized patients (n=34) used only estradiol, orally (2 mg/day) (n=31) or transdermally (1.5 mg/day) (n=3).
The patients using ERT were carefully examined 6 and 12 months later, and then annually at a specific outpatient department, and the mean follow-up time is now 2.5 years (range from 1 month to 5.2 years, 216 woman-years). The 43 patients not wishing to receive ERT were followed annually at the oncologic department for a mean of 2.6 years (range from 1 month to 4.7 years), and served as a control group.
Results: ERT significantly reduced climacteric symptoms, and the Kupperman score fell by 63%, from 26.9+/-8.6 to 9.9+/-6.7 (mean+/-SD). In non-hysterectomized women, medroxyprogesterone acetate triggered withdrawal bleeding in all except seven women. Seven patients (13%) experienced spotting during ERT. In 27 women, endometrial thickness exceeded 10 mm, and two of the total of 54 patients (3.7%) had simple hyperplasia. This vanished spontaneously in 3-6 months. Ten patients terminated the use of ERT within the first 12 to 39 months due to the lack of severe vasomotor symptoms (n=4) or due to the recurrence of breast cancer or to cancer of the contralateral breast (n=6).
Eighty-one of the 88 patients (92%) using ERT showed no evidence of recurrence, whereas five patients (5.7%) had recurrence in 12-36 months and two patients (2.3%) developed a cancer of the contralateral breast in 14-24 months; another one of those wanted to continue with ERT. Thus the combined risk of recurrence or a new cancer of the contralateral breast in ERT users was 7/216 woman-years (3% per year).
In the control group, 38 of 43 patients (88.4%) showed no evidence of recurrence or contralateral cancer, whereas four patients had recurrence and one developed a contralateral breast cancer (5/112 woman-years, 4% per year).
Large prospective, preferably placebo-controlled trials would be necessary to assess the final safety of ERT in healthy women, not to mention patients with a history of breast cancer. In practice such a study would be difficult to perform, and perhaps even unethical, because one cannot offer a placebo for a symptomatic climacteric patient wishing to receive ERT. Moreover, due to the good present prognosis of breast cancer patients [1–3], thousands of such patients would be needed if the end point of the trial were to be a recurrence of the cancer.
The role of progestin on the development of breast cancer is still unclear. The data from Sweden and United States have shown that adding progestin to estrogen replacement increases the risk of breast cancer by 14–40% in healthy women [27–29], whereas a progestin addition does not affect the risk of cancer recurrence in women with a history of breast cancer who are using estrogen replacement [21,23,25].
Why should this be the case ?
27.Persson I, Weiderpass E, Bergstro¨m R, Schairer C. Risks
of breast and endometrial cancer after estrogen and estrogen-
progestin replacement therapy. Cancer Causes Control
1999;10:253–60.
[28] Schairer C, Lubin J, Troisi R, Sturgeon S, Brinton L,
Hoover R. Menopausal estrogen and estrogen-progestin
replacement therapy and beast cancer risk. JAMA
2000;283:485–91.
[29] Ross RK, Paganini-Hill A, Wan PC, Pike MC. Effect of
hormone replacement therapy on breast cancer risk: estrogen
versus estrogen-progestin. J Natl Cancer Inst
2000;92:328–32.
[21] Eden JA, Bush T, Nand S, Wren BG. A case-control
study of combined continuous oestrogen-progestin replacement
therapy among women with a personal history
of breast cancer. Menopause 1995;2:67–72.
[23] Natrajan PK, Soumakis K, Gambrell RD. Estrogen replacement
therapy in women with previous breast cancer.
Am J Obstet Gynecol 1999;181:288–95.
[25] Dew J, Eden J, Beller E, Magarey C, Schwartz P, Crea P,
Wren B. A cohort study of hormone replacement therapy
given to women previously treated for breast cancer.
Climactric 1998;1:137–42.
Conclusions: Symptomatic climacteric patients with a history of breast cancer benefited from ERT without increasing their risk of recurrence, but the short follow-up and the small number of patients limit any definitive recommendations.
=====================================================================
abstract only
41 pts
no info as to HRT type
24) DiSaia, PhilipJ, et al. “Hormone replacement therapy in breast cancer.” The Lancet 342.8881 (1993): 1232.
25) DiSaia PJ, Grosen EA, Kurasaki T, Gildea M, Cowen B, Anton-Culver H. Hormone replacement therapy in breast cancer survivors:a cohort study. Am J Obstet Gynecol 1996;174:1494-8.
Objective: Our purpose was to measure any adverse effect (if one exists) of hormone replacement therapy administered to breast cancer survivors.
Study design: Forty-one patients from a group of 77 patients who received hormone replacement therapy after therapy for breast cancer were matched with 82 comparison patients not receiving hormone replacement therapy. Both groups were taken from the same population on the basis of cancer registry of the Cancer Surveillance Program of Orange County and were compared with regard to survival results.
Results: An analysis of survival time and disease-free time revealed no statistically significant difference between the two groups.
Conclusions: No obvious adverse effect of hormone replacement therapy could be shown in this pilot study. A case is made for a prospective randomized trial.
LTED 46 months
The risk of death was lower among the HRT survivors; odds ratio 0.28
26) Disaia PJ, Brewster WR, Ziogas A, et al. Breast cancer survival and hormone
replacement therapy. A cohort analysis. Am J Clin Oncol. 2000;23:541–545.
Controversy exists regarding the safety of hormone replacement therapy (HRT) after a diagnosis of breast cancer. The objective of this study is to perform a matched cohort analysis to evaluate the impact of HRT on mortality in breast cancer survivors. Patients with breast cancer who received HRT after diagnosis of breast cancer were identified. Control subjects were identified from the regional cancer registry. Matching criteria included age at diagnosis, stage of breast cancer, and year of diagnosis. Controls were selected only if they were alive at the time of initiation of HRT of the matched case. Only subjects not included in a previously reported matched analysis were selected.
One hundred twenty-five cases were matched with 362 controls.
Ninety-eight percent (123/125) of the cases received systemic estrogen; 90/125 (72%) also received a progestational agent.
The median interval between diagnosis of breast cancer and initiation of HRT was 46 months (range 0-401 months). The median duration of HRT was 22 months (range 1-357 months).
The risk of death was lower among the HRT survivors; odds ratio 0.28 (95% confidence interval 0.11-0.71). This analysis does not suggest that HRT after the treatment of breast cancer is associated with an adverse outcome.
========================================================
NICE STUDY Showing value of LTED > 2 years (no recurrence), less than 2 years LTED recurrence in 3/25.
Abstract only
27) Wile AG, Opfell RW, Margileth DA. Hormone replacement therapy in previously treated breast cancer patients. Am J Surg 1993;165:372-5.
Orange California
25 pts
LTED group I <24 months (7.9 months average)
LTED group II >24 months (64.5 mo. average)
Three of 25 patients have had a recurrence, all in group I.
We report our experience with 25 women previously treated for breast cancer who subsequently received hormone replacement therapy (HRT) for the relief of menopausal symptoms and the prevention of postmenopausal cardiovascular disease and osteoporosis. Two patients had in situ disease, 13 had stage I disease, 7 had stage II disease, 1 had stage III disease, and 2 had invasive cancer of undetermined stage.
Seventeen patients (group I) began HRT less than 24 months after primary breast cancer therapy, and 8 patients (group II) began HRT more than 24 months after breast cancer therapy.
The HRT-free interval for group I patients averaged 7.9 months and for group II patients averaged 64.5 months.
The average period of observation while receiving HRT for the entire group was 35.2 months (range: 24 to 82 months).
Three of 25 patients have had a recurrence, all in group I.
One patient developed local recurrence after breast conservation treatment, and her condition was salvaged by further wide excision.
Two patients developed recurrence after mastectomy, and one patient ultimately died of systemic disease. The overall survival rate for the entire group was 96%.
Overall survival of high-risk group I patients, with a mean follow-up of 30.4 months, was 94%. We recognize that this report of HRT in a small group of patients does not have the power to demonstrate an adverse effect of HRT on breast cancer. However, the lack of an obvious adverse effect of HRT in this group of breast cancer patients and the known beneficial effect of HRT on postmenopausal cardiovascular disease and osteoporosis warrant formal prospective trials of HRT in such patients.
================================================================
Full pdf
28) Guidozzi F. Estrogen replacement therapy in breast cancer survivors. Int J
Gynecol Obstet. 1999;64:59–63.
Department of Obstetrics and Gynaecology, Johannesburg Hospital, University of the Witwatersrand Medical School, South Africa.
20 premarin and medroxyprogesterone and four tibolone.
no recurrences to date after 24-44 months
Results: Twenty-four patients who had previously been treated for breast cancer 8-91 months prior to their initiating ERT have been observed for 24-44 months. There were 15 patients with stage 1, eight with stage 2 and one with stage 4 breast cancer. The mean age of the patients at commencement of ERT was 48 years (range 42-61). Two patients had a biopsy of a suspicious breast nodule: both of which were benign. There have not been any recurrences to date.
Conclusion: Breast cancer survivors did not have their outcome adversely affected by ERT during an observation period of 24-44 months.
========================================================
full pdf
Randominzed Controlled Trial
LTED 37 months
Duration of study ONLY 6 months
estradiol valerate 2 mg/day plus levonorgestrel if intact uterus
100 patients randomized to HRT vs no HRT
some used TAM.
Full text
29) Marsden J, Whitehead M, A’Hern R, et al. Are randomized trials of hormone replacement therapy in symptomatic women with breast cancer feasible? Fertil Steril. 2000;73:292–299.
Postmenopausal women (i.e., >1 year since their last menstrual period), with a previous diagnosis of in situ disease or stage I/II breast carcinoma were identified from preliminary inspection of medical records before their attendance at breast cancer follow-up clinics at The Royal Marsden and St. George’s Hospital NHS Trusts, London.
Women were randomized (1:1) to HRT or no HRT for the 6-month duration of the study (Fig. 1).
The HRT used in this study was either estradiol valerate 2 mg/d (Progynova; Schering Health Care Ltd., Burgess Hill, West Sussex, UK) in hysterectomized women or the same estrogen plus levonorgestrel 75 μg/d for 12 of 28 days (Nuvelle; Schering Health Care Ltd.) in those with an intact uterus.
A total of 100 women were randomized so that continuance with HRT and no HRT could be estimated in 50 patients
Half were on tamoxifen
TAM − ve (n = 51) TAM + ve (n = 49)
Of the 100 women recruited, 51 were not currently taking tamoxifen (TAM − ve), whereas 49 did take the drug (TAM + ve).
To date, three women have developed recurrent breast cancer, two of which were taking HRT. One was randomized to receive opposed HRT 38 months after diagnosis and developed recurrence after taking HRT for 2 years. The second patient received unopposed HRT 9 years after diagnosis and developed recurrent disease after only 6 weeks of treatment. The third patient, who never took HRT, developed recurrent disease 6 months after her initial diagnosis.
===================================================================
HABITS
Estradiol plus Progestin, norethisterone
LTED ??? However, for 80% of the women in both arms, the time between first diagnosis and random assignment was between 7 months and approximately 8 years.
HABITS was a randomized, non-placebo-controlled noninferiority trial that aimed to be at a power of 80% to detect a 36% increase in the hazard ratio (HR) for a new breast cancer event following HT.
continuous combined or sequential estradiol hemihydrate and norethisterone.
39 of the 221 women in the HT arm and
17 of the 221 women in the control arm
experienced a new breast cancer event (HR = 2.4, 95% CI = 1.3 to 4.2).
30) Holmberg L, Anderson H. HABITS (hormonal replacement therapy after
breast cancer—is it safe?), a randomised comparison: trial stopped. Lancet. 2004;363:453–455. Full pdf Holmberg HABITS Lancet 2004
Regional Oncologic Centre, University Hospital, SE-751 85 Uppsala, Sweden.
Abstract
In the 1990s, two randomised clinical trials started in Scandinavia addressing whether hormone replacement therapy (HRT) is safe for women with previous breast cancer. We report the findings of the safety analysis in HABITS (hormonal replacement therapy after breast cancer–is it safe?), an open randomised clinical trial with allocation to either HRT or best treatment without hormones. The main endpoint was any new breast cancer event. All analyses were done according to intention-to-treat.
Until September, 2003, 434 women were randomised; 345 had at least one follow-up report.
After a median follow-up of 2.1 years, 26 women in the HRT group and seven in the non-HRT group had a new breast-cancer event. All women with an event in the HRT group and two of those in the non-HRT group were exposed to HRT and most women had their event when on treatment. We decided that these findings indicated an unacceptable risk for women exposed to HRT in the HABITS trial, and the trial was terminated on Dec 17, 2003.
——————————————– —————————————–
HABITS 2008 Holmberg full pdf
continuous combined or sequential estradiol hemihydrate and norethisterone
HR = 2.4
continuous combined or sequential estradiol hemihydrate and norethisterone.testosterone-like progestagen, NETA, either continuously or for at least 10 days of a 28-day cycle
There are also indications that NETA is associated with a higher risk of
breast cancer than MPA, especially if continuous combined regimens are used ( 21 , 22 ).
Nice review of different estrogens and progestins and cancer risk
full text
21. Stahlberg C , Tonnes PedersenA , LyngeE , OttesenB . Hormone replacement therapy and risk of breast cancer: the role of progestins . Acta Obstet Gynecol Scand . 2003 ; 82 ( 7 ) : 335 – 344
In the USA the predominant compounds are conjugated estrogens and medroxyprogesterone-acetate, whereas oestradiol combined with testosterone-like progestins is commonly used in Europe. The progestin preferred in the USA is medroxyprogesterone-acetate (MPA), which resembles natural progesterone and is administered either cyclically or continuously. In Europe the predominant regimen prescribed is 17-b-oestradiaol opposed by testosterone-derived progestins, mainly norethistosteroneacetate (NETA) or levonorgestrel (LNG); while the less androgenic progestin MPA is used to a lesser extent.
Magnusson et al. found a 5-fold increased breast cancer risk with the continuous treatment
with NETA/LNG after more than 10 years of use as compared with never-users. This was not found for the continuous treatment with MPA in the study by Ross et al.
full text
22. Magnusson C , Baron JA , CorreiaN , Bergström R , Adami HO , Persson I .Breast cancer risk following long-term oestrogen- and oestrogenprogestin-replacement therapy . Int J Cancer . 1999 ; 81 ( 3 ) : 339 – 344 .1Department of Medical Epidemiology, Karolinska Institutet, Stockholm, Sweden
Exclusive use of oestrogen with progesterone-derived progestins did not appear to confer an excess breast-cancer risk (OR per year of use 0.95, 95% CI 0.80–1.14) (Table IV), an analysis based, however, on few exposed women (32 cases and 34 controls). In contrast, use of oestrogen with testosterone-derived progestins increased the risk of breast cancer by 8% per year of use (OR 1.08,95% CI 1.07–1.13), and use for more than 10 years conferred an OR of 3.41 (95% CI 1.91–6.08), as compared with never-use
While use of hormone-replacement therapy (HRT) effectively alleviates menopausal symptoms and prevents osteoporosis and possibly cardiovascular disease, there is concern of a detrimental impact on breast-cancer risk. There is a particular lack of data regarding the effect of long-term use of oestrogen-progestin combinations on breast-cancer risk. We conducted a large epidemiological study in Sweden, where combined oestrogen-progestin treatment has been predominant, to examine the influence of different regimens of menopausal hormone therapy on breast-cancer risk. In this population-based case-control study, 3,345 women aged 50 to 74 years with invasive breast cancer (84% of all eligible) and 3,454 controls of similar age (82% of all selected) were included. Mailed questionnaires and telephone interviews were used to collect detailed information on use of hormone replacement and on potential confounding factors. Odds ratios (OR) and 95% confidence intervals (CI) were estimated through multiple logistic regression. There was a trend of increasing breast-cancer risk with duration of oestrogen/oestrogen-progestin use (OR for women treated at least 10 years, 2.43; 95% CI, 1.79-3.30, as compared to never-users), with statistically significant estimates only for women with BMI<27 kg/m2.
Excess risks were observed to current use and use that ceased more than 10 years ago (OR for women treated at least 5 years, OR was 2.68, 95% CI, 2.09-3.42, and OR 2.57, 95% CI, 1.28-5.15, as compared with never-users, respectively). A positive association which was noted for use of oestrogen combined with testosterone-derived progestins appeared especially pronounced with continuously combined regimens. Long-term use of replacement oestrogens with or without progestins may substantially increase the incidence of post-menopausal breast cancer, particularly among non-obese women.
LTED ??? Time in years between primary treatment and randomization, median (range) 2.1 years (0.1 – 23.2) 2.2 (0.1 – 26.5)
33.5% were taking tamoxifen at the time of random assignment
A total of 442 women could be followed for a median of 4 years.
Women in the HT arm experienced new breast cancer events more than twice as often as women in the control arm (39 out of 221 vs 17 out of 221; hazard ratio = 2.4, 95% confidence interval = 1.3 to 4.2).
The cumulative incidence of a new breast cancer event at 5 years was estimated at 22.2% for the HT arm and at 8.0% for the control arm.
Continuation of Holmberg………………………..2008
Holmberg L, Iversen OE, Rudenstam CM, et al. Increased risk of recurrence after hormone replacement therapy in breast cancer survivors.J Natl Cancer Inst. 2008;100:475–482.
Regional Oncologic Centre, University Hospital, SE-751 85 Uppsala, Sweden.
Background: Hormone replacement therapy (HT) is known to increase the risk of breast cancer in healthy women, but its effect on breast cancer risk in breast cancer survivors is less clear. The randomized HABITS study, which compared HT for menopausal symptoms with best management without hormones among women with previously treated breast cancer, was stopped early due to suspicions of an increased risk of new breast cancer events following HT. We present results after extended follow-up.
Methods: HABITS was a randomized, non-placebo-controlled noninferiority trial that aimed to be at a power of 80% to detect a 36% increase in the hazard ratio (HR) for a new breast cancer event following HT. Cox models were used to estimate relative risks of a breast cancer event, the maximum likelihood method was used to calculate 95% confidence intervals (CIs), and chi(2) tests were used to assess statistical significance, with all P values based on two-sided tests. The absolute risk of a new breast cancer event was estimated with the cumulative incidence function. Most patients who received HT were prescribed continuous combined or sequential estradiol hemihydrate and norethisterone.
Results: Of the 447 women randomly assigned, 442 could be followed for a median of 4 years. Thirty-nine of the 221 women in the HT arm and 17 of the 221 women in the control arm experienced a new breast cancer event (HR = 2.4, 95% CI = 1.3 to 4.2). Cumulative incidences at 5 years were 22.2% in the HT arm and 8.0% in the control arm.
By the end of follow-up, six women in the HT arm had died of breast cancer and six were alive with distant metastases. In the control arm, five women had died of breast cancer and four had metastatic breast cancer (P = .51, log-rank test).
Conclusion: After extended follow-up, there was a clinically and statistically significant increased risk of a new breast cancer event in survivors who took HT.
When preliminary results based on a median follow-up of 2 years were reported ( 4 ), 33 breast cancer events had occurred and the hazard ratio had reached 3.5 (95% CI = 1.5 to 7.4). Here, we report results from a median follow-up of 4 years and a total of 56 breast cancer events.
We recommended that the patients receive mammograms every 12 – 24 months or participate in routine mammographic screening with target intervals of 18 – 24 months.
HT was given for 2 years in the HT arm. The majority of centers prescribed a sequential or continuously com-bined regimen of estradiol hemihydrate (E2) and norethisterone acetate (NETA) for women with an intact uterus or estradiol for women who had undergone hysterectomy.
For the 54 women exposed to estrogen only (47 from the HT arm and seven from the control arm), 20 women took 1 – 2 mg E2 (Estrofem) daily and 10 took 0.625 or 1.25 mg conjugated estrogens (Compremin) daily.
Exposure to estrogen alone yielded a HR of 1.4 (95% CI = 0.55 to 3.3) as compared with exposure to a continuously combined regimen. (1.4 x 1.6=2.24)
All women in the HT arm who developed a breast cancer event had been exposed to HT. Five of the women with a breast cancer event in the non-HT arm had also taken HT after random assign-ment. Nineteen women in the HT arm had a new breast cancer event after the 2-year follow-up period (when the treatment should have stopped according to protocol). Nine of these women had con-tinued to use HT until the time of the event, and one had exposure until 4 months before the event.
By the end of follow-up, six women from the HT group had died of breast cancer and six were alive with distant metastases. For the non-HT group, five women had died of breast cancer and four were alive with distant metastases. The difference in distant metastasis –free survival was not statistically significant ( P = .51, log-rank test). Three women in the HT arm died of causes other than breast cancer, compared with none in the non-HT arm.
Because it was open label and not placebo-controlled, there was a risk for a bias related to possibly more vigorous follow-up and diagnosis of events in the HT arm.
All women with breast cancer events assigned to the HT arm were exposed to HT, and fi ve of the 17 women in the non-HT arm with a new breast cancer event were also exposed to HT.
It is not surprising that the results from this randomized trial deviate from those in the observational series ( 2 ). The majority of the observational studies were not formal studies that could con-trol suffi ciently for bias and confounding. Even in the carefully designed observational study from Seattle ( 12 ) that studied the risk of breast cancer recurrence following HT among breast cancer survivors and found decreased risk for HT users compared with nonusers (HR = 0.50, 95% CI = 0.30 to 0.85), it was diffi cult to completely exclude selection bias. An important methodological problem in observational studies is to adjust for the effects of any pretreatment screening for metastases and restaging of those exposed to HT. Furthermore, reliable data on patterns of HT exposure are seldom available in the observational setting. In several of the case series ( 13 – 18 ), one or more of the investigators were also the patients’ physicians, which increases the risk that patients do not report back bad outcomes
First, there was a higher proportion of women with node-positive breast cancer in the HABITS trial than in the Stockholm trial.
Second, a higher proportion of the women in the Stockholm study were being treated with tamoxifen, which would theoretically confer protection from breast cancer recurrence.
A third possible reason for the different results between the HABITS trial and the Stockholm study is that in the HABITS trial the most commonly used HT regimens included a potent testosterone-like progestagen, NETA, either continuously or for at least 10 days of a 28-day cycle. By contrast, the Stockholm study was specifi cally designed to keep doses of progestagen as low as possible and to use a naturally occurring progesterone, [error-this is not anaturally ocurring progesterone] medroxyprogesterone acetate (MPA), instead of NETA.
The addition of progestagens in HT has been found to increase the risk of breast cancer ( 6 , 7 , 19 – 21 ). There are also indications that NETA is associated with a higher risk of breast cancer than MPA, especially if continuous combined regimens are used (21, 22).
21. Stahlberg C , Tonnes Pedersen A , Lynge E , Ottesen B . Hormone replace-ment therapy and risk of breast cancer: the role of progestins . Acta Obstet Gynecol Scand . 2003 ; 82 ( 7 ) : 335 – 344 .
22. Magnusson C , Baron JA , Correia N , Bergström R , Adami HO , Persson I . Breast cancer risk following long-term oestrogen- and oestrogen-progestin-replacement therapy . Int J Cancer . 1999 ; 81 ( 3 ) : 339 – 344 .
In the HABITS trial, the new breast cancer events in the HT arm were mainly local events, and there was no convincing evi-dence for a higher breast cancer mortality associated with HT exposure.
-============================================================
Stockholm trial full pdf
32) von Schoultz Eva, Rutqvist LE, Stockholm Breast Cancer Study Group. Menopausal hormone therapy after breast cancer: the Stockholm randomized trial. J Natl Cancer Inst. 2005;97:533–535.
Among the 188 patients who had been randomly assigned to receive menopausal
hormone therapy, 42 (22%) started cyclic estradiol and medroxyprogesterone
acetate, 94 (50%) started spacing out estradiol and medroxyprogesterone acetate,
and 43 (23%) started estradiol valerate alone.
At a median follow-up of 4.1 years (end date for follow-up = January 2004)
in the Stockholm trial, there was a total of 24 breast cancer recurrences: 11 in the
menopausal hormone therapy group and 13 in the control no-treatment group
( Table 2 ). The total number of deaths in the menopausal hormone therapy group
was four (two from breast cancer) and in the control group nine (four from breast
cancer).
In 1997 two independent randomized clinical trials, Hormonal Replacement Therapy After Breast Cancer–Is It Safe? (HABITS; 434 patients) and the Stockholm trial (378 patients), were initiated in Sweden to compare menopausal hormone therapy with no menopausal hormone therapy after diagnosis of early-stage breast cancer. Much of the design of both studies was similar; however, a goal of the Stockholm protocol, not shared with the HABITS trial, was to minimize the use of progestogen combined with estrogen. The HABITS trial was prematurely stopped in December 2003, because, at a median follow-up of 2.1 years, the risk for recurrence of breast cancer among patients receiving menopausal hormone therapy was statistically significantly higher (relative hazard [RH] = 3.3, 95% confidence interval [CI] = 1.5 to 7.4) than among those receiving no treatment. In the Stockholm trial, however, at a median follow-up of 4.1 years, the risk of breast cancer recurrence was not associated with menopausal hormone therapy (RH = 0.82, 95% CI = 0.35 to 1.9). Statistically significant heterogeneity in the rate of recurrence was observed (P = .02; two-sided likelihood-ratio test) between the two studies, indicating that chance may not be the only explanation. Doses of estrogen and progestogen and treatment regimens for menopausal hormone therapy may be associated with the recurrence of breast cancer.
Patients who had had a hysterectomy in the menopausal hormone therapy group
were given continuous treatment with 2 mg of estradiol valerate daily.
Patients in the group receiving no menopausal hormone therapy were asked
to refrain from use of all types of menopausal hormone therapy. Local vaginal
treatment with low-dose estrogen gels or vagitories was allowed.
For example, the proportion of lymph node – positive patients was higher in the
HABITS trial than in the Stockholm trial (26% versus 16%, respectively), and
fewer patients received concomitant adjuvant tamoxifen therapy in the HABITS
trial than in the Stockholm trial (21% versus 52%, respectively). In addition, the
Stockholm protocol attempted to minimize the use of progestogen in combination
with estrogen. In contrast to the HABITS trial, the Stockholm trial recommended
that patients avoid continuous combined treatment with estrogen and progestogen
and use regimens that incorporated 1 week of no treatment every 1 (cyclic regimen)
or 3 (spacing out regimen) months
These treatment recommendations were based on the following results, available when the trial was initiated, that indicated differential effects on the breast when treatment with estrogen alone was compared with combined treatment with estrogen and progestogen
( 2 – 4 ) . First, breast cell proliferation is increased only during the luteal phase of
the menstrual cycle when levels of both estrogen and progesterone are high ( 2 ) .
Second, using a relevant prospective monkey model for menopausal hormone
therapy, we reported statistically significantly higher proliferation in the breast
during continuous combined estrogen and progestogen treatment than during
estrogen-only treatment ( 3 ) . Finally, cyclic discontinuation of hormonal treatment
was hypothesized to decrease the expression of local growth factors in
breast tissue and to initiate and stimulate apoptosis ( 4 ) .
In the Stockholm trial, 73% of the women were first assigned to menopausal
hormone therapy containing either estrogen alone or the spacing out
regimen, in which progestogen was given for only 14 days at 3-month intervals.
This protocol could provide one explanation for the lack of difference in
breast cancer recurrence between the menopausal hormone therapy group and
the no treatment group in the Stockholm trial.
===============================
full text
Stockholm randomized trial 10 year follow up
33) Fahlén M, Fornander T, Johansson H, et al. Hormone replacement therapy after breast cancer: 10 year follow-up of the Stockholm randomized trial. Eur J Cancer. 2013;49:52–59.
Background: The management of hormonal deficiency symptoms in breast cancer survivors is an unsolved problem. While hormone replacement therapy (HRT) may increase the risk of breast cancer in healthy women, its effects on recurrence is unclear. Observational studies have suggested decreased recurrence rates from HRT. The few clinical trials in this field have all been closed preterm.
Methods: The Stockholm trial was started in 1997 and designed to minimise the dose of progestogen in the HRT arm. Disease-free women with a history of breast cancer were randomised to HRT (n=188) or no HRT (n=190). The trial was stopped in 2003 when another Swedish study (HABITS, the Hormonal Replacement After Breast Cancer – Is it Safe?) reported increased recurrence. However the Stockholm material showed no excess risk after 4 years of follow-up. A long term follow-up has now been performed.
Findings: After 10.8 years of follow-up, there was no difference in new breast cancer events: 60 in the HRT group versus 48 among controls (hazard ratio (HR)=1.3; 95% confidence interval (CI)=0.9-1.9). Among women on HRT, 11 had local recurrence and 12 distant metastases versus 15 and 12 for the controls. There were 14 contra-lateral breast cancers in the HRT group and four in the control group (HR=3.6; 95% CI=1.2-10.9; p=0.013). No differences in mortality or new primary malignancies were found.
Interpretation: The number of new events did not differ significantly between groups, in contrast to previous reports. The increased recurrence in HABITS has been attributed to higher progestogen exposure. As both trials were prematurely closed, data do not allow firm conclusions. Both studies found no increased mortality from breast cancer or other causes from HRT. Current guidelines typically consider HRT contraindicated in breast cancer survivors. Findings suggest that, in some women symptom relief may outweigh the potential risks of HRT.
=======================================================
No abstract available
34) Bluming AZ. Hormone replacement therapy (HRT) in women with previously treated primary breast cancer: update XIV. Proc ASCO. J Clin Oncol.2008;15 s:20693.
============================================================
OCP’s
35) Figueiredo JC, Bernstein L, Capanu M, et al. Oral contraceptives, postmenopausal hormones, and risk of asynchronous bilateral breast cancer: the WECARE study group. J Clin Oncol. 2008;26:1411–1418
Abstract/ Purpose: To investigate whether oral contraceptive (OC) use and postmenopausal hormones (PMH) are associated with an increased risk of developing asynchronous bilateral breast cancer among women diagnosed with breast cancer younger than 55 years.
Patients and methods: The WECARE (Women’s Environment, Cancer, and Radiation Epidemiology) study is a population-based, multicenter, case-control study of 708 women with asynchronous bilateral breast cancer and 1,395 women with unilateral breast cancer. Risk factor information collected during a telephone interview focused on exposures before and after the first breast cancer diagnosis. Treatment and tumor characteristics were abstracted from medical records. Multivariable conditional logistic regression was used to estimate rate ratios (RR) and 95% CIs.
Results: OC use before the first breast cancer diagnosis was not associated with risk of asynchronous bilateral breast cancer (RR = 0.88; 95% CI, 0.67 to 1.16). OC use after breast cancer diagnosis was also not significantly associated with risk (RR = 1.56; 95% CI, 0.71 to 3.45). Risk did not increase with longer duration of use or among women who had begun using OCs at a younger age. No evidence of an increased risk of asynchronous bilateral breast cancer was observed with PMH use before (RR = 1.21; 95% CI, 0.90 to 1.61) or after breast cancer diagnosis (RR = 1.10; 95% CI, 0.67 to 1.77). Neither duration nor type of PMH were associated with risk. Age at and time since first breast cancer diagnosis did not substantially affect these results.
Conclusion: This study provides no strong evidence that OC or PMH use increases the risk of a second cancer in the contralateral breast.
===================================================
LTED from Marttunen,2001
===================================================
Mean interval between surgery and start of ERT / follow-up months
Wile 1992 26
Powles 1993 31
DiSaia 1993 24
Eden 1995 60
V-Sellin 1995 84
Narrajan 1999 17
U-Vrscaj 1999 62
===================================
duplicate: See (1)
36) Bluming, Avrum Zvi. “Hormone replacement therapy after breast cancer: it is time.” The Cancer Journal 28.3 (2022): 183-190.
1a. Palshof et al. 1980 Prospective randomized 37/95 2 3 Reduced recurrence* 61
1b. Palshof et al. 1985 Updates of the original study 51/103 2 6.5 Reduced recurrence
Reduced mortality
62
2. Stoll and Parbhoo 1988 Prospective single-arm 14/ 0.25–0.5 2 No recurrence 63
3. Powles et al. 1993 Retrospective observational 35/ 1.2/0.1–3.7 3.6 2 of 35 developed recurrence No breast cancer deaths 64
4. Eden et al. 1995 Retrospective case-control 90/811 1.5/0.25–12 3 Reduced recurrence 65
5. Vassilopoulou-Sellin et al. (feasibility study) 1997 Prospective single-arm 43/ 2.6/2–12 12
1 of 43 developed recurrence No breast cancer deaths 66
6. Dew et al. 1998 Retrospective cohort 167/1305 1.6/0.25–22 4 No difference† 67
7. Espie et al. 1999 Retrospective cohort 120/240 2.4/1–10.6 2.4 No difference 68
8. Guidozzi 1999 Prospective single-arm 20/ 2.7/2–3.7 5.7 No recurrence 69
9. Natrajan et al. 1999 Retrospective cohort 50/26 5.5/0.5–32 7 No difference 70
10. Uršič-Vrščaj and Bebar 1999 Prospective cohort 21/42 2.3/0.25–6 2.3 No difference 71
11. Vassilopoulou-Sellin et al. 1999 Prospective cohort 39/280 4/2–6 3.8 No difference 72
12. Disaia et al. 2000 Retrospective cohort 125/362 1.8/0.1–30 Reduced mortality 73
13. Marsden et al. 2000 Prospective randomized 51/49 0.5 No difference 74
14. Peters et al. 2001 Prospective cohort 56/551 6.4/1–20.9 12.8 No difference 75
15. Marttunen et al. 2001 Prospective cohort 88/43 2.6 2.6 No difference 76
16. Beckmann et al. 2001 Retrospective cohort 64/121 3.5/3 5 No difference 77
17. O’Meara et al. 2001 Retrospective case-control 174/695 1.25 4.6 Reduced recurrence
Reduced mortality 78
18. Vassilopoulou-Sellin et al. 2002 Prospective randomized 56/243 5 6 No difference 79
19. Durna et al. 2002 Retrospective observational 286/836 1.75/0.17–34 6 Reduced recurrence Reduced mortality 80
20. Decker et al. 2003 Prospective cohort 277/554 3.7 No difference 81
21. Gorins et al. 2003 Prospective cohort 230/ 2.5 No difference 82
22a. Holmberg and Anderson (HABITS) 2004 Prospective randomized 174/171 2 2.1 Increased risk of local or contralateral tumors only No increased risk of metastases or death 83
22b. Holmberg et al. (HABITS) 2008 Updates of the original study 221/221 2 5 No increased mortality 84
23a. von Schoultz and Rutqvist (Stockholm) 2005 Prospective randomized 175/184 4.1/0.2–7 4.1 No difference 85
23b. Fahlén et al. (Stockholm) 2013 Updates of the original study 188/190 2.6 10.8 No difference 86
24. Bluming 2008 Prospective cohort 117/63 7.5/1–15 7.5 Reduced recurrence 87
25. Figueiredo et al. 2008 Retrospective case-control 708/1399 No difference 88
Boldface is employed to identify the prospective randomized trials and to identify significant positive or negative findings.
*Reduced recurrence = breast cancer survivors given HRT had fewer recurrences of breast cancer, or lower risk of death, than control group not on HRT.
†
===============================
No abstract available
37) Hayes, Theresa M., et al. “Hormone replacement therapy after a diagnosis of breast cancer: cancer recurrence and mortality.” The Medical Journal of Australia 178.8 (2003): 412-413.
===============================
Testosterone Pellets
Full pdf
38) Glaser, Rebecca L., Anne E. York, and Constantine Dimitrakakis. “Incidence of invasive breast cancer in women treated with testosterone implants: a prospective 10-year cohort study.” BMC cancer 19.1 (2019): 1271.
39) Glaser, R. L., A. E. York, and C. Dimitrakakis. “Abstract P6-13-02: Reduced incidence of breast cancer with testosterone implant therapy: A 10-year cohort study.” Cancer Research 79.4_Supplement (2019): P6-13.
Testosterone implants have been used for over eighty years to treat symptoms of hormone deficiency in pre and postmenopausal women. Evidence supports that androgens are breast protective. However, there is a lack of data on the long-term effect of testosterone therapy on the incidence of invasive breast cancer (IBC). This study was specifically designed to investigate the incidence of IBC in pre and postmenopausal women (presenting with symptoms of androgen deficiency) treated with subcutaneous testosterone implants or testosterone implants combined with anastrozole.
Methods
The 10-year prospective cohort study was approved in March 2008 at which time recruitment was initiated. Recruitment was closed March 2013. Pre and postmenopausal women receiving at least two pellet insertions were eligible for analysis (N = 1267). Breast cancer incidence rates were reported as an unadjusted, un-weighted value of newly diagnosed cases divided by the sum of ‘person-time of observation’ for the at-risk population. Incidence rates on testosterone therapy were compared to age-specific Surveillance Epidemiology and End Results (SEER) incidence rates and historical controls. Bootstrap sampling distributions were constructed to verify comparisons and tests of significance that existed between our results and SEER data.
Results
As of March 2018, a total of 11 (versus 18 expected) cases of IBC were diagnosed in patients within 240-days following their last testosterone insertion equating to an incidence rate of 165/100000 p-y, which is significantly less than the age-matched SEER expected incidence rate of 271/100000 p-y (p < 0.001) and historical controls.
Conclusion
Long term therapy with subcutaneous testosterone, or testosterone combined with anastrozole, did not increase the incidence of IBC. Testosterone should be further investigated for hormone therapy and breast cancer prevention.
============================
40) Donovitz, Gary, and Mandy Cotten. “Breast cancer incidence reduction in women treated with subcutaneous testosterone: testosterone therapy and breast cancer incidence study.” European journal of breast health 17.2 (2021): 150.
Testosterone (T) therapy has been shown to be breast protective in both pre- and post-menopausal patients. Additionally, estradiol (E) does not cause breast cancer (BC) in the majority of the world’s literatures. This study aimed to investigate the incidence of invasive BC (IBC) in pre- and postmenopausal women treated with T therapy and T in combination with E (T/E).
Materials and Methods:
Since January 2010, a total of 2,377 pre- and post-menopausal women were treated with T or T/E implants. IBC rates were reported based on newly diagnosed IBC cases in the total study. Total cases divided by the total sample size and years in study was expressed as an incidence per 100,000 person-years (P-Ys). The BC incidence was compared with age-specific Surveillance Epidemiology and End Results (SEER) incidence rates.
Results:
As of October 2020, 14 cases diagnosed with IBC have been found in 9,746 P-Y of follow up for an incidence of 144 cases per 100,000 P-Y, substantially less than the age-specific SEER incidence rates (223/100,000), placebo arm of Women’s Health Initiative Study (330/100,000), and never users of hormone therapy from the Million Women Study (312/100,000).
Conclusion:
T and/or T/E pellet implants significantly reduced the incidence of BC in pre- and post-menopausal women. The addition of E did not increase the incidence over using T alone. This is the second multi-year long-term study demonstrating the benefits of T therapy in reducing the incidence of IBC.
============================================
41) Cline, J. M., et al. “Effects of hormone replacement therapy on the mammary gland of surgically postmenopausal cynomolgus macaques.” American journal of obstetrics and gynecology 174.1 Pt 1 (1996): 93-100.
OBJECTIVE: Our purpose was to define the proliferative response and receptor status in the mammary glands of surgically postmenopausal macaques given hormone replacement therapy, equivalent for monkeys to that given women.
STUDY DESIGN: Surgically postmenopausal adult female cynomolgus macaques (Macaca fascicularis) were given either no treatment (n = 26), conjugated equine estrogens (n = 22), or combined therapy with conjugated equine estrogens and medroxyprogesterone acetate (n = 21). Drugs were administered in the diet, at doses equivalent on a caloric basis to 0.625 mg per woman per day for conjugated equine estrogens and 2.5 mg per woman per day for medroxyprogesterone acetate, for 30 months. Mammary gland proliferation was assessed
subjectively and by morphometric and stereologic means. Estrogen receptor and progesterone receptor content and proliferation were studied by immunohistochemistry.
RESULTS:In this model combined therapy with conjugated equine estrogens and medroxyprogesterone acetate induced greater proliferation than did conjugated equine estrogens alone. The percentage of estrogen receptor – positive cells was decreased in the conjugated equine estrogens plus medroxyprogesterone acetate group. The percentage of progesterone receptor -positive cells was increased by treatment with conjugated equine estrogens alone.
CONCLUSION: These results indicate a proliferative response of mammary gland epithelium to therapy with conjugated equine estrogens plus medroxyprogesterone acetate in postmenopausal macaques. The clinical implication of this finding may be a greater risk for development of breast neoplasms in women receiving combined hormone replacement therapy.
42) Reid SE, Murthy MS, Kaufman M, Scanlon EF. Endocrine and paracrine hormones in the promotion, progression and recurrence of breast cancer. Br J Surg 1996 ; 83 : 1037 – 46.
43) Santen RJ, Pinkerton J, McCartney C, Petroni GR. Risk of breast cancer with progestins
in combination with estrogen as hormone replacement therapy. J Clin Endocrinol
Metab 2001 ; 86 : 16 – 23.
44) Weiss LK, Burkman RT, Cushing-Haugen KL, Voigt LF, Simon MS, Daling JR, et al. Hormone replacement therapy regimens and breast cancer risk. Obstet Gynecol 2002; 100 : 1148 – 58.
45) Olsson HL, Ingvar C, Bladström A. Hormone replacement therapy containing progestins and given continuously increases breast carcinoma risk in Sweden. Cancer 2003 ; 97 : 1387 – 92.
46) Chlebowski RT, Hendrix SL, Langer RD, Stefanick ML, Gass M, Lane D, et al. Influence
of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal
women: the Women’s Health Initiative Randomized Trial. JAMA 2003; 289: 3243 – 53.
47) Bluming, Avrum Z. “Progesterone and breast cancer pathogenesis.” Journal of molecular endocrinology 66.1 (2021): C1-C2.
48) Bluming, Avrum Z, Estrogen Matters, Little, Brown Spark; 1st edition (September 4, 2018)
49) Fournier, Agnès, et al. “Postmenopausal hormone therapy initiation before and after the Women’s Health Initiative in two French cohorts.” Menopause (New York, NY) 18.2 (2011): 219-223.
50) Fournier, Agnes, et al. “Breast cancer risk in relation to different types of hormone replacement therapy in the E3N‐EPIC cohort.” International journal of cancer 114.3 (2005): 448-454.
51) Fournier, Agnès, Franco Berrino, and Françoise Clavel-Chapelon. “Unequal risks for breast cancer associated with different hormone replacement therapies: results from the E3N cohort study.” Breast cancer research and treatment 107 (2008): 103-111.
52) Fournier, Agnès, et al. “Use of different postmenopausal hormone therapies and risk of histology-and hormone receptor–defined invasive breast cancer.” Journal of Clinical Oncology 26.8 (2008): 1260-1268.
53) Fournier, Agnès, et al. “Estrogen-progestagen menopausal hormone therapy and breast cancer: does delay from menopause onset to treatment initiation influence risks?.” Journal of Clinical Oncology 27.31 (2009): 5138-5143.
54) Fournier, Agnès, et al. “Risk of breast cancer after stopping menopausal hormone therapy in the E3N cohort.” Breast cancer research and treatment 145 (2014): 535-543.
55) Berkson, Devaki Lindsey. “Estrogen Vindication, Part 1: Estrogen and the WHI.” Townsend Letter 445-446 (2020): 36-40.
56) Berkson, Devaki Lindsey. “Estrogen Vindication, Part 2: Estrogen, Cancer Stem Cells, and Studies.” Townsend Letter 447 (2020): 60-66. Berkson Devaki Lindsey Estrogen Vindication Part 1 and 2 Townsend Letter 445-446 2020 36-40
57) Lippman, Marc, Gall Bolan, and Karen Huff. “The effects of estrogens and antiestrogens on hormone-responsive human breast cancer in long-term tissue culture.” Cancer research 36.12 (1976): 4595-4601.
58) Ellis, Matthew J., et al. “Lower-dose vs high-dose oral estradiol therapy of hormone receptor–positive, aromatase inhibitor–resistant advanced breast cancer: A phase 2 randomized study.” Jama 302.7 (2009): 774-780.
59) Bennink, Herjan JT Coelingh, et al. “The use of high-dose estrogens for the treatment of breast cancer.” Maturitas 95 (2017): 11-23.
60) Lønning, Per E., et al. “High-dose estrogen treatment in postmenopausal breast cancer patients heavily exposed to endocrine therapy.” Breast cancer research and treatment 67 (2001): 111-116.
61) Haddow, Alexander, et al. “Influence of synthetic oestrogens on advanced malignant disease.” British medical journal 2.4368 (1944): 393.
62) Giulianelli, Sebastián, et al. “Estrogen receptor alpha mediates progestin-induced mammary tumor growth by interacting with progesterone receptors at the cyclin D1/MYC promoters.” Cancer research 72.9 (2012): 2416-2427.
63) O’Malley, Bert W., and Sohaib Khan. “Elwood V. Jensen (1920–2012): father of the nuclear receptors.” Proceedings of the National Academy of Sciences 110.10 (2013): 3707-3708.
64) Gustafsson, Jan-Åke. “Estrogen Receptor β—A Multifaceted Player.” Women’s Health and Menopause: New Strategies—Improved Quality of Life. Boston, MA: Springer US, 2002. 1-5.
65) Koehler, Konrad F., et al. “Reflections on the discovery and significance of estrogen receptor β.” Endocrine reviews 26.3 (2005): 465-478.
66) Mohammed, Hisham, et al. “Progesterone receptor modulates ERα action in breast cancer.” Nature 523.7560 (2015): 313-317.
67) Maximov, Philipp Y., et al. “Estrogen receptor complex to trigger or delay estrogen-induced apoptosis in long-term estrogen deprived breast cancer.” Frontiers in Endocrinology 13 (2022): 869562.
68) Feigman, Mary J., et al. “Pregnancy reprograms the epigenome of mammary epithelial cells and blocks the development of premalignant lesions.” Nature communications 11.1 (2020): 2649.
69) Subramani, Ramadevi, et al. “Pregnancy Inhibits Mammary Carcinogenesis by Persistently Altering the Hypothalamic–Pituitary Axis.” Cancers 13.13 (2021): 3207.
70) Barton, Maria, Julia Santucci-Pereira, and Jose Russo. “Molecular pathways involved in pregnancy-induced prevention against breast cancer.” Frontiers in endocrinology 5 (2014): 213.
74) Mal, Rahul, et al. “Estrogen receptor beta (ERβ): a ligand activated tumor suppressor.” Frontiers in oncology 10 (2020): 587386.
75) McVicker, Lauren, et al. “Vaginal estrogen therapy use and survival in females with breast cancer.” JAMA oncology 10.1 (2024): 103-108.
76) Beste, Mary E., et al. “Vaginal Estrogen Use in Breast Cancer Survivors: A Systematic Review and Meta-Analysis of Recurrence and Mortality Risks.” American Journal of Obstetrics and Gynecology (2024).
77) Agrawal, Pranjal, et al. “Safety of vaginal estrogen therapy for genitourinary syndrome of menopause in women with a history of breast cancer.” Obstetrics & Gynecology (2022): 10-1097.
78) Moegele, M., et al. “Vaginal estrogen therapy for patients with breast cancer.” Geburtshilfe und Frauenheilkunde 73.10 (2013): 1017-1022.
79) Dew, J. E., B. G. Wren, and J. A. Eden. “A cohort study of topical vaginal estrogen therapy in women previously treated for breast cancer.” Climacteric 6.1 (2003): 45-52.
80) Lambrinoudaki, Irene. “Menopausal hormone therapy and breast cancer risk: all progestogens are not the same.” Case Reports in Women’s Health 29 (2021).
81) Vinogradova Y. Use of hormone replacement therapy and risk of breast cancer: nested case-control studies using the QResearch and CPRD databases. BMJ. 2020 Oct 28;371
82) Hsieh, Robert W., et al. “Molecular characterization of a B-ring unsaturated estrogen: implications for conjugated equine estrogen components of premarin.” Steroids 73.1 (2008): 59-68.
83) Bhavnani, Bhagu R., Shui-Pang Tam, and XiaoFeng Lu. “Structure activity relationships and differential interactions and functional activity of various equine estrogens mediated via estrogen receptors (ERs) ERα and ERβ.” Endocrinology 149.10 (2008): 4857-4870.
84) Flores, Valerie A., and Hugh S. Taylor. “The effect of menopausal hormone therapies on breast cancer: avoiding the risk.” Endocrinology and Metabolism Clinics 44.3 (2015): 587-602.
85) Saltelli, Andrea, et al. “Science, the endless frontier of regulatory capture.” Futures 135 (2022): 102860.
86) Nelson, Mark M. “What is to be done? Options for combating the menace of media capture.” In the Service of Power: Media Capture and the Threat to Democracy (2017): 143.
87) Abraham, John, and Rachel Ballinger. “Science, politics, and health in the brave new world of pharmaceutical carcinogenic risk assessment: Technical progress or cycle of regulatory capture?.” Social Science & Medicine 75.8 (2012): 1433-1440.
88) Marsden, Tracy. “Bioidentical Hormone Replacement: Guiding Principles for Practice.” Nat Med J 2.3 (2010): 2010-03.
89) Plu-Bureau G, Le M, Thalabard J, et al. Percutaneous progesterone use and risk of breast cancer: results from a French cohort study of premenopausal women with benign breast disease. Cancer Detect Prev. 1999;23:290-296.
90) L’hermite M, Simoncini T, Fuller S, Genazzani AR. Could transdermal estradiol + progesterone be a safer postmenopausal HRT? A review. Maturitas. 2008;60:185-201.
91) Lieberman, Allan, and Luke Curtis. “In defense of progesterone: a review of the literature.” Alternative Therapies in Health & Medicine 23.7 (2017).
92) Ferretti, Gianluigi, Alessandra Felici, and Francesco Cognetti. “The protective side of progesterone.” Breast Cancer Research 9 (2007): 1-1.
93) Stevenson, John C., et al. “Progestogens as a component of menopausal hormone therapy: the right molecule makes the difference.” Drugs in context 9 (2020).
94) V. Craig Jordan, PhD, a Founding Father of Targeted Therapy in Cancer, Dies at Age 76
By Ronald Piana July 10, 2024
95) Ganzina, Fabrizio. “High-dose medroxyprogesterone acetate (MPA) treatment in advanced breast cancer. A review.” Tumori Journal 65.5 (1979): 563-585.
96) Bentel, Jacqueline M., et al. “Androgen receptor agonist activity of the synthetic progestin, medroxyprogesterone acetate, in human breast cancer cells.” Molecular and cellular endocrinology 154.1-2 (1999): 11-20.
97) Wood, Charles E., et al. “Effects of estradiol with micronized progesterone or medroxyprogesterone acetate on risk markers for breast cancer in postmenopausal monkeys.” Breast cancer research and treatment 101 (2007): 125-134.
98) Peters, Amelia A., et al. “Androgen receptor inhibits estrogen receptor-alpha activity and is prognostic in breast cancer.” Cancer research 69.15 (2009): 6131-6140.
99) Santen, R. J., and W. Yue. “Cause or prevention of breast cancer with estrogens: analysis from tumor biologic data, growth kinetic model and Women’s Health Initiative study.” Climacteric 22.1 (2019): 3-12.
100) Le Ray, Isabelle, et al. “Local estrogen therapy and risk of breast cancer recurrence among hormone-treated patients: a nested case-control study.” Breast cancer research and treatment 135.2 (2012): 603-609.
Women with estrogen-positive breast cancers receive endocrine treatment such as tamoxifen and aromatase inhibitors (AI) for 5-10 years. An important side effect of these drugs is vaginal dryness for which local hormonal therapy (LHT) represents the most effective treatment but is theoretically contraindicated. This study aimed to assess whether the use of LHT increases the risk of breast cancer recurrence among women receiving endocrine treatment. We conducted a cohort study with nested case-control analysis using the United Kingdom General Practice Research Database (GPRD). The cohort included female patients at least 18 years of age, newly diagnosed with breast cancer who received at least one AI or tamoxifen prescription between January 1, 1998 and June 30, 2008. Cases, who were patients experiencing a breast cancer recurrence during follow-up, were each matched with up to 10 controls based on age, date of cohort entry, type of endocrine treatment received, and duration of follow-up. Conditional logistic regression was used to estimate rate ratios (RR), and 95 % confidence intervals. A total of 13,479 women were included in the study, of which 2,673 received AIs, 10,806 received tamoxifen, and 271 received LHT. Mean (SD) age at cohort entry was 63.7 (14.1) years, and mean follow-up was 3.5 (2.6) years. The crude recurrence rate 25.9 per 1,000 per year. Overall, the use of LHT was not associated with an increased risk of recurrence (RR: 0.78, 95 % CI 0.48-1.25) compared with non-use. In stratified analyses, LHT did not increase the risk of recurrence among tamoxifen-treated patients (RR: 0.83, 95 % CI 0.51-1.34), while the risk was not estimable among AI-treated patients since no patients receiving LHT experienced a recurrence. The use of LHT is not associated with an increase in breast cancer recurrence among women receiving a hormone therapy.
101) full pdf
Laing, Abbie J., Louise Newson, and James A. Simon. “Individual benefits and risks of intravaginal estrogen and systemic testosterone in the management of women in the menopause, with a discussion of any associated risks for cancer development.” The Cancer Journal 28.3 (2022): 196-203.
Symptoms are very common, affecting approximately 70% of all menopausal women.4 Symptoms include genital changes (dryness, itching, burning, pain, and irritation), sexual concerns (dyspareunia, decreased arousal, reduced lubrication, postcoital bleeding, reduced or absent orgasm), and urinary problems (urgency, nocturia, dysuria, recurrent UTIs, and urinary incontinence).
4,5
102) Palshof T, Mouridsen HT, Daehnfeldt JL. Adjuvant endocrine therapy of
breast cancer—a controlled clinical trial of estrogen and anti-oestrogen:
preliminary results of the Copenhagen breast cancer trials.Recent Results Cancer
Res. 1980;71:185–189.
Abstract
In an attempt to clarify the value of adjuvant endocrine therapy as an alternative to other treatment modalities, we have started two controlled, prospective, double-blind trials, the Copenhagen Breast Cancer Trials. In these studies postmenopausal women are randomised after primary local treatment to treatment with DES, tamoxifen, or placebo. Premenopausal women are similarly randomised to receive treatment with tamoxifen or placebo. Entry to the trials was closed in March 1978, when 343 patients had entered the studies. The preliminary results indicate that postmenopausal patients with ER+ tumours benefit from adjuvant treatment with tamoxifen. Adjuvant treatment with DES seems to be effective in ER- postmenopausal patients. In postmenopausal patients the ER+ tumors have a lower rate of recurrence. At present these preliminary results do not permit definite conclusions.
103) Palshof T, Carstensen B, Mouridsen HT, et al. Adjuvant endocrine therapy
in pre- and postmenopausal women with operable breast cancer. Reviews
on. Endocr Relat Cancer. 1985;(suppl 17):43–50. No abstract available
104) full pdf Eigeliene N, Elo T, LinhalaM, et al. Androgens inhibit the stimulatory action
of 17β-estradiol on normal human breast tissue in explant cultures.J Clin EndocrinolMetab. 2012;97:E1116–E1127.
Background: The data concerning the effects and safety of androgen in human breast tissue are conflicting.Objective: Our aim was to analyze the effects of androgens on normal human breast tissue (HBT).Approach: We cultured explants of HBT (obtained from reduction mammoplasty operations of postmenopausal women) with or without testosterone (T) and 5α-dihydrotestosterone (DHT) or in combination with 17β-estradiol (E(2)) for 7 and 14 d to study the effects of androgens on proliferation, apoptosis, target gene expression, and steroid receptors. The androgen receptor (AR) and estrogen receptor (ER) dependences of the effects were studied with the antihormones bicalutamide and fulvestrant, respectively.
Results: The hormone responsiveness of cultured breast tissue was assessed by assaying apolipoprotein-D and prostate-specific antigen expression increased by androgens and amphiregulin and trefoil factor-1 expression induced by E(2) treatment. T and DHT reduced proliferation and increased apoptosis in breast epithelium, the effects of which were reversed by bicalutamide [androgen blocker]. In combination with E(2), they suppressed E(2)-stimulated proliferation and cell survival. DHT also inhibited basal (P < 0.05) and E(2)-induced expression of cyclin-D1 mRNA (P < 0.05). Immunohistochemistry showed that T (P < 0.05) and DHT (P < 0.05) increased the relative number of AR-positive cells, whereas ERα-positive (P < 0.001) cell numbers were strongly decreased. The percentage of ERβ-positive cells remained unchanged. E(2) treatment increased ERα-positive (P < 0.01) cells, whereas AR- (P < 0.05) and ERβ-expressing (P < 0.001) cells diminished. These effects were repressed in combination cultures of E(2) with T and DHT.
Conclusion: T and DHT inhibited proliferation and increased apoptosis in the epithelium of cultured normal HBT and opposed E(2)-stimulated proliferation and cell survival in an AR-dependent manner. These effects were associated with changes in the proportions of ERα- and AR-positive epithelial cells.
105) full pdf
Boni, Corrado, et al. “Therapeutic activity of testosterone in metastatic breast cancer.” Anticancer Research 34.3 (2014): 1287-1290.
retrospective study to assess the effectiveness and tolerability of testosterone treatment in a consecutive series of 53 patients with hormone receptor-positive breast cancer who had stopped responding following several lines of previous hormone therapy and whose disease was progressing.
Since September 2007, testosterone propionate (registered in Italy for treatment of metastatic breast cancer) has been used at the Oncology Unit of Reggio Emilia for the treatment of patients with ER/PgR-positive metastatic breast cancer whose disease was
progressing despite several lines of previous hormonal treatment. In order to critically evaluate the efficacy and toxicity of this treatment, we retrospectively studied a consecutive series of 53 patients treated between September 2007 and November 2010.
All patients were menopausal and their disease was progressing after a median of
three lines of hormonal therapy (range 2-5) and a median of three lines of chemotherapy (range 0-11). Testosterone propionate, 250 mg intramuscolarly, was given once every two weeks, twice, and then once every four weeks until progression of disease, limiting
toxicity, or death. The median duration of treatment was seven months, without dose reductions or delays. All patients had been previously responsive to at least one hormonal line of treatment and had received at least two such lines (among them, tamoxifen,
anastrozole, letrozole, exemestane, fulvestrant, and megestrol); all had been treated with an aromatase inhibitor. Most of the patients had also been treated with several types of chemotherapy before beginning treatment with testosterone.
Disease regression was seen in 9 patients (17%); regression was complete in 1 patient and partial in the other 8. Stabilization of disease was seen in 22 patients (41.5%). The total number of patients with a favorable outcome (regression or stabilization) was 31 (58.5%).
ZZZZZZZZZZZZZZZZ END OF REFERENCES ZZZZZZZZZZZZZZZZZZZZZZZZ
==============================================================
Palshof T, Mouridsen HT, Daehnfeldt JL. Adjuvant endocrine therapy of
breast cancer—a controlled clinical trial of oestrogen and anti-oestrogen:
preliminary results of the Copenhagen breast cancer trials.Recent Results Cancer
Res. 1980;71:185–189.
Abstract
In an attempt to clarify the value of adjuvant endocrine therapy as an alternative to other treatment modalities, we have started two controlled, prospective, double-blind trials, the Copenhagen Breast Cancer Trials. In these studies postmenopausal women are randomised after primary local treatment to treatment with DES, tamoxifen, or placebo. Premenopausal women are similarly randomised to receive treatment with tamoxifen or placebo. Entry to the trials was closed in March 1978, when 343 patients had entered the studies. The preliminary results indicate that postmenopausal patients with ER+ tumours benefit from adjuvant treatment with tamoxifen. Adjuvant treatment with DES seems to be effective in ER- postmenopausal patients. In postmenopausal patients the ER+ tumors have a lower rate of recurrence. At present these preliminary results do not permit definite conclusions.
62. Palshof T, Carstensen B, Mouridsen HT, et al. Adjuvant endocrine therapy
in pre- and postmenopausal women with operable breast cancer. Reviews
on. Endocr Relat Cancer. 1985;(suppl 17):43–50. No abstract available
63. Stoll BA, Parbhoo S. Treatment of menopausal symptoms in breast cancer
patients. Lancet. 1988;1:1278–1279.joint Breast Clinic, Academic Department of Surgery, Royal Free Hospital, London NW3 2QG, United Kingdon
No abstract available
64. Powles TJ, Hickish T, Casey S, et al. Hormone replacement after breast
cancer. Lancet. 1993;342:60–61.
65. Eden JA, Bush T, Nand S, et al. A case-controlled study of combined continuous estrogen-progestin replacement therapy amongst women with a
personal history of breast cancer. Menopause. 1995;2:67–72.
66. Vassilopoulou-Sellin R, Theriault R, Klein MJ. Estrogen replacement
therapy in women with prior diagnosis and treatment for breast cancer.
Gynecol Oncol. 1997;65:89–93.
67. Dew J, Eden J, Beller E, et al. A cohort study of hormone replacement
therapy given to women previously treated for breast cancer. Climacteric.
1998;1:137–142.
68. Espie M, Gorins A, Perret F, et al. Hormone replacement therapy (HRT) in
patients treated for breast cancer: analysis of a cohort of 120 patients. Proc
ASCO. 1999(abstract);18:2262.
69. Guidozzi F. Estrogen replacement therapy in breast cancer survivors. Int J
Gynecol Obstet. 1999;64:59–63.
20 premarin and medroxyprogesterone and four tibolone.
70. Natrajan PK, Soumakis K, Gambrell RD Jr. Estrogen replacement therapy
in women with previous breast cancer. Obstet Gynecol. 1999;181:288–295.
71. Uršič-Vrščaj M, Bebar S. A case-control study of hormone replacement
therapy after primary surgical breast cancer treatment. Eur J Surg Oncol.
1999;25:146–151.
72. Vassilopoulou-Sellin R, Asmar L, Hortobagyi GN, et al. Estrogen replacement therapy after localized breast cancer: clinical outcome of 319 women
followed prospectively. J Clin Oncol. 1999;17:1482–1487.
73. Disaia PJ, Brewster WR, Ziogas A, et al. Breast cancer survival and hormone
replacement therapy. A cohort analysis. Am J Clin Oncol. 2000;23:541–545.
74. Marsden J, Whitehead M, A’Hern R, et al. Are randomized trials of hormone replacement therapy in symptomatic women with breast cancer feasible? Fertil Steril. 2000;73:292–299.
75. Peters GN, Fodera T, Sabol J, et al. Estrogen replacement therapy after breast cancer: a 12-year follow-up. Ann Surg Oncol. 2001;8:828–832.
76. Marttunen MB, Hietanen P, Pyrhonen S, et al. A prospective study on
women with a history of breast cancer and with or without estrogen replacement therapy. Maturitas. 2001;39:217–225.
77. Beckmann MW, Jap D, Djahansouzi S, et al. Hormone replacement therapy after treatment of breast cancer: effects on postmenopausal symptoms, bone mineral density and recurrence rates. Oncology. 2001; 60:199–20
78. O’Meara ES, Rossing MA, Daling JR, et al. Hormone replacement therapy after a diagnosis of breast cancer in relation to recurrence and mortality. J Natl Cancer Inst. 2001;93:754–762.
79. Vassilopoulou-Sellin R, Cohen DS, Hortobagyi GN, et al. Estrogen replacement therapy for menopausal women with a history of breast carcinoma: results of a 5-year, prospective study. Cancer. 2002;95:1817–1826.
80. Durna EM, Wren BG, Heller GZ, et al. Hormone replacement therapy after a diagnosis of breast cancer: cancer recurrence and mortality. Med J Aust. 2002;177:347–351.
81. Decker DA, Pettinga JE, VanderVelde N, et al. Estrogen replacement therapy in breast cancer survivors: a matched-controlled series. Menopause.
2003;10:277–285.
82. Gorins A, Espié M, Bedairia N, et al. Hormone replacement therapy in breast cancer patients: a study of 230 patients, with a case-control study. Gynecol Obstet Fertil. 2003;31:614–619.
France
83. Holmberg L, Anderson H. HABITS (hormonal replacement therapy after
breast cancer—is it safe?), a randomised comparison: trial stopped. Lancet. 2004;363:453–455.
Regional Oncologic Centre, University Hospital, SE-751 85 Uppsala, Sweden.
84. Holmberg L, Iversen OE, Rudenstam CM, et al. Increased risk of recurrence after hormone replacement therapy in breast cancer survivors.J Natl Cancer Inst. 2008;100:475–482.
Regional Oncologic Centre, University Hospital, SE-751 85 Uppsala, Sweden.
85. von Schoultz E, Rutqvist LE, Stockholm Breast Cancer Study Group. Menopausal hormone therapy after breast cancer: the Stockholm randomized trial. J Natl Cancer Inst. 2005;97:533–535.
86. Fahlén M, Fornander T, Johansson H, et al. Hormone replacement therapy after breast cancer: 10 year follow-up of the Stockholm randomized trial. Eur J Cancer. 2013;49:52–59.
87. Bluming AZ. Hormone replacement therapy (HRT) in women with previously treated primary breast cancer: update XIV. Proc ASCO. J Clin Oncol.2008;15 s:20693.
88. Figueiredo JC, Bernstein L, Capanu M, et al. Oral contraceptives, postmenopausal hormones, and risk of asynchronous bilateral breast cancer: the WECARE study group. J Clin Oncol. 2008;26:1411–1418
=====================================================
Chlebowski, Rowan T., et al. “Breast cancer after use of estrogen plus progestin in postmenopausal women.” New England journal of medicine 360.6 (2009): 573-587.
Following the release of the 2002 report of the Women’s Health Initiative (WHI) trial of estrogen plus progestin, the use of menopausal hormone therapy in the United States decreased substantially. Subsequently, the incidence of breast cancer also dropped, suggesting a cause-and-effect relation between hormone treatment and breast cancer. However, the cause of this decrease remains controversial.
Methods
We analyzed the results of the WHI randomized clinical trial — in which one study group received 0.625 mg of conjugated equine estrogens plus 2.5 mg of medroxyprogesterone acetate daily and another group received placebo — and examined temporal trends in breast-cancer diagnoses in the WHI observational-study cohort. Risk factors for breast cancer, frequency of mammography, and time-specific incidence of breast cancer were assessed in relation to combined hormone use.
Results
In the clinical trial, there were fewer breast-cancer diagnoses in the group receiving estrogen plus progestin than in the placebo group in the initial 2 years of the study, but the number of diagnoses increased over the course of the 5.6-year intervention period. The elevated risk decreased rapidly after both groups stopped taking the study pills, despite a similar frequency of mammography. In the observational study, the incidence of breast cancer was initially about two times as high in the group receiving menopausal hormones as in the placebo group, but this difference in incidence decreased rapidly in about 2 years, coinciding with year-to-year reductions in combined hormone use. During this period, differences in the frequency of mammography between the two groups were unchanged.
Conclusions
The increased risk of breast cancer associated with the use of estrogen plus progestin declined markedly soon after discontinuation of combined hormone therapy and was unrelated to changes in frequency of mammography.
===========================================
VAGINAL ESTROGEN – Breast Cancer Survivors
Le Ray, Isabelle, et al. “Local estrogen therapy and risk of breast cancer recurrence among hormone-treated patients: a nested case-control study.” Breast cancer research and treatment 135.2 (2012): 603-609.
Dijon Cedex, France.
13,479 women
United Kingdom General Practice Research Database (GPRD).
2,673 received AIs,
10,806 received tamoxifen, and
271 received LHT.local hormonal therapy
Exact HRT type and dosage not seen in abstract
Abstract
Women with estrogen-positive breast cancers receive endocrine treatment such as tamoxifen and aromatase inhibitors (AI) for 5-10 years. An important side effect of these drugs is vaginal dryness for which local hormonal therapy (LHT) represents the most effective treatment but is theoretically contraindicated. This study aimed to assess whether the use of LHT increases the risk of breast cancer recurrence among women receiving endocrine treatment. We conducted a cohort study with nested case-control analysis using the United Kingdom General Practice Research Database (GPRD).
The cohort included female patients at least 18 years of age, newly diagnosed with breast cancer who received at least one AI or tamoxifen prescription between January 1, 1998 and June 30, 2008. Cases, who were patients experiencing a breast cancer recurrence during follow-up, were each matched with up to 10 controls based on age, date of cohort entry, type of endocrine treatment received, and duration of follow-up. Conditional logistic regression was used to estimate rate ratios (RR), and 95 % confidence intervals.
A total of 13,479 women were included in the study, of which 2,673 received AIs, 10,806 received tamoxifen, and 271 received LHT. Mean (SD) age at cohort entry was 63.7 (14.1) years, and mean follow-up was 3.5 (2.6) years.
The crude recurrence rate 25.9 per 1,000 per year.
Overall, the use of LHT was not associated with an increased risk of recurrence (RR: 0.78, 95 % CI 0.48-1.25) compared with non-use. In stratified analyses, LHT did not increase the risk of recurrence among tamoxifen-treated patients (RR: 0.83, 95 % CI 0.51-1.34), while the risk was not estimable among AI-treated patients since no patients receiving LHT experienced a recurrence.The use of LHT is not associated with an increase in breast cancer recurrence among women receiving a hormone therapy.
===================
The dream team , perfect study on HRT for breast cancer survivors
1) metabolic testing for 2 and 4 hydroxy-estrogens
2) genetic testing for SNPs involving estrogen metabolism
3) stratify patients according to length of time of LTED- longer LTED associated with better outcome with lesss recurrence.
4) record type of estrogen/progesterone and dosage and delivery
premarin CEE common in US
estradiol common in Europe
estriol (Bi-Est)?? usually not used. requires compounding pharmacy.
Progesterone dosage – cancer protective
or synthetic progestin – carcinogenic – increased recurrence
5) record route of delivery Oral-blood clots, transdermal safest
==================================
163 JAMA. 1994 Aug 17; 272(7):540-5. Estrogen replacement therapy in breast cancer survivors. A time for change. Breast Cancer Committees of the Eastern Cooperative Oncology Group.
164 Avrum Bluming MD, Carol Tavris PhD. Estrogen Matters: Why Taking Hormones in Menopause Can Improve Women’s Well-Being and Lengthen Their Lives — Without Raising the Risk of Breast Cancer 1st Edition. Hachett Book Group 2018.
165 Proc ASCO J Clini Oncol. 2008.15s:20693. Hormone Replacement Therapy (HRT) in women with previously treated primary breast cancer: Update XIV.
166 JAMA. 1995; 273:620-21. Estrogen Replacement therapy in breast cancer survivors. Letter to the editor.
167 JAMA. 1994; 272:540-45. Estrogen Replacement therapy in breast cancer survivors: A time for change. Breast Cancer Committees of the Eastern Cooperative Oncology Group.
168 Gynecol Endocrinol. 2002; 16:469-78. A case-control study of hormone replacement therapy in women with a history of breast cancer.
169 Surg Oncol. 1999; 25:146-51. A case-control study of combined continuous estrogen-progestin replacement therapy among women with a personal history of breast cancer.
170 Climacteric 1998; 1:137-42. A cohort study of hormone replacement therapy given to women previously treated for breast cancer.
171 Climacteric. 2002; 5:151-55. Tamoxifen, hormone receptors and hormone replacement therapy in women previously treated for breast cancer: A cohort study.
172 Med J Aust. 2002177:347-51. Hormone replacement therapy after a diagnosis of breast cancer. Cancer recurrence and mortality.
173 Climacteric. 2004; 7:284-91. Breast cancer in premenopausal women: Recurrence and survival rates and relationship to hormone replacement therapy.
174 Proc ASCO. 1999(abstract); 18:2262. Hormone replacement therapy (HRT) in patients treated for breast cancer: Analysis of cohort of 120 patients.
175 Maturitas. 200139; 217-25. A Prospective study on women with a history of breast cancer and with or without estrogen replacement therapy.
176 Oncol. 2001; 60:199-206. Hormone Replacement therapy after breast cancer: Effects on postmenopausal symptoms, bone mineral density and recurrence rates.
177 J Clin Oncol. 1999; 17:1482-92. Estrogen replacement therapy after localized breast cancer; Clinical outcome of 319 women followed prospectively.
178 Int J Fertili Women’s Med. 1999; 44:186-92. Experience with estrogen replacement therapy in breast cancer survivors.
179 Menopause 2003; 10:277-85. Estrogen replacement therapy in breast cancer survivors: A matched-controlled series.
180 J Natl Cancer Instit. 2001; 93754-62. Hormone Replacement Therapy after a diagnosis of breast cancer in relation to recurrence and mortality.
181 Medscape Oncology and Medscape Internal Medicine Dec. 13 2019 Megan Brooks News > Medscape Medical News > Conference News > SABCS 2019
182 Maturitas. 2006; 53:123-32. Menopausal hormone therapy (HT) in patients with breast cancer.
183 J Natl Cancer Inst. 2005; 97:533-35. Menopausal hormone therapy after breast cancer. The Stockholm randomized trial.
184 Eur J Cancer. 2013; 49:52-59. Hormone replacement therapy after breast cancer: 10 yr. follow-up of the Stockholm randomized trial.
185 N Engl J Med. 1991; 325; 800-802. Uncertainty about postmenopausal estrogen: Time for action, not debate.
190 Avrum Bluming MD, Carol Tavris PhD. Estrogen Matters: Why Taking Hormones in Menopause Can Improve Women’s Well-Being and Lengthen Their Lives — Without Raising the Risk of Breast Cancer 1st Edition. Hachett Book Group 2018 Page 50.
191 Berkson, DL. “How to Prescribe Hormones in Breast Cancer Patients” a CME approved talk. Presented at: Private M.D. Function; Salt Lake City Utah; 2019
192 J Clin Oncol. 1999 May; 17(5):1482-7. Estrogen replacement therapy after localized breast cancer: clinical outcome of 319 women followed prospectively.
193 Int J Fertil Womens Med. 1999 Jun-Aug; 44(4):186-92. An experience with estrogen replacement therapy in breast cancer survivors.
194 Menopause. 2003 Jul-Aug; 10(4):277-85. Estrogen replacement therapy in breast cancer survivors: a matched-controlled series.
195 Ann Surg Oncol. 2001 Dec; 8(10):828-32. Estrogen replacement therapy after breast cancer: a 12-year follow-up
196 J Natl Cancer Inst. 2001 May 16; 93(10):754-62. Hormone replacement therapy after a diagnosis of breast cancer in relation to recurrence and mortality.
197 Fam Pract. 2002 Dec; 51(12):1056-62. Cancer recurrence and mortality in women using hormone replacement therapy: meta-analysis.
198 Eur J Surg Oncol. 1999 Apr; 25(2):146-51. A case-control study of hormone replacement therapy after primary surgical breast cancer treatment.
199 Menopause 1995:2; 67-72. A case-control study of combined continuous estrogen-progestin replacement therapy among women with a personal history of breast cancer.
200 Climacteric. 1998 Jun; 1(2):137-42. A cohort study of hormone replacement therapy given to women previously treated for breast cancer.
201 Med J Aust. 2002 Oct 7; 177(7):347-51. Hormone replacement therapy after a diagnosis of breast cancer: cancer recurrence and mortality.
202 Proc ASCO. 1999 (abstract):18; 2262. Hormone replacement therapy (HRT) in patients treated for breast cancer: Analysis of a cohort of 120 patients.
203 Maturitas. 2001 Sep 28; 39(3):217-25. A prospective study on women with a history of breast cancer and with or without estrogen replacement therapy.
204 Oncology. 2001; 60(3):199-206. Hormone replacement therapy after treatment of breast cancer: effects on postmenopausal symptoms, bone mineral density and recurrence rates.
========================================================
Meta-Analysis of 8 studies
Meurer, Linda N., and Sarah Lena. “Cancer recurrence and mortality in women using hormone replacement therapy after breast cancer: Meta-analysis.” Journal of family practice 51.12 (2002): 1056-1064.
Department of Family and Community Medicine, Medical
College of Wisconsin, Milwaukee, WI
Studies included 717 subjects who used hormone replacement therapy (HRT) at
some time after their diagnosis of breast cancer, as well as 2545 subjects who did not use HRT.
A meta-analysis of these studies showed that breast cancer survivors using ERT experienced no increase in the risk of recurrence compared with nonusers (8.2% vs 10.2%; RR,0.72, 95% confidence interval [CI], 0.47–1.10).
Six studies were included in a combined analysis of overall mortality (Figure 2). The ERT users in these studies experienced significantly fewer deaths (3.0%) than the nonusers (11.4%) over the combined study periods (RR, 0.18; 95% CI, 0.10–0.31; numbers needed to treat = 12). Subanalyses of those studies in which groups were comparable showed similar results (RR, 0.21; 95% CI, 0.10–0.46).
1.Beckmann MW, Jap D, Djahansouzi S, et al. Hormone replacement
therapy after treatment of breast cancer: effects on postmenopausal
symptoms, bone mineral density and recurrence rates. Oncology
2001; 60:199–206
2.Dew J, Eden J, Beller E, et al. A cohort study of hormone replacement
therapy given to women previously treated for breast cancer.
Climacteric 1998; 1:137–42.
3. DiSaia PJ, Grosen EA, Kurosaki T, Gildea M, Cowan B, Anton-Culver
H. Hormone replacement therapy in breast cancer survivors: a
cohort study [see comments]. Am J Obstet Gynecol 1996;
174:1494–8.
4. DiSaia PJ, Brewster WR, Ziogas A, Anton-Culver H. Breast cancer
survival and hormone replacement therapy: a cohort analysis. Am J
Clin Oncol 2000; 23:541–5.
5. Eden J, Bush T, Natrajan PK, Wren B. A case-control study of combined
continuous estrogen–progestin replacement therapy among
women with a personal history of breast cancer. Menopause 1995;
2:67–72.
6.Habel LA, Daling JR, Newcoomb PA, et al. Risk of recurrence after
ductal carcinoma in situ of the breast. Cancer Epidemiol Biomarkers
Prev 1998; 7:689–96.
7. Marsden J, Whitehead M, A’Hern R, Baum M, Sacks N. Are randomized
trials of hormone replacement therapy in symptomatic
women with breast cancer feasible? Fertil Steril 2000; 73:292–9.
8. Natrajan PK, Soumakis K, Gambrell RD Jr. Estrogen replacement
therapy in women with previous breast cancer. Am J Obstet
Gynecol 1999; 181:288–95.
9. Ursic-Vrscaj M, Bebar S. A case-control study of hormone replacement
therapy after primary surgical breast cancer treatment. Eur J
Surg Oncol 1999; 25:146–51.
10.Vassilopoulou-Sellin R, Theriault R, Klein MJ. Estrogen replacement
therapy in women with prior diagnosis and treatment for breast cancer.
ynecol Oncol 1997; 65:89–93.
Jeffrey Dach MD
7450 Griffin Road, Suite 190
Davie, Fl 33314
954-792-4663
my blog: www.jeffreydachmd.com
Natural Thyroid Toolkit by Jeffrey Dach MD
Cracking Cancer Toolkit by Jeffrey Dach MD
Heart Book by Jeffrey Dach MD
www.naturalmedicine101.com
www.bioidenticalhormones101.com
www.truemedmd.com
www.drdach.com
Click Here for: Dr Dach’s Online Store for Pure Encapsulations Supplements
Click Here for: Dr Dach’s Online Store for Nature’s Sunshine Supplements
Web Site and Discussion Board Links:
jdach1.typepad.com/blog/
disc.yourwebapps.com/Indices/244066.html
disc.yourwebapps.com/Indices/244067.html
http://sci.med.narkive.com/covV2Qo2/jeffrey-dach-book-announcment-natural-medicine-101
The reader is advised to discuss the comments on these pages with his/her personal physicians and to only act upon the advice of his/her personal physician. Also note that concerning an answer which appears as an electronically posted question, I am NOT creating a physician — patient relationship. Although identities will remain confidential as much as possible, as I can not control the media, I can not take responsibility for any breaches of confidentiality that may occur.
Link to this Article
Copyright © 2024 Jeffrey Dach MD All Rights Reserved. This article may be reproduced on the internet without permission, provided there is a link to this page and proper credit is given. See Repost Guidelines.
FAIR USE NOTICE: This site contains copyrighted material the use of which has not always been specifically authorized by the copyright owner. We are making such material available in our efforts to advance understanding of issues of significance. We believe this constitutes a ‘fair use’ of any such copyrighted material as provided for in section 107 of the US Copyright Law. In accordance with Title 17 U.S.C. Section 107, the material on this site is distributed without profit to those who have expressed a prior interest in receiving the included information for research and educational purposes.
Serving Areas of: Hollywood, Aventura, Miami, Fort Lauderdale, Pembroke Pines, Miramar, Davie, Coral Springs, Cooper City, Sunshine Ranches, Hallandale, Surfside, Miami Beach, Sunny Isles, Normandy Isles, Coral Gables, Hialeah, Golden Beach ,Kendall,sunrise, coral springs, parkland,pompano, boca raton, palm beach, weston, dania beach, tamarac, oakland park, boynton beach, delray,lake worth,wellington,plantation
Leave a Reply
You must be logged in to post a comment.