fbpx
Wikipedia

Medroxyprogesterone acetate

Medroxyprogesterone acetate (MPA), also known as depot medroxyprogesterone acetate (DMPA) in injectable form and sold under the brand name Depo-Provera among others, is a hormonal medication of the progestin type.[10][4] It is used as a method of birth control and as a part of menopausal hormone therapy.[10][4] It is also used to treat endometriosis, abnormal uterine bleeding, paraphilia, and certain types of cancer.[10] The medication is available both alone and in combination with an estrogen.[11][12] It is taken by mouth, used under the tongue, or by injection into a muscle or fat.[10]

Medroxyprogesterone acetate
Clinical data
Pronunciation/mɛˌdrɒksiprˈɛstərn ˈæsɪtt/ me-DROKS-ee-proh-JES-tər-ohn ASS-i-tayt[1]
Trade namesProvera, Depo-Provera, Depo-SubQ Provera 104, Curretab, Cycrin, Farlutal, Gestapuran, Perlutex, Veramix, others[2]
Other namesMPA; DMPA; Methylhydroxyprogesterone acetate; Methylacetoxyprogesterone; MAP; Methypregnone; Metipregnone; 6α-Methyl-17α-hydroxyprogesterone acetate; 6α-Methyl-17α-acetoxyprogesterone; 6α-Methyl-17α-hydroxypregn-4-ene-3,20-dione acetate; NSC-26386
AHFS/Drugs.comMonograph
MedlinePlusa604039
Routes of
administration
By mouth, sublingual, intramuscular injection, subcutaneous injection
Drug classProgestogen; Progestin; Progestogen ester; Antigonadotropin; Steroidal antiandrogen
ATC code
Legal status
Legal status
  • AU: S4 (Prescription only)
  • US: WARNING[3]Rx-only
  • In general: ℞ (Prescription only)
Pharmacokinetic data
BioavailabilityBy mouth: ~100%[4][5]
Protein binding88% (to albumin)[5]
MetabolismLiver (hydroxylation (CYP3A4), reduction, conjugation)[6][4][9]
Elimination half-lifeBy mouth: 12–33 hours[6][4]
IM (aq. susp.): ~50 days[7]
SC (aq. susp.): ~40 days[8]
ExcretionUrine (as conjugates)[6]
Identifiers
  • [(6S,8R,9S,10R,13S,14S,17R)-17-acetyl-6,10,13-trimethyl-3-oxo-2,6,7,8,9,11,12,14,15,16-decahydro-1H-cyclopenta[a]phenanthren-17-yl] acetate
CAS Number
  • 71-58-9
PubChem CID
  • 6279
IUPHAR/BPS
  • 2879
DrugBank
  • DB00603
ChemSpider
  • 6043
UNII
  • C2QI4IOI2G
KEGG
  • C08150 Y
ChEBI
  • CHEBI:6716
ChEMBL
  • ChEMBL717 Y
CompTox Dashboard (EPA)
  • DTXSID0025527
ECHA InfoCard100.000.689
Chemical and physical data
FormulaC24H34O4
Molar mass386.532 g·mol−1
3D model (JSmol)
  • Interactive image
Melting point207 to 209 °C (405 to 408 °F)
  • C[C@H]1C[C@@H]2[C@H](CC[C@]3([C@H]2CC[C@@]3(C(=O)C)OC(=O)C)C)[C@@]4(C1=CC(=O)CC4)C
  • InChI=InChI=1S/C24H34O4/c1-14-12-18-19(22(4)9-6-17(27)13-21(14)22)7-10-23(5)20(18)8-11-24(23,15(2)25)28-16(3)26/h13-14,18-20H,6-12H2,1-5H3/t14-,18+,19-,20-,22+,23-,24-/m0/s1
  • Key:PSGAAPLEWMOORI-PEINSRQWSA-N
  (verify)

Common side effects include menstrual disturbances such as absence of periods, abdominal pain, and headaches.[10] More serious side effects include bone loss, blood clots, allergic reactions, and liver problems.[10] Use is not recommended during pregnancy as it may harm the baby.[10] MPA is an artificial progestogen, and as such activates the progesterone receptor, the biological target of progesterone.[4] It also has androgenic activity and weak glucocorticoid activity. Due to its progestogenic activity, MPA decreases the body's release of gonadotropins and can suppress sex hormone levels.[13] It works as a form of birth control by preventing ovulation.[10]

MPA was discovered in 1956 and was introduced for medical use in the United States in 1959.[14][15][10] It is on the World Health Organization's List of Essential Medicines.[16] MPA is the most widely used progestin in menopausal hormone therapy and in progestogen-only birth control.[17][18] DMPA is approved for use as a form of long-acting birth control in more than 100 countries.[19][20] In 2020, it was the 214th most commonly prescribed medication in the United States, with more than 2 million prescriptions.[21][22]

Medical uses edit

The most common use of MPA is in the form of DMPA as a long-acting progestogen-only injectable contraceptive to prevent pregnancy in women. It is an extremely effective contraceptive when used with relatively high doses to prevent ovulation. MPA is also used in combination with an estrogen in menopausal hormone therapy in postmenopausal women to treat and prevent menopausal symptoms such as hot flashes, vaginal atrophy, and osteoporosis.[4] It is used in menopausal hormone therapy specifically to prevent endometrial hyperplasia and cancer that would otherwise be induced by prolonged unopposed estrogen therapy in women with intact uteruses.[4][23] In addition to contraception and menopausal hormone therapy, MPA is used in the treatment of gynecological and menstrual disorders such as dysmenorrhea, amenorrhea, and endometriosis.[24] Along with other progestins, MPA was developed to allow for oral progestogen therapy, as progesterone (the progestogen hormone made by the human body) could not be taken orally for many decades before the process of micronization was developed and became feasible in terms of pharmaceutical manufacturing.[25]

DMPA reduces sex drive in men and is used as a form of chemical castration to control inappropriate or unwanted sexual behavior in those with paraphilias or hypersexuality, including in convicted sex offenders.[26][27] DMPA has also been used to treat benign prostatic hyperplasia, as a palliative appetite stimulant for cancer patients, and at high doses (800 mg per day) to treat certain hormone-dependent cancers including endometrial cancer, renal cancer, and breast cancer.[28][29][30][31][32] MPA has also been prescribed in feminizing hormone therapy for transgender women due to its progestogenic and functional antiandrogenic effects.[33] It has been used to delay puberty in children with precocious puberty but is not satisfactory for this purpose as it is not able to completely suppress puberty.[34] DMPA at high doses has been reported to be definitively effective in the treatment of hirsutism as well.[35]

Though not used as a treatment for epilepsy, MPA has been found to reduce the frequency of seizures and does not interact with antiepileptic medications. MPA does not interfere with blood clotting and appears to improve blood parameters for women with sickle cell anemia. Similarly, MPA does not appear to affect liver metabolism, and may improve primary biliary cirrhosis and chronic active hepatitis. Women taking MPA may experience spotting shortly after starting the medication but is not usually serious enough to require medical intervention. With longer use amenorrhea (absence of menstruation) can occur as can irregular menstruation which is a major source of dissatisfaction, though both can result in improvements with iron deficiency and risk of pelvic inflammatory disease and often do not result in discontinuation of the medication.[29]

Birth control edit

Depot medroxyprogesterone acetate (DMPA)
 
Background
TypeHormonal
First use1969[36]
Trade namesDepo-Provera, Depo-SubQ Provera 104, others
AHFS/Drugs.comdepo-provera
Failure rates (first year)
Perfect use0.2%[37]
Typical use6%[37]
Usage
Duration effect3 months
(12–14 weeks)
Reversibility3–18 months
User remindersMaximum interval is just under 3 months
Clinic review12 weeks
Advantages and disadvantages
STI protectionNo
Period disadvantagesEspecially in first injection may be frequent spotting
Period advantagesUsually no periods from 2nd injection
BenefitsEspecially good if poor pill compliance.
Reduced endometrial cancer risk.
RisksReduced bone density, which may reverse after discontinuation
Medical notes
For those intending to start family, suggest switch 6 months prior to alternative method (e.g. POP) allowing more reliable return fertility.

DMPA, under brand names such as Depo-Provera and Depo-SubQ Provera 104, is used in hormonal birth control as a long-lasting progestogen-only injectable contraceptive to prevent pregnancy in women.[38][39] It is given by intramuscular or subcutaneous injection and forms a long-lasting depot, from which it is slowly released over a period of several months. It takes one week to take effect if given after the first five days of the period cycle, and is effective immediately if given during the first five days of the period cycle. Estimates of first-year failure rates are about 0.3%.[40]

Effectiveness edit

Trussell's estimated perfect use first-year failure rate for DMPA as the average of failure rates in seven clinical trials at 0.3%.[40][41] It was considered perfect use because the clinical trials measured efficacy during actual use of DMPA defined as being no longer than 14 or 15 weeks after an injection (i.e., no more than 1 or 2 weeks late for a next injection).

Prior to 2004, Trussell's typical use failure rate for DMPA was the same as his perfect use failure rate: 0.3%.[42]

  • DMPA estimated typical use first-year failure rate = 0.3% in:
    • Contraceptive Technology, 16th revised edition (1994)[43]
    • Contraceptive Technology, 17th revised edition (1998)[44]
      • Adopted in 1998 by the FDA for its current Uniform Contraceptive Labeling guidance[45]

In 2004, using the 1995 NSFG failure rate, Trussell increased (by 10 times) his typical use failure rate for DMPA from 0.3% to 3%.[40][41]

  • DMPA estimated typical use first-year failure rate = 3% in:
    • Contraceptive Technology, 18th revised edition (2004)[40]
    • Contraceptive Technology, 19th revised edition (2007)[46]

Trussell did not use 1995 NSFG failure rates as typical use failure rates for the other two then newly available long-acting contraceptives, the Norplant implant (2.3%) and the ParaGard copper T 380A IUD (3.7%), which were (as with DMPA) an order of magnitude higher than in clinical trials. Since Norplant and ParaGard allow no scope for user error, their much higher 1995 NSFG failure rates were attributed by Trussell to contraceptive overreporting at the time of a conception leading to a live birth.[40][47][41]

Advantages edit

DMPA has a number of advantages and benefits:[48][49][39][50]

The United Kingdom Department of Health has actively promoted Long Acting Reversible Contraceptive use since 2008, particularly for young people;[58] following on from the October 2005 National Institute for Health and Clinical Excellence guidelines.[59] Giving advice on these methods of contraception has been included in the 2009 Quality and Outcomes Framework "good practice" for primary care.[60]

Comparison edit

Proponents of bioidentical hormone therapy believe that progesterone offers fewer side effects and improved quality of life compared to MPA.[61] The evidence for this view has been questioned; MPA is better absorbed when taken by mouth, with a much longer elimination half-life leading to more stable blood levels[62] though it may lead to greater breast tenderness and more sporadic vaginal bleeding.[61] The two compounds do not differentiate in their ability to suppress endometrial hyperplasia,[61] nor does either increase the risk of pulmonary embolism.[63] The two medications have not been adequately compared in direct tests to clear conclusions about safety and superiority.[25]

Available forms edit

MPA is available alone in the form of 2.5, 5, and 10 mg oral tablets, as a 150 mg/mL (1 mL) or 400 mg/mL (2.5 mL) microcrystalline aqueous suspension for intramuscular injection, and as a 104 mg (0.65 mL of 160 mg/mL) microcrystalline aqueous suspension for subcutaneous injection.[64][65] It has also been marketed in the form of 100, 200, 250, 400, and 500 mg oral tablets; 500 and 1,000 mg oral suspensions; and as a 50 mg/mL microcrystalline aqueous suspension for intramuscular injection.[66][67] A 100 mg/mL microcrystalline aqueous suspension for intramuscular injection was previously available as well.[64] In addition to single-drug formulations, MPA is available in the form of oral tablets in combination with conjugated estrogens (CEEs), estradiol, and estradiol valerate for use in menopausal hormone therapy, and is available in combination with estradiol cypionate in a microcrystalline aqueous suspension as a combined injectable contraceptive.[11][12][64][19]

Depo-Provera is the brand name for a 150 mg microcrystalline aqueous suspension of DMPA that is administered by intramuscular injection. The shot must be injected into thigh, buttock, or deltoid muscle four times a year (every 11 to 13 weeks), and provides pregnancy protection instantaneously after the first injection.[68] Depo-subQ Provera 104 is a variation of the original intramuscular DMPA that is instead a 104 mg microcrystalline dose in aqueous suspension administered by subcutaneous injection. It contains 69% of the MPA found in the original intramuscular DMPA formulation. It can be injected using a smaller injection needle inserting the medication just below the skin, instead of into the muscle, in either the abdomen or thigh. This subcutaneous injection claims to reduce the side effects of DMPA while still maintaining all the same benefits of the original intramuscular DMPA.

Contraindications edit

MPA is not usually recommended because of unacceptable health risk or because it is not indicated in the following cases:[69][70]

Conditions where the theoretical or proven risks usually outweigh the advantages of using DMPA:

Conditions which represent an unacceptable health risk if DMPA is used:

Conditions where use is not indicated and should not be initiated:

MPA is not recommended for use prior to menarche or before or during recovery from surgery.[71]

Side effects edit

In women, the most common adverse effects of MPA are acne, changes in menstrual flow, drowsiness, and can cause birth defects if taken by pregnant women. Other common side effects include breast tenderness, increased facial hair, decreased scalp hair, difficulty falling or remaining asleep, stomach pain, and weight loss or gain.[24] Lowered libido has been reported as a side effect of MPA in women.[72] DMPA can affect menstrual bleeding. After a year of use, 55% of women experience amenorrhea (missed periods); after two years, the rate rises to 68%. In the first months of use "irregular or unpredictable bleeding or spotting, or, rarely, heavy or continuous bleeding" was reported.[73] MPA does not appear to be associated with vitamin B12 deficiency.[74] Data on weight gain with DMPA likewise are inconsistent.[75][76]

At high doses for the treatment of breast cancer, MPA can cause weight gain and can worsen diabetes mellitus and edema (particularly of the face). Adverse effects peak at five weeks, and are reduced with lower doses. Less frequent effects may include thrombosis (though it is not clear if this is truly a risk, it cannot be ruled out), painful urination, headache, nausea, and vomiting. When used as a form of androgen deprivation therapy in men, more frequent complaints include reduced libido, impotence, reduced ejaculate volume, and within three days, chemical castration. At extremely high doses (used to treat cancer, not for contraception) MPA may cause adrenal suppression and may interfere with carbohydrate metabolism, but does not cause diabetes.[29]

When used as a form of injected birth control, there is a delayed return of fertility. The average return to fertility is 9 to 10 months after the last injection, taking longer for overweight or obese women. By 18 months after the last injection, fertility is the same as that in former users of other contraceptive methods.[48][49] Fetuses exposed to progestogens have demonstrated higher rates of genital abnormalities, low birth weight, and increased ectopic pregnancy particularly when MPA is used as an injected form of long-term birth control. A study of accidental pregnancies among poor women in Thailand found that infants who had been exposed to DMPA during pregnancy had a higher risk of low birth weight and an 80% greater-than-usual chance of dying in the first year of life.[77]

Mood changes edit

There have been concerns about a possible risk of depression and mood changes with progestins like MPA, and this has led to reluctance of some clinicians and women to use them.[78][79] However, contrary to widely-held beliefs, most research suggests that progestins do not cause adverse psychological effects such as depression or anxiety.[78] A 2018 systematic review of the relationship between progestin-based contraception and depression included three large studies of DMPA and reported no association between DMPA and depression.[80] According to a 2003 review of DMPA, the majority of published clinical studies indicate that DMPA is not associated with depression, and the overall data support the notion that the medication does not significantly affect mood.[81]

In the largest study to have assessed the relationship between MPA and depression to date, in which over 3,900 women were treated with DMPA for up to 7 years, the incidence of depression was infrequent at 1.5% and the discontinuation rate due to depression was 0.5%.[80][38][82] This study did not include baseline data on depression,[82] and due to the incidence of depression in the study, the FDA required package labeling for DMPA stating that women with depression should be observed carefully and that DMPA should be discontinued if depression recurs.[80] A subsequent study of 495 women treated with DMPA over the course of 1 year found that the mean depression score slightly decreased in the whole group of continuing users from 7.4 to 6.7 (by 9.5%) and decreased in the quintile of that group with the highest depression scores at baseline from 15.4 to 9.5 (by 38%).[82] Based on the results of this study and others, a consensus began emerging that DMPA does not in fact increase the risk of depression nor worsen the severity of pre-existing depression.[76][82][38]

Similarly to the case of DMPA for hormonal contraception, the Heart and Estrogen/Progestin Replacement Study (HERS), a study of 2,763 postmenopausal women treated with 0.625 mg/day oral CEEs plus 2.5 mg/day oral MPA or placebo for 36 months as a method of menopausal hormone therapy, found no change in depressive symptoms.[83][84][85] However, some small studies have reported that progestins like MPA might counteract beneficial effects of estrogens against depression.[78][4][86]

Long-term effects edit

The Women's Health Initiative investigated the use of a combination of oral CEEs and MPA compared to placebo. The study was prematurely terminated when previously unexpected risks were discovered, specifically the finding that though the all-cause mortality was not affected by the hormone therapy, the benefits of menopausal hormone therapy (reduced risk of hip fracture, colorectal and endometrial cancer and all other causes of death) were offset by increased risk of coronary heart disease, breast cancer, strokes and pulmonary embolism.[87]

When combined with CEEs, MPA has been associated with an increased risk of breast cancer, dementia, and thrombus in the eye. In combination with estrogens in general, MPA may increase the risk of cardiovascular disease, with a stronger association when used by postmenopausal women also taking CEEs. It was because of these unexpected interactions that the Women's Health Initiative study was ended early due to the extra risks of menopausal hormone therapy,[88] resulting in a dramatic decrease in both new and renewal prescriptions for hormone therapy.[89]

Long-term studies of users of DMPA have found slight or no increased overall risk of breast cancer. However, the study population did show a slightly increased risk of breast cancer in recent users (DMPA use in the last four years) under age 35, similar to that seen with the use of combined oral contraceptive pills.[73]

Results of the Women's Health Initiative (WHI) menopausal hormone therapy randomized controlled trials
Clinical outcome Hypothesized
effect on risk
Estrogen and progestogen
(CEs 0.625 mg/day p.o. + MPA 2.5 mg/day p.o.)
(n = 16,608, with uterus, 5.2–5.6 years follow up)
Estrogen alone
(CEs 0.625 mg/day p.o.)
(n = 10,739, no uterus, 6.8–7.1 years follow up)
HR 95% CI AR HR 95% CI AR
Coronary heart disease Decreased 1.24 1.00–1.54 +6 / 10,000 PYs 0.95 0.79–1.15 −3 / 10,000 PYs
Stroke Decreased 1.31 1.02–1.68 +8 / 10,000 PYs 1.37 1.09–1.73 +12 / 10,000 PYs
Pulmonary embolism Increased 2.13 1.45–3.11 +10 / 10,000 PYs 1.37 0.90–2.07 +4 / 10,000 PYs
Venous thromboembolism Increased 2.06 1.57–2.70 +18 / 10,000 PYs 1.32 0.99–1.75 +8 / 10,000 PYs
Breast cancer Increased 1.24 1.02–1.50 +8 / 10,000 PYs 0.80 0.62–1.04 −6 / 10,000 PYs
Colorectal cancer Decreased 0.56 0.38–0.81 −7 / 10,000 PYs 1.08 0.75–1.55 +1 / 10,000 PYs
Endometrial cancer 0.81 0.48–1.36 −1 / 10,000 PYs
Hip fractures Decreased 0.67 0.47–0.96 −5 / 10,000 PYs 0.65 0.45–0.94 −7 / 10,000 PYs
Total fractures Decreased 0.76 0.69–0.83 −47 / 10,000 PYs 0.71 0.64–0.80 −53 / 10,000 PYs
Total mortality Decreased 0.98 0.82–1.18 −1 / 10,000 PYs 1.04 0.91–1.12 +3 / 10,000 PYs
Global index 1.15 1.03–1.28 +19 / 10,000 PYs 1.01 1.09–1.12 +2 / 10,000 PYs
Diabetes 0.79 0.67–0.93 0.88 0.77–1.01
Gallbladder disease Increased 1.59 1.28–1.97 1.67 1.35–2.06
Stress incontinence 1.87 1.61–2.18 2.15 1.77–2.82
Urge incontinence 1.15 0.99–1.34 1.32 1.10–1.58
Peripheral artery disease 0.89 0.63–1.25 1.32 0.99–1.77
Probable dementia Decreased 2.05 1.21–3.48 1.49 0.83–2.66
Abbreviations: CEs = conjugated estrogens. MPA = medroxyprogesterone acetate. p.o. = per oral. HR = hazard ratio. AR = attributable risk. PYs = person–years. CI = confidence interval. Notes: Sample sizes (n) include placebo recipients, which were about half of patients. "Global index" is defined for each woman as the time to earliest diagnosis for coronary heart disease, stroke, pulmonary embolism, breast cancer, colorectal cancer, endometrial cancer (estrogen plus progestogen group only), hip fractures, and death from other causes. Sources: See template.

Blood clots edit

DMPA has been associated in multiple studies with a higher risk of venous thromboembolism (VTE) when used as a form of progestogen-only birth control in premenopausal women.[90][91][92][93] The increase in incidence of VTE ranges from 2.2-fold to 3.6-fold.[90][91][92][93] Elevated risk of VTE with DMPA is unexpected, as DMPA has little or no effect on coagulation and fibrinolytic factors,[94][95] and progestogens by themselves normally do not increase the risk of thrombosis.[91][92] It has been argued that the higher incidence with DMPA has reflected preferential prescription of DMPA to women considered to be at an increased risk of VTE.[91] Alternatively, it is possible that MPA may be an exception among progestins in terms of VTE risk.[96][97][98] A 2018 meta-analysis reported that MPA was associated with a 2.8-fold higher risk of VTE than other progestins.[97] It is possible that the glucocorticoid activity of MPA may increase the risk of VTE.[4][99][98]

Bone density edit

DMPA may cause reduced bone density in premenopausal women and in men when used without an estrogen, particularly at high doses, though this appears to be reversible to a normal level even after years of use.

On 17 November 2004, the United States Food and Drug Administration put a black box warning on the label, indicating that there were potential adverse effects of loss of bone mineral density.[100][101] While it causes temporary bone loss, most women fully regain their bone density after discontinuing use.[75] The World Health Organization (WHO) recommends that the use not be restricted.[102][103] The American College of Obstetricians and Gynecologists notes that the potential adverse effects on BMD be balanced against the known negative effects of unintended pregnancy using other birth control methods or no method, particularly among adolescents.

Three studies have suggested that bone loss is reversible after the discontinuation of DMPA.[104][105][106] Other studies have suggested that the effect of DMPA use on postmenopausal bone density is minimal,[107] perhaps because DMPA users experience less bone loss at menopause.[108] Use after peak bone mass is associated with increased bone turnover but no decrease in bone mineral density.[109]

The FDA recommends that DMPA not be used for longer than two years, unless there is no viable alternative method of contraception, due to concerns over bone loss.[101] However, a 2008 Committee Opinion from the American Congress of Obstetricians and Gynecologists (ACOG) advises healthcare providers that concerns about bone mineral density loss should neither prevent the prescription of or continuation of DMPA beyond two years of use.[110]

HIV risk edit

There is uncertainty regarding the risk of HIV acquisition among DMPA users; some observational studies suggest an increased risk of HIV acquisition among women using DMPA, while others do not.[111] The World Health Organization issued statements in February 2012 and July 2014 saying the data did not warrant changing their recommendation of no restriction – Medical Eligibility for Contraception (MEC) category 1 – on the use of DMPA in women at high risk for HIV.[112][113] Two meta-analyses of observational studies in sub-Saharan Africa were published in January 2015.[114] They found a 1.4- to 1.5-fold increase risk of HIV acquisition for DMPA users relative to no hormonal contraceptive use.[115][116] In January 2015, the Faculty of Sexual & Reproductive Healthcare of the Royal College of Obstetricians and Gynaecologists issued a statement reaffirming that there is no reason to advise against use of DMPA in the United Kingdom even for women at 'high risk' of HIV infection.[117] A systematic review and meta-analysis of risk of HIV infection in DMPA users published in fall of 2015 stated that "the epidemiological and biological evidence now make a compelling case that DMPA adds significantly to the risk of male-to-female HIV transmission."[118] In 2019, a randomized controlled trial found no significant association between DMPA use and HIV.[119]

Breastfeeding edit

MPA may be used by breastfeeding mothers. Heavy bleeding is possible if given in the immediate postpartum time and is best delayed until six weeks after birth. It may be used within five days if not breast feeding. While a study showed "no significant difference in birth weights or incidence of birth defects" and "no significant alternation of immunity to infectious disease caused by breast milk containing DMPA", a subgroup of babies whose mothers started DMPA at two days postpartum had a 75% higher incidence of doctor visits for infectious diseases during their first year of life.[120]

A larger study with longer follow-up concluded that "use of DMPA during pregnancy or breastfeeding does not adversely affect the long-term growth and development of children". This study also noted that "children with DMPA exposure during pregnancy and lactation had an increased risk of suboptimal growth in height," but that "after adjustment for socioeconomic factors by multiple logistic regression, there was no increased risk of impaired growth among the DMPA-exposed children." The study also noted that effects of DMPA exposure on puberty require further study, as so few children over the age of 10 were observed.[121]

Overdose edit

MPA has been studied at "massive" dosages of up to 5,000 mg per day orally and 2,000 mg per day via intramuscular injection, without major tolerability or safety issues described.[122][123][124] Overdose is not described in the Food and Drug Administration (FDA) product labels for injected MPA (Depo-Provera or Depo-SubQ Provera 104).[7][8] In the FDA product label for oral MPA (Provera), it is stated that overdose of an estrogen and progestin may cause nausea and vomiting, breast tenderness, dizziness, abdominal pain, drowsiness, fatigue, and withdrawal bleeding.[6] According to the label, treatment of overdose should consist of discontinuation of MPA therapy and symptomatic care.[6]

Interactions edit

MPA increases the risk of breast cancer, dementia, and thrombus when used in combination with CEEs to treat menopausal symptoms.[71] When used as a contraceptive, MPA does not generally interact with other medications. The combination of MPA with aminoglutethimide to treat metastases from breast cancer has been associated with an increase in depression.[29] St John's wort may decrease the effectiveness of MPA as a contraceptive due to acceleration of its metabolism.[71]

Pharmacology edit

Pharmacodynamics edit

MPA acts as an agonist of the progesterone, androgen, and glucocorticoid receptors (PR, AR, and GR, respectively),[5] activating these receptors with EC50 values of approximately 0.01 nM, 1 nM, and 10 nM, respectively.[125] It has negligible affinity for the estrogen receptor.[5] The medication has relatively high affinity for the mineralocorticoid receptor, but in spite of this, it has no mineralocorticoid or antimineralocorticoid activity.[4] The intrinsic activities of MPA in activating the PR and the AR have been reported to be at least equivalent to those of progesterone and dihydrotestosterone (DHT), respectively, indicating that it is a full agonist of these receptors.[126][127]

Relative affinities (%) of MPA and related steroids
Progestogen
PR AR ER GR MR
Progesterone 50 0 0 10 100
Chlormadinone acetate 67 5 0 8 0
Cyproterone acetate 90 6 0 6 8
Medroxyprogesterone acetate 115 5 0 29 160
Megestrol acetate 65 5 0 30 0
Notes: Values are percentages (%). Reference ligands (100%) were promegestone for the PR, metribolone for the AR, estradiol for the ER, dexamethasone for the GR, and aldosterone for the MR. Sources: [4]

Progestogenic activity edit

MPA is a potent agonist of the progesterone receptor with similar affinity and efficacy relative to progesterone.[128] While both MPA and its deacetylated analogue medroxyprogesterone bind to and agonize the PR, MPA has approximately 100-fold higher binding affinity and transactivation potency in comparison.[128] As such, unlike MPA, medroxyprogesterone is not used clinically, though it has seen some use in veterinary medicine.[2] The oral dosage of MPA required to inhibit ovulation (i.e., the effective contraceptive dosage) is 10 mg/day, whereas 5 mg/day was not sufficient to inhibit ovulation in all women.[129] In accordance, the dosage of MPA used in oral contraceptives in the past was 10 mg per tablet.[130] For comparison to MPA, the dosage of progesterone required to inhibit ovulation is 300 mg/day, whereas that of the 19-nortestosterone derivatives norethisterone and norethisterone acetate is only 0.4 to 0.5 mg/day.[131]

The mechanism of action of progestogen-only contraceptives like DMPA depends on the progestogen activity and dose. High-dose progestogen-only contraceptives, such as DMPA, inhibit follicular development and prevent ovulation as their primary mechanism of action.[132][133] The progestogen decreases the pulse frequency of gonadotropin-releasing hormone (GnRH) release by the hypothalamus, which decreases the release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) by the anterior pituitary. Decreased levels of FSH inhibit follicular development, preventing an increase in estradiol levels. Progestogen negative feedback and the lack of estrogen positive feedback on LH release prevent a LH surge. Inhibition of follicular development and the absence of a LH surge prevent ovulation.[48][49] A secondary mechanism of action of all progestogen-containing contraceptives is inhibition of sperm penetration by changes in the cervical mucus.[134] Inhibition of ovarian function during DMPA use causes the endometrium to become thin and atrophic. These changes in the endometrium could, theoretically, prevent implantation. However, because DMPA is highly effective in inhibiting ovulation and sperm penetration, the possibility of fertilization is negligible. No available data support prevention of implantation as a mechanism of action of DMPA.[134]

MPA and related steroids at the progesterone receptor
Compound Ki (nM) EC50 (nM)a EC50 (nM)b
Progesterone 4.3 0.9 25
Medroxyprogesterone 241 47 32
Medroxyprogesterone acetate 1.2 0.6 0.15
Footnotes: a = Coactivator recruitment. b = Reporter cell line. Sources: [128]
Oral potencies of MPA and related steroids
Progestogen OID
(mg/day)
TFD
(mg/cycle)
TFD
(mg/day)
ODP
(mg/day)
ECD
(mg/day)
Progesterone 300 4200 200–300 200
Chlormadinone acetate 1.7 20–30 10 2.0 5–10
Cyproterone acetate 1.0 20 1.0 2.0 1.0
Medroxyprogesterone acetate 10 50 5–10 ? 5.0
Megestrol acetate ? 50 ? ? 5.0
Abbreviations: OID = ovulation-inhibiting dosage (without additional estrogen). TFD = endometrial transformation dosage. ODP = oral dosage in commercial contraceptive preparations. ECD = estimated comparable dosage. Sources: [131][99][135]
Parenteral potencies and durations of progestogens[a][b]
Compound Form Dose for specific uses (mg)[c] DOA[d]
TFD[e] POICD[f] CICD[g]
Algestone acetophenide Oil soln. - 75–150 14–32 d
Gestonorone caproate Oil soln. 25–50 8–13 d
Hydroxyprogest. acetate[h] Aq. susp. 350 9–16 d
Hydroxyprogest. caproate Oil soln. 250–500[i] 250–500 5–21 d
Medroxyprog. acetate Aq. susp. 50–100 150 25 14–50+ d
Megestrol acetate Aq. susp. - 25 >14 d
Norethisterone enanthate Oil soln. 100–200 200 50 11–52 d
Progesterone Oil soln. 200[i] 2–6 d
Aq. soln. ? 1–2 d
Aq. susp. 50–200 7–14 d
Notes and sources:
  1. ^ Sources: [136][137][138][139][140][141][142][143][144][145][146][147][148][149][150][151][152][153][154]
  2. ^ All given by intramuscular or subcutaneous injection.
  3. ^ Progesterone production during the luteal phase is ~25 (15–50) mg/day. The OID of OHPC is 250 to 500 mg/month.
  4. ^ Duration of action in days.
  5. ^ Usually given for 14 days.
  6. ^ Usually dosed every two to three months.
  7. ^ Usually dosed once monthly.
  8. ^ Never marketed or approved by this route.
  9. ^ a b In divided doses (2 × 125 or 250 mg for OHPC, 10 × 20 mg for P4).

Antigonadotropic and anticorticotropic effects edit

MPA suppresses the hypothalamic–pituitary–adrenal (HPA) and hypothalamic–pituitary–gonadal (HPG) axes at sufficient dosages, resulting decreased levels of gonadotropins, androgens, estrogens, adrenocorticotropic hormone (ACTH), and cortisol, as well as levels of sex hormone-binding globulin (SHBG).[13] There is evidence that the suppressive effects of MPA on the HPG axis are mediated by activation of both the PR and the AR in the pituitary gland.[155][156] Due to its effects on androgen levels, MPA can produce strong functional antiandrogenic effects, and is used in the treatment of androgen-dependent conditions such as precocious puberty in boys and hypersexuality in men.[157] In addition, since the medication suppresses estrogen levels as well, MPA can produce strong functional antiestrogenic effects similarly, and has been used to treat estrogen-dependent conditions such as precocious puberty in girls and endometriosis in women. Due to low estrogen levels, the use of MPA without an estrogen poses a risk of decreased bone mineral density and other symptoms of estrogen deficiency.[158]

Oral MPA has been found to suppress testosterone levels in men by about 30% (from 831 ng/dL to 585 ng/dL) at a dosage of 20 mg/day, by about 45 to 75% (average 60%; to 150–400 ng/dL) at a dosage of 60 mg/day,[159][160][161] and by about 70 to 75% (from 832–862 ng/dL to 214–251 ng/dL) at a dosage of 100 mg/day.[162][163] Dosages of oral MPA of 2.5 to 30 mg/day in combination with estrogens have been used to help suppress testosterone levels in transgender women.[164][165][166][167][168][169] One study of injectable MPA in men with benign prostatic hyperplasia reported that a single 150 mg dose suppressed testosterone levels into the defined male castrate range (<58 ng/dL) within 7 days and that castration levels of testosterone were maintained for 3 months.[170] Very high doses of intramuscular MPA of 150 to 500 mg per week (but up to 900 mg per week) have similarly been reported to suppress testosterone levels to less than 100 ng/dL.[159][171] The typical initial dose of intramuscular MPA for testosterone suppression in men with paraphilias is 400 or 500 mg per week.[159]

Androgenic activity edit

MPA is a potent full agonist of the AR. Its activation of the AR may play an important and major role in its antigonadotropic effects and in its beneficial effects against breast cancer.[155][172][173] However, although MPA may produce androgenic side effects such as acne and hirsutism in some women,.[174][175] In fact, likely due to its suppressive actions on androgen levels, it has been reported that MPA is generally highly effective in improving pre-existing symptoms of hirsutism in women with the condition.[176][177] However, MPA has been seen to cause androgenic effects in children with precocious puberty.[178] The reason for the general lack of virilizing effects with MPA, despite it binding to and activating the AR with high affinity and this action potentially playing an important role in many of its physiological and therapeutic effects, is not entirely clear. However, MPA has been found to interact with the AR differently compared to other agonists of the receptor such as dihydrotestosterone (DHT).[126] The result of this difference appears to be that MPA binds to the AR with a similar affinity and intrinsic activity to that of DHT, but requires about 100-fold higher concentrations for a comparable induction of gene transcription, while at the same time not antagonizing the transcriptional activity of normal androgens like DHT at any concentration.[126] Thus, this may explain the low propensity of MPA for producing androgenic side effects.[126]

MPA shows weak androgenic effects on liver protein synthesis, similarly to other weakly androgenic progestins like megestrol acetate and 19-nortestosterone derivatives.[4][9] While it does not antagonize estrogen-induced increases in levels of triglycerides and HDL cholesterol, DMPA every other week may decrease levels of HDL cholesterol.[4] In addition, MPA has been found to suppress sex hormone-binding globulin (SHBG) production by the liver.[9][179][180] At a dosage of 10 mg/day oral MPA, it has been found to decrease circulating SHBG levels by 14 to 18% in women taking 4 mg/day oral estradiol valerate.[9] Conversely, in a study that combined 2.5 mg/day oral MPA with various oral estrogens, no influence of MPA on estrogen-induced increases in SHBG levels was discerned.[180] In another, higher-dose study, SHBG levels were lower by 59% in a group of women treated with 50 mg/day oral MPA alone relative to an untreated control group of women.[179] In massive-dose studies of oral or injectable MPA (e.g., 500–1,000 mg/day), the medication decreased SHBG levels by about 80%.[181][182][183]

Unlike the related steroids megestrol acetate and cyproterone acetate, MPA is not an antagonist of the AR and does not have direct antiandrogenic activity.[4] As such, although MPA is sometimes described as an antiandrogen, it is not a "true" antiandrogen (i.e., AR antagonist).[160]

Glucocorticoid activity edit

As an agonist of the GR, MPA has glucocorticoid activity, and as a result can cause symptoms of Cushing's syndrome,[184] steroid diabetes, and adrenal insufficiency at sufficiently high doses.[185] It has been suggested that the glucocorticoid activity of MPA may contribute to bone loss.[186] The glucocorticoid activity of MPA may also result in an upregulation of the thrombin receptor in blood vessel walls, which may contribute to procoagulant effects of MPA and risk of venous thromboembolism and atherosclerosis.[4] The relative glucocorticoid activity of MPA is among the highest of the clinically used progestins.[4]

Glucocorticoid activity of selected steroids in vitro
Steroid Class TR ()a GR (%)b
Dexamethasone Corticosteroid ++ 100
Ethinylestradiol Estrogen 0
Etonogestrel Progestin + 14
Gestodene Progestin + 27
Levonorgestrel Progestin 1
Medroxyprogesterone acetate Progestin + 29
Norethisterone Progestin 0
Norgestimate Progestin 1
Progesterone Progestogen + 10
Footnotes: a = Thrombin receptor (TR) upregulation (↑) in vascular smooth muscle cells (VSMCs). b = RBA (%) for the glucocorticoid receptor (GR). Strength: – = No effect. + = Pronounced effect. ++ = Strong effect. Sources: [187]

Steroidogenesis inhibition edit

MPA has been found to act as a competitive inhibitor of rat 3α-hydroxysteroid dehydrogenase (3α-HSD).[188][189][190][191] This enzyme is essential for the transformation of progesterone, deoxycorticosterone, and DHT into inhibitory neurosteroids such as allopregnanolone, THDOC, and 3α-androstanediol, respectively.[192] MPA has been described as very potent in its inhibition of rat 3α-HSD, with an IC50 of 0.2 μM and a Ki (in rat testicular homogenates) of 0.42 μM.[188][189] However, inhibition of 3α-HSD by MPA does not appear to have been confirmed using human proteins yet, and the concentrations required with rat proteins are far above typical human therapeutic concentrations.[188][189]

MPA has been identified as a competitive inhibitor of human 3β-hydroxysteroid dehydrogenase/Δ5-4 isomerase II (3β-HSD II).[193] This enzyme is essential for the biosynthesis of sex steroids and corticosteroids.[193] The Ki of MPA for inhibition of 3β-HSD II is 3.0 μM, and this concentration is reportedly near the circulating levels of the medication that are achieved by very high therapeutic dosages of MPA of 5 to 20 mg/kg/day (dosages of 300 to 1,200 mg/day for a 60 kg (132 lb) person).[193] Aside from 3β-HSD II, other human steroidogenic enzymes, including cholesterol side-chain cleavage enzyme (P450scc/CYP11A1) and 17α-hydroxylase/17,20-lyase (CYP17A1), were not found to be inhibited by MPA.[193] MPA has been found to be effective in the treatment of gonadotropin-independent precocious puberty and in breast cancer in postmenopausal women at high dosages, and inhibition of 3β-HSD II could be responsible for its effectiveness in these conditions.[193]

GABAA receptor allosteric modulation edit

Progesterone, via transformation into neurosteroids such as 5α-dihydroprogesterone, 5β-dihydroprogesterone, allopregnanolone, and pregnanolone (catalyzed by the enzymes 5α- and 5β-reductase and 3α- and 3β-HSD), is a positive allosteric modulator of the GABAA receptor, and is associated with a variety of effects mediated by this property including dizziness, sedation, hypnotic states, mood changes, anxiolysis, and cognitive/memory impairment, as well as effectiveness as an anticonvulsant in the treatment of catamenial epilepsy.[192][194] It has also been found to produce anesthesia via this action in animals when administered at sufficiently high dosages.[194] MPA was found to significantly reduce seizure incidence when added to existing anticonvulsant regimens in 11 of 14 women with uncontrolled epilepsy, and has also been reported to induce anesthesia in animals, raising the possibility that it might modulate the GABAA receptor similarly to progesterone.[195][196]

MPA shares some of the same metabolic routes of progesterone and, analogously, can be transformed into metabolites such as 5α-dihydro-MPA (DHMPA) and 3α,5α-tetrahydro-MPA (THMPA).[195] However, unlike the reduced metabolites of progesterone, DHMPA and THMPA have been found not to modulate the GABAA receptor.[195] Conversely, unlike progesterone, MPA itself actually modulates the GABAA receptor, although notably not at the neurosteroid binding site.[195] However, rather than act as a potentiator of the receptor, MPA appears to act as a negative allosteric modulator.[195] Whereas the reduced metabolites of progesterone enhance binding of the benzodiazepine flunitrazepam to the GABAA receptor in vitro, MPA can partially inhibit the binding of flunitrazepam by up to 40% with half-maximal inhibition at 1 μM.[195] However, the concentrations of MPA required for inhibition are high relative to therapeutic concentrations, and hence, this action is probably of little or no clinical relevance.[195] The lack of potentiation of the GABAA receptor by MPA or its metabolites is surprising in consideration of the apparent anticonvulsant and anesthetic effects of MPA described above, and they remain unexplained.[195]

Clinical studies using massive dosages of up to 5,000 mg/day oral MPA and 2,000 mg/day intramuscular MPA for 30 days in women with advanced breast cancer have reported "no relevant side effects", which suggests that MPA has no meaningful direct action on the GABAA receptor in humans even at extremely high dosages.[122]

Appetite stimulation edit

Although MPA and the closely related medication megestrol acetate are effective appetite stimulants at very high dosages,[197] the mechanism of action of their beneficial effects on appetite is not entirely clear. However, glucocorticoid, cytokine, and possibly anabolic-related mechanisms are all thought to possibly be involved, and a number of downstream changes have been implicated, including stimulation of the release of neuropeptide Y in the hypothalamus, modulation of calcium channels in the ventromedial hypothalamus, and inhibition of the secretion of proinflammatory cytokines including IL-1α, IL-1β, IL-6, and TNF-α, actions that have all been linked to an increase in appetite.[198]

Other activity edit

MPA weakly stimulates the proliferation of MCF-7 breast cancer cells in vitro, an action that is independent of the classical PRs and is instead mediated via the progesterone receptor membrane component-1 (PGRMC1).[199] Certain other progestins are also active in this assay, whereas progesterone acts neutrally.[199] It is unclear if these findings may explain the different risks of breast cancer observed with progesterone, dydrogesterone, and other progestins such as medroxyprogesterone acetate and norethisterone in clinical studies.[200]

Pharmacokinetics edit

Absorption edit

Surprisingly few studies have been conducted on the pharmacokinetics of MPA at postmenopausal replacement dosages.[201][4] The bioavailability of MPA with oral administration is approximately 100%.[4] A single oral dose of 10 mg MPA has been found to result in peak MPA levels of 1.2 to 5.2 ng/mL within 2 hours of administration using radioimmunoassay.[201][202] Following this, levels of MPA decreased to 0.09 to 0.35 ng/mL 12 hours post-administration.[201][202] In another study, peak levels of MPA were 3.4 to 4.4 ng/mL within 1 to 4 hours of administration of 10 mg oral MPA using radioimmunoassay.[201][203] Subsequently, MPA levels fell to 0.3 to 0.6 ng/mL 24 hours after administration.[201][203] In a third study, MPA levels were 4.2 to 4.4 ng/mL after an oral dose of 5 mg MPA and 6.0 ng/mL after an oral dose of 10 mg MPA, both using radioimmunoassay as well.[201][204]

Treatment of postmenopausal women with 2.5 or 5 mg/day MPA in combination with estradiol valerate for two weeks has been found to rapidly increase circulating MPA levels, with steady-state concentrations achieved after three days and peak concentrations occurring 1.5 to 2 hours after ingestion.[4][205] With 2.5 mg/day MPA, levels of the medication were 0.3 ng/mL (0.8 nmol/L) in women under 60 years of age and 0.45 ng/mL (1.2 nmol/L) in women 65 years of age or over, and with 5 mg/day MPA, levels were 0.6 ng/mL (1.6 nmol/L) in women under 60 years of age and in women 65 years of age or over.[4][205] Hence, area-under-curve levels of the medication were 1.6 to 1.8 times higher in those who were 65 years of age or older relative to those who were 60 years of age or younger.[9][205] As such, levels of MPA have been found to vary with age, and MPA may have an increased risk of side effects in elderly postmenopausal women.[9][4][205] This study assessed MPA levels using liquid-chromatography–tandem mass spectrometry (LC–MS/MS), a more accurate method of blood determinations.[205]

Oral MPA tablets can be administered sublingually instead of orally.[206][207][208] Rectal administration of MPA has also been studied.[209]

With intramuscular administration of 150 mg microcrystalline MPA in aqueous suspension, the medication is detectable in the circulation within 30 minutes, serum concentrations vary but generally plateau at 1.0 ng/mL (2.6 nmol/L) for 3 months.[210] Following this, there is a gradual decline in MPA levels, and the medication can be detected in the circulation for as long as 6 to 9 months post-injection.[210] The particle size of MPA crystals significantly influences its rate of absorption into the body from the local tissue depot when used as a microcrystalline aqueous suspension via intramuscular injection.[211][212][213] Smaller crystals dissolve faster and are absorbed more rapidly, resulting in a shorter duration of action.[211][212][213] Particle sizes can differ between different formulations of MPA, potentially influencing clinical efficacy and tolerability.[211][212][213][214]

Distribution edit

The plasma protein binding of MPA is 88%.[4][9] It is weakly bound to albumin and is not bound to sex hormone-binding globulin or corticosteroid-binding globulin.[4][9]

Metabolism edit

The elimination half-life of MPA via oral administration has been reported as both 11.6 to 16.6 hours[6] and 33 hours,[4] whereas the elimination half-lives with intramuscular and subcutaneous injection of microcrystalline MPA in aqueous suspension are 50 and 40 days, respectively.[7][8] The metabolism of MPA is mainly via hydroxylation, including at positions C6β, C21, C2β, and C1β, mediated primarily via CYP3A4, but 3- and 5-dihydro and 3,5-tetrahydro metabolites of MPA are also formed.[4][9] Deacetylation of MPA and its metabolites (into, e.g., medroxyprogesterone) has been observed to occur in non-human primate research to a substantial extent as well (30 to 70%).[215] MPA and/or its metabolites are also metabolized via conjugation.[71] The C6α methyl and C17α acetoxy groups of MPA make it more resistant to metabolism and allow for greater bioavailability than oral progesterone.[9]

Elimination edit

MPA is eliminated 20 to 50% in urine and 5 to 10% in feces following intravenous administration.[216] Less than 3% of a dose is excreted in unconjugated form.[216]

Level–effect relationships edit

With intramuscular administration, the high levels of MPA in the blood inhibit luteinizing hormone and ovulation for several months, with an accompanying decrease in serum progesterone to below 0.4 ng/mL.[210] Ovulation resumes when once blood levels of MPA fall below 0.1 ng/mL.[210] Serum estradiol remains at approximately 50 pg/mL for approximately four months post-injection (with a range of 10–92 pg/mL after several years of use), rising once MPA levels fall below 0.5 ng/mL.[210]

Hot flashes are rare while MPA is found at significant blood levels in the body, and the vaginal lining remains moist and creased. The endometrium undergoes atrophy, with small, straight glands and a stroma that is decidualized. Cervical mucus remains viscous. Because of its steady blood levels over the long term and multiple effects that prevent fertilization, MPA is a very effective means of birth control.[210]

Time–concentration curves edit

Chemistry edit

MPA is a synthetic pregnane steroid and a derivative of progesterone and 17α-hydroxyprogesterone.[220][2] Specifically, it is the 17α-acetate ester of medroxyprogesterone or the 6α-methylated analogue of hydroxyprogesterone acetate.[220][2] MPA is known chemically as 6α-methyl-17α-acetoxyprogesterone or as 6α-methyl-17α-acetoxypregn-4-ene-3,20-dione, and its generic name is a contraction of 6α-methyl-17α-hydroxyprogesterone acetate.[220][2] MPA is closely related to other 17α-hydroxyprogesterone derivatives such as chlormadinone acetate, cyproterone acetate, and megestrol acetate, as well as to medrogestone and nomegestrol acetate.[220][2] 9α-fluoromedroxyprogesterone acetate (FMPA), the C9α fluoro analogue of MPA and an angiogenesis inhibitor with two orders of magnitude greater potency in comparison to MPA, was investigated for the potential treatment of cancers but was never marketed.[221][222]

History edit

MPA was independently discovered in 1956 by Syntex and the Upjohn Company.[14][15][223][224] It was first introduced on 18 June 1959 by Upjohn in the United States under the brand name Provera (2.5, 5, and 10 mg tablets) for the treatment of amenorrhea, metrorrhagia, and recurrent miscarriage.[225][226] An intramuscular formulation of MPA, now known as DMPA (400 mg/mL MPA), was also introduced, under the brand name brand name Depo-Provera, in 1960 in the U.S. for the treatment of endometrial and renal cancer.[28] MPA in combination with ethinylestradiol was introduced in 1964 by Upjohn in the U.S. under the brand name Provest (10 mg MPA and 50 μg ethinylestradiol tablets) as an oral contraceptive, but this formulation was discontinued in 1970.[227][228][130] This formulation was marketed by Upjohn outside of the U.S. under the brand names Provestral and Provestrol, while Cyclo-Farlutal (or Ciclofarlutal) and Nogest-S[229] were formulations available outside of the U.S. with a different dosage (5 mg MPA and 50 or 75 μg ethinylestradiol tablets).[230][231]

Following its development in the late 1950s, DMPA was first assessed in clinical trials for use as an injectable contraceptive in 1963.[232] Upjohn sought FDA approval of intramuscular DMPA as a long-acting contraceptive under the brand name Depo-Provera (150 mg/mL MPA) in 1967, but the application was rejected.[233][234] However, this formulation was successfully introduced in countries outside of the United States for the first time in 1969, and was available in over 90 countries worldwide by 1992.[36] Upjohn attempted to gain FDA approval of DMPA as a contraceptive again in 1978, and yet again in 1983, but both applications failed similarly to the 1967 application.[233][234] However, in 1992, the medication was finally approved by the FDA, under the brand name Depo-Provera, for use in contraception.[233] A subcutaneous formulation of DMPA was introduced in the United States as a contraceptive under the brand name Depo-SubQ Provera 104 (104 mg/0.65 mL MPA) in December 2004, and subsequently was also approved for the treatment of endometriosis-related pelvic pain.[235]

MPA has also been marketed widely throughout the world under numerous other brand names such as Farlutal, Perlutex, and Gestapuran, among others.[2][11]

Society and culture edit

Generic names edit

Medroxyprogesterone acetate is the generic name of the drug and its INN, USAN, BAN, and JAN, while medrossiprogesterone is the DCIT and médroxyprogestérone the DCF of its free alcohol form.[220][12][2][236][11] It is also known as 6α-methyl-17α-acetoxyprogesterone (MAP) or 6α-methyl-17α-hydroxyprogesterone acetate.[220][12][2][11]

Brand names edit

MPA is marketed under a large number of brand names throughout the world.[11][12][2] Its most major brand names are Provera as oral tablets and Depo-Provera as an aqueous suspension for intramuscular injection.[11][12][2] A formulation of MPA as an aqueous suspension for subcutaneous injection is also available in the United States under the brand name Depo-SubQ Provera 104.[11][12] Other brand names of MPA formulated alone include Farlutal and Sayana for clinical use and Depo-Promone, Perlutex, Promone-E, and Veramix for veterinary use.[11][12][2] In addition to single-drug formulations, MPA is marketed in combination with the estrogens CEEs, estradiol, and estradiol valerate.[11][12][2] Brand names of MPA in combination with CEEs as oral tablets in different countries include Prempro, Premphase, Premique, Premia, and Premelle.[11][12][2] Brand names of MPA in combination with estradiol as oral tablets include Indivina and Tridestra.[11][12][2]

Availability edit

Oral MPA and DMPA are widely available throughout the world.[11] Oral MPA is available both alone and in combination with the estrogens CEEs, estradiol, and estradiol valerate.[11] DMPA is registered for use as a form of birth control in more than 100 countries worldwide.[19][20][11] The combination of injected MPA and estradiol cypionate is approved for use as a form of birth control in 18 countries.[19]

United States edit

As of November 2016, MPA is available in the United States in the following formulations:[64]

  • Oral pills: Amen, Curretab, Cycrin, Provera – 2.5 mg, 5 mg, 10 mg
  • Aqueous suspension for intramuscular injection: Depo-Provera – 150 mg/mL (for contraception), 400 mg/mL (for cancer)
  • Aqueous suspension for subcutaneous injection: Depo-SubQ Provera 104 – 104 mg/0.65 mL (for contraception)

It is also available in combination with an estrogen in the following formulations:

  • Oral pills: CEEs and MPA (Prempro, Prempro (Premarin, Cycrin), Premphase (Premarin, Cycrin 14/14), Premphase 14/14, Prempro/Premphase) – 0.3 mg / 1.5 mg; 0.45 mg / 1.5 mg; 0.625 mg / 2.5 mg; 0.625 mg / 5 mg

While the following formulations have been discontinued:

  • Oral pills: ethinylestradiol and MPA (Provest) – 50 μg / 10 mg
  • Aqueous suspension for intramuscular injection: estradiol cypionate and MPA (Lunelle) – 5 mg / 25 mg (for contraception)

The state of Louisiana permits sex offenders to be given MPA.[237]

Generation edit

Progestins in birth control pills are sometimes grouped by generation.[238][239] While the 19-nortestosterone progestins are consistently grouped into generations, the pregnane progestins that are or have been used in birth control pills are typically omitted from such classifications or are grouped simply as "miscellaneous" or "pregnanes".[238][239] In any case, based on its date of introduction in such formulations of 1964, MPA could be considered a "first-generation" progestin.[240]

Controversy edit

Outside the United States edit

  • In 1994, when DMPA was approved in India, India's Economic and Political Weekly reported that "The FDA finally licensed the drug in 1990 in response to concerns about the population explosion in the third world and the reluctance of third world governments to license a drug not licensed in its originating country."[241] Some scientists and women's groups in India continue to oppose DMPA.[242] In 2016, India introduced DMPA depo-medroxyprogesterone IM preparation in the public health system.[243]
  • The Canadian Coalition on Depo-Provera, a coalition of women's health professional and advocacy groups, opposed the approval of DMPA in Canada.[244] Since the approval of DMPA in Canada in 1997, a $700 million class-action lawsuit has been filed against Pfizer by users of DMPA who developed osteoporosis. In response, Pfizer argued that it had met its obligation to disclose and discuss the risks of DMPA with the Canadian medical community.[245]
  • Clinical trials for this medication regarding women in Zimbabwe were controversial with regard to human rights abuses and Medical Experimentation in Africa.
  • A controversy erupted in Israel when the government was accused of giving DMPA to Ethiopian immigrants without their consent. Some women claimed they were told it was a vaccination. The Israeli government denied the accusations but instructed the four health maintenance organizations to stop administering DMPA injections to women "if there is the slightest doubt that they have not understood the implications of the treatment".[246]

United States edit

There was a long, controversial history regarding the approval of DMPA by the U.S. Food and Drug Administration. The original manufacturer, Upjohn, applied repeatedly for approval. FDA advisory committees unanimously recommended approval in 1973, 1975 and 1992, as did the FDA's professional medical staff, but the FDA repeatedly denied approval. Ultimately, on 29 October 1992, the FDA approved DMPA for birth control, which had by then been used by over 30 million women since 1969 and was approved and being used by nearly 9 million women in more than 90 countries, including the United Kingdom, France, Germany, Sweden, Thailand, New Zealand and Indonesia.[247] Points in the controversy included:

  • Animal testing for carcinogenicity – DMPA caused breast cancer tumors in dogs. Critics of the study claimed that dogs are more sensitive to artificial progesterone, and that the doses were too high to extrapolate to humans. The FDA pointed out that all substances carcinogenic to humans are carcinogenic to animals as well, and that if a substance is not carcinogenic it does not register as a carcinogen at high doses. Levels of DMPA which caused malignant mammary tumors in dogs were equivalent to 25 times the amount of the normal luteal phase progesterone level for dogs. This is lower than the pregnancy level of progesterone for dogs, and is species-specific.[248]
    DMPA caused endometrial cancer in monkeys – 2 of 12 monkeys tested, the first ever recorded cases of endometrial cancer in rhesus monkeys.[249] However, subsequent studies have shown that in humans, DMPA reduces the risk of endometrial cancer by approximately 80%.[51][52][53]
    Speaking in comparative terms regarding animal studies of carcinogenicity for medications, a member of the FDA's Bureau of Drugs testified at an agency DMPA hearing, "...Animal data for this drug is more worrisome than any other drug we know of that is to be given to well people."
  • Cervical cancer in Upjohn/NCI studies. Cervical cancer was found to be increased as high as 9-fold in the first human studies recorded by the manufacturer and the National Cancer Institute.[250] However, numerous larger subsequent studies have shown that DMPA use does not increase the risk of cervical cancer.[251][252][253][254][255]
  • Coercion and lack of informed consent. Testing or use of DMPA was focused almost exclusively on women in developing countries and poor women in the United States,[256] raising serious questions about coercion and lack of informed consent, particularly for the illiterate[257] and for mentally disabled people, who in some reported cases were given DMPA long-term for reasons of "menstrual hygiene", although they were not sexually active.[258]
  • Atlanta/Grady Study – Upjohn studied the effect of DMPA for 11 years in Atlanta, mostly on black women who were receiving public assistance, but did not file any of the required follow-up reports with the FDA. Investigators who eventually visited noted that the studies were disorganized. "They found that data collection was questionable, consent forms and protocol were absent; that those women whose consent had been obtained at all were not told of possible side effects. Women whose known medical conditions indicated that use of DMPA would endanger their health were given the shot. Several of the women in the study died; some of cancer, but some for other reasons, such as suicide due to depression. Over half the 13,000 women in the study were lost to followup due to sloppy record keeping." Consequently, no data from this study was usable.[256]
  • WHO Review – In 1992, the WHO presented a review of DMPA in four developing countries to the FDA. The National Women's Health Network and other women's organizations testified at the hearing that the WHO was not objective, as the WHO had already distributed DMPA in developing countries. DMPA was approved for use in United States on the basis of the WHO review of previously submitted evidence from countries such as Thailand, evidence which the FDA had deemed insufficient and too poorly designed for assessment of cancer risk at a prior hearing.
  • The Alan Guttmacher Institute has speculated that United States approval of DMPA may increase its availability and acceptability in developing countries.[256][259]
  • In 1995, several women's health groups asked the FDA to put a moratorium on DMPA, and to institute standardized informed consent forms.[260]

Research edit

DMPA was studied by Upjohn for use as a progestogen-only injectable contraceptive in women at a dose of 50 mg once a month but produced poor cycle control and was not marketed for this use at this dosage.[261] A combination of DMPA and polyestradiol phosphate, an estrogen and long-lasting prodrug of estradiol, was studied in women as a combined injectable contraceptive for use by intramuscular injection once every three months.[262][263][264]

High-dose oral and intramuscular MPA monotherapy has been studied in the treatment of prostate cancer but was found to be inferior to monotherapy with cyproterone acetate or diethylstilbestrol.[265][266][267] High-dose oral MPA has been studied in combination with diethylstilbestrol and CEEs as an addition to high-dose estrogen therapy for the treatment of prostate cancer in men, but was not found to provide better effectiveness than diethylstilbestrol alone.[268]

DMPA has been studied for use as a potential male hormonal contraceptive in combination with the androgens/anabolic steroids testosterone and nandrolone (19-nortestosterone) in men.[269] However, it was never approved for this indication.[269]

MPA was investigated by InKine Pharmaceutical, Salix Pharmaceuticals, and the University of Pennsylvania as a potential anti-inflammatory medication for the treatment of autoimmune hemolytic anemia, Crohn's disease, idiopathic thrombocytopenic purpura, and ulcerative colitis, but did not complete clinical development and was never approved for these indications.[270][271] It was formulated as an oral medication at very high dosages, and was thought to inhibit the signaling of proinflammatory cytokines such as interleukin 6 and tumor necrosis factor alpha, with a mechanism of action that was said to be similar to that of corticosteroids.[270][271] The formulation of MPA had the tentative brand names Colirest and Hematrol for these indications.[270]

MPA has been found to be effective in the treatment of manic symptoms in women with bipolar disorder.[272]

Veterinary use edit

MPA has been used to reduce aggression and spraying in male cats.[273] It may be particularly useful for controlling such behaviors in neutered male cats.[273] The medication can be administered in cats as an injection once per month.[273]

See also edit

References edit

  1. ^ "Medroxyprogesterone Uses, Dosage & Side Effects".
  2. ^ a b c d e f g h i j k l m n o Index Nominum 2000: International Drug Directory. Taylor & Francis. January 2000. pp. 638–. ISBN 978-3-88763-075-1. from the original on 19 June 2013.
  3. ^ "FDA-sourced list of all drugs with black box warnings (Use Download Full Results and View Query links.)". nctr-crs.fda.gov. FDA. Retrieved 22 October 2023.
  4. ^ a b c d e f g h i j k l m n o p q r s t u v w x y z Kuhl H (2005). "Pharmacology of estrogens and progestogens: influence of different routes of administration" (PDF). Climacteric. 8 (Suppl 1): 3–63. doi:10.1080/13697130500148875. PMID 16112947. S2CID 24616324.
  5. ^ a b c d Schindler AE, Campagnoli C, Druckmann R, Huber J, Pasqualini JR, Schweppe KW, Thijssen JH (2008). "Classification and pharmacology of progestins". Maturitas. 61 (1–2): 171–80. doi:10.1016/j.maturitas.2008.11.013. PMID 19434889.
  6. ^ a b c d e f (PDF). FDA. 2015. Archived from the original (PDF) on 11 February 2017. Retrieved 31 March 2018.
  7. ^ a b c "Depo_Provera" (PDF). FDA. 2016. Retrieved 31 March 2018.
  8. ^ a b c d "depo-subQ Provera" (PDF). FDA. 2017. Retrieved 31 March 2018.
  9. ^ a b c d e f g h i j Stanczyk FZ, Bhavnani BR (September 2015). "Reprint of "Use of medroxyprogesterone acetate for hormone therapy in postmenopausal women: Is it safe?"". J. Steroid Biochem. Mol. Biol. 153: 151–9. doi:10.1016/j.jsbmb.2015.08.013. PMID 26291834. S2CID 23985966.
  10. ^ a b c d e f g h i "Medroxyprogesterone Acetate". The American Society of Health-System Pharmacists. from the original on 24 December 2016. Retrieved 8 December 2016.
  11. ^ a b c d e f g h i j k l m n o "Medroxyprogesterone Uses, Dosage & Side Effects". Drugs.com.
  12. ^ a b c d e f g h i j k Sweetman, Sean C., ed. (2009). "Sex hormones and their modulators". Martindale: The Complete Drug Reference (36th ed.). London: Pharmaceutical Press. pp. 2113–2114. ISBN 978-0-85369-840-1.
  13. ^ a b Genazzani AR (15 January 1993). Frontiers in Gynecologic and Obstetric Investigation. Taylor & Francis. p. 320. ISBN 978-1-85070-486-7. from the original on 20 May 2016.
  14. ^ a b Roberts SM (7 May 2013). Introduction to Biological and Small Molecule Drug Research and Development: Chapter 12. Hormone replacement therapy. Elsevier Science. pp. 9–. ISBN 978-0-12-806202-9. [...] medroxyprogesterone acetate, also known as Provera (discovered simultaneously by Searle and Upjohn in 1956) [..]
  15. ^ a b Sneader W (2005). "Chapter 18: Hormone analogs". Drug discovery: a history. New York: Wiley. p. 204. ISBN 0-471-89980-1.
  16. ^ World Health Organization (2019). World Health Organization model list of essential medicines: 21st list 2019. Geneva: World Health Organization. hdl:10665/325771. WHO/MVP/EMP/IAU/2019.06. License: CC BY-NC-SA 3.0 IGO.
  17. ^ Meikle AW (1 June 1999). Hormone Replacement Therapy. Springer Science & Business Media. pp. 383–. ISBN 978-1-59259-700-0.
  18. ^ Special Programme of Research, Development, and Research Training in Human Reproduction (World Health Organization); World Health Organization (2002). Research on Reproductive Health at WHO: Biennial Report 2000-2001. World Health Organization. pp. 17–. ISBN 978-92-4-156208-9.{{cite book}}: CS1 maint: multiple names: authors list (link)
  19. ^ a b c d Bagade O, Pawar V, Patel R, Patel B, Awasarkar V, Diwate S (2014). (PDF). World J Pharm Pharm Sci. 3 (10): 364–392. ISSN 2278-4357. Archived from the original (PDF) on 10 August 2017. Retrieved 2 August 2018.
  20. ^ a b Gunasheela S (14 March 2011). Practical Management of Gynecological Problems. JP Medical Ltd. pp. 39–. ISBN 978-93-5025-240-6.
  21. ^ "The Top 300 of 2020". ClinCalc. Retrieved 7 October 2022.
  22. ^ . ClinCalc. Archived from the original on 2 July 2020. Retrieved 7 October 2022.
  23. ^ Furness S, Roberts H, Marjoribanks J, Lethaby A (August 2012). "Hormone therapy in postmenopausal women and risk of endometrial hyperplasia". The Cochrane Database of Systematic Reviews. 2012 (8): CD000402. doi:10.1002/14651858.CD000402.pub4. PMC 7039145. PMID 22895916.
  24. ^ a b "Medroxyprogesterone". MedlinePlus. 9 January 2008. from the original on 12 July 2010. Retrieved 2 July 2010.
  25. ^ a b Panay N, Fenton A (February 2010). "Bioidentical hormones: what is all the hype about?". Climacteric. 13 (1): 1–3. doi:10.3109/13697130903550250. PMID 20067429. S2CID 244295.
  26. ^ Light SA, Holroyd S (March 2006). "The use of medroxyprogesterone acetate for the treatment of sexually inappropriate behaviour in patients with dementia" (PDF). Journal of Psychiatry & Neuroscience. 31 (2): 132–4. PMC 1413960. PMID 16575429. (PDF) from the original on 7 March 2016.
  27. ^ . Archived from the original on 7 January 2009. Retrieved 22 January 2009.
  28. ^ a b Depo-Provera (medroxyprogesterone acetate) (NDA # 012541) - Drugs@FDA: FDA Approved Drug Products, retrieved 2 April 2018, Original Approvals or Tentative Approvals: 09/23/1960.
  29. ^ a b c d Meyler L (2009). Meyler's side effects of endocrine and metabolic drugs. Amsterdam: Elsevier Science. pp. 281–284]. ISBN 978-0-444-53271-8. from the original on 23 October 2014.
  30. ^ Ganzina F (October 1979). "High-dose medroxyprogesterone acetate (MPA) treatment in advanced breast cancer. A review". Tumori. 65 (5): 563–85. doi:10.1177/030089167906500507. PMID 390798. S2CID 23378403.
  31. ^ Kjaer M (September 1988). "The role of medroxyprogesterone acetate (MPA) in the treatment of renal adenocarcinoma". Cancer Treat. Rev. 15 (3): 195–209. doi:10.1016/0305-7372(88)90003-5. PMID 2974757.
  32. ^ Vanderstappen D, Bonte J (1992). "New trends in the use of medroxyprogesterone acetate as a chemotherapeutic agent in gynecologic malignancies". Eur. J. Gynaecol. Oncol. 13 (2): 113–23. PMID 1534051.
  33. ^ Gooren L (2005). "Hormone treatment of the adult transsexual patient". Horm. Res. 64 (Suppl 2): 31–6. doi:10.1159/000087751. PMID 16286768. S2CID 42507159.
  34. ^ Sachdeva A, Dutta AK (31 August 2012). Advances in Pediatrics. JP Medical Ltd. pp. 1202–. ISBN 978-93-5025-777-7.
  35. ^ Hammerstein J (1990). "Antiandrogens: Clinical Aspects". Hair and Hair Diseases. pp. 827–886. doi:10.1007/978-3-642-74612-3_35. ISBN 978-3-642-74614-7.
  36. ^ a b Nadakavukaren A (28 February 2011). Our Global Environment: A Health Perspective, Seventh Edition. Waveland Press. pp. 63–. ISBN 978-1-4786-0976-6.
  37. ^ a b Trussell J (2011). "Contraceptive efficacy". In Hatcher RA, Trussell J, Nelson AL, Cates W, Kowal D, Policar MS (eds.). Contraceptive technology (20th revised ed.). New York: Ardent Media. pp. 779–863. ISBN 978-1-59708-004-0. OCLC 781956734. Table 26–1 = Table 3–2 Percentage of women experiencing an unintended pregnancy during the first year of typical use and the first year of perfect use of contraception, and the percentage continuing use at the end of the first year. United States. 15 February 2017 at the Wayback Machine
  38. ^ a b c Bakry S, Merhi ZO, Scalise TJ, Mahmoud MS, Fadiel A, Naftolin F (July 2008). "Depot-medroxyprogesterone acetate: an update". Arch. Gynecol. Obstet. 278 (1): 1–12. doi:10.1007/s00404-007-0497-z. PMID 18470526. S2CID 11340062.
  39. ^ a b c Westhoff C (August 2003). "Depot-medroxyprogesterone acetate injection (Depo-Provera): a highly effective contraceptive option with proven long-term safety". Contraception. 68 (2): 75–87. doi:10.1016/S0010-7824(03)00136-7. PMID 12954518.
  40. ^ a b c d e Trussell J (2004). "Contraceptive Efficacy". In Hatcher RA, Trussell J, Stewart FH, Nelson AL, Cates Jr W, Guest F, Kowal D (eds.). Contraceptive Technology (18th rev. ed.). New York: Ardent Media. pp. 773–845. ISBN 0-9664902-5-8.
  41. ^ a b c Trussell J (August 2004). "Contraceptive failure in the United States". Contraception. 70 (2): 89–96. doi:10.1016/j.contraception.2004.03.009. PMC 3638209. PMID 15288211.
  42. ^ Trussell J, Hatcher RA, Cates W, Stewart FH, Kost K (September 1990). "A guide to interpreting contraceptive efficacy studies". Obstetrics and Gynecology. 76 (3 Pt 2): 558–67. PMID 2199875.
  43. ^ Trussell J (1994). "Contraceptive Failure Rates". In Hatcher RA, Trussell J, Nelson AL, Cates W, Kowal D, Policar MS (eds.). Contraceptive Technology (16th rev. ed.). New York: Irvington Publishers. pp. 637–688. ISBN 0-8290-3171-5.
  44. ^ Trussell J (1998). "Contraceptive Efficacy". In Hatcher RA, Trussell J, Stewart F, Cates W, Stewart GK, Guest F, Kowal D (eds.). Contraceptive Technology (17th rev. ed.). New York: Ardent Media. pp. 779–844. ISBN 0-9664902-0-7.
  45. ^ FDA (1998). (PDF). Food and Drug Administration. Archived from the original (PDF) on 25 February 2007. Retrieved 21 June 2007.
  46. ^ Trussell J (2007). . In Hatcher RA, Trussell J, Nelson AL, Cates W, Stewart FH, Kowal D (eds.). Contraceptive Technology (19th rev. ed.). New York: Ardent Media. Archived from the original on 31 May 2008. Retrieved 21 June 2007.
  47. ^ Trussell J, Vaughan B (1999). "Contraceptive failure, method-related discontinuation and resumption of use: results from the 1995 National Survey of Family Growth" (PDF). Family Planning Perspectives. 31 (2): 64–72, 93. doi:10.2307/2991641. JSTOR 2991641. PMID 10224544. (PDF) from the original on 2 December 2008.
  48. ^ a b c Hatcher RA (2004). "Depo-Provera Injections, Implants, and Progestin-Only Pills (Minipills)". In Hatcher RA, Trussell J, Stewart F, Cates W, Stewart GK, Guest F, Kowal D (eds.). Contraceptive Technology (18th rev. ed.). New York: Ardent Media. pp. 461–494. ISBN 0-9664902-5-8.
  49. ^ a b c Speroff L, Darney PD (2005). "Injectable Contraception". A Clinical Guide for Contraception (4th ed.). Philadelphia: Lippincott Williams & Wilkins. pp. 201–220. ISBN 0-7817-6488-2.
  50. ^ Mishell DR (2004). "Contraception". In Strauss III JF, Barbieri RL (eds.). Yen and Jaffe's Reproductive Endocrinology (5th ed.). Philadelphia: Elsevier Saunders. pp. 899–938. ISBN 0-7216-9546-9.
  51. ^ a b Kaunitz AM (December 2001). "Current options for injectable contraception in the United States". Seminars in Reproductive Medicine. 19 (4): 331–7. doi:10.1055/s-2001-18641. PMID 11727175. S2CID 39556669.
  52. ^ a b Bigrigg A, Evans M, Gbolade B, Newton J, Pollard L, Szarewski A, Thomas C, Walling M (July 1999). "Depo Provera. Position paper on clinical use, effectiveness and side effects". The British Journal of Family Planning. 25 (2): 69–76. PMID 10454658.
  53. ^ a b "Depot-medroxyprogesterone acetate (DMPA) and risk of endometrial cancer. The WHO Collaborative Study of Neoplasia and Steroid Contraceptives". International Journal of Cancer. 49 (2): 186–90. September 1991. doi:10.1002/ijc.2910490207. PMID 1831802. S2CID 221776781.
  54. ^ Santen RJ (2004). "Endocrinology of Breast and Endometrial Cancer". In Strauss III JF, Barbieri RL (eds.). Yen and Jaffe's Reproductive Endocrinology (5th ed.). Philadelphia: Elsevier Saunders. pp. 787–809. ISBN 0-7216-9546-9.
  55. ^ Bartz D, Goldberg AB (2011). "Injectable contraceptives". In Hatcher RA, Trussell J, Nelson AL, Cates W Jr, Kowal D, Policar MS (eds.). Contraceptive technology (20th revised ed.). New York: Ardent Media. pp. 212–213. ISBN 978-1-59708-004-0. OCLC 781956734. Advantages of DMPA Injectables. 5. Reduced risk of ectopic pregnancy. Compared with women who use no contraceptive at all, women who use DMPA have a reduced risk for having an ectopic pregnancy. Although the overall risk of pregnancy and thus ectopic pregnancy is lowered by DMPA, the possibility of an ectopic pregnancy should be excluded if a woman using DMPA becomes pregnant. One study showed that 1.5% of women who got pregnant on DMPA had an ectopic pregnancy, the same ectopic rate as women who conceived while not using contraception.27
  56. ^ Borgatta L, Murthy A, Chuang C, Beardsley L, Burnhill MS (September 2002). "Pregnancies diagnosed during Depo-Provera use". Contraception. 66 (3): 169–72. doi:10.1016/S0010-7824(02)00340-2. PMID 12384205.
  57. ^ J. Julie Kim, Takeshi Kurita, Serdar E. Bulun, Progesterone Action in Endometrial Cancer, Endometriosis, Uterine Fibroids, and Breast Cancer, Endocrine Reviews, Volume 34, Issue 1, 1 February 2013, Pages 130–162, https://doi.org/10.1210/er.2012-1043 <ref>
  58. ^ "Increasing use of long-acting reversible contraception". Nursing Times.net. 21 October 2008. from the original on 26 August 2009. Retrieved 19 June 2009.
  59. ^ (PDF). National Institute for Health and Clinical Excellence. Archived from the original (PDF) on 20 September 2009. Retrieved 19 June 2009.
  60. ^ "Sexual Health Ruleset" (PDF). New GMS Contract Quality and Outcome Framework - Implementation Dataset and Business Rules. Primary Care Commissioning. 1 May 2009. (PDF) from the original on 10 August 2011. Retrieved 19 June 2009.
    Sumarized at
    * "Contraception - Management QOF indicators". NHS Clinical Knowledge Summaries. NHS Institute for Innovation and Improvement. Retrieved 19 June 2009.[permanent dead link]
  61. ^ a b c Holtorf K (January 2009). (PDF). Postgraduate Medicine. 121 (1): 73–85. doi:10.3810/pgm.2009.01.1949. PMID 19179815. S2CID 2060730. Archived from the original (PDF) on 8 July 2011.
  62. ^ Cirigliano M (June 2007). (PDF). Journal of Women's Health. 16 (5): 600–31. doi:10.1089/jwh.2006.0311. PMID 17627398. Archived from the original (PDF) on 6 January 2011.
  63. ^ Boothby LA, Doering PL (August 2008). "Bioidentical hormone therapy: a panacea that lacks supportive evidence". Current Opinion in Obstetrics and Gynecology. 20 (4): 400–7. doi:10.1097/GCO.0b013e3283081ae9. PMID 18660693. S2CID 22449765.
  64. ^ a b c d "Drugs@FDA: FDA Approved Drug Products". United States Food and Drug Administration. from the original on 16 November 2016. Retrieved 31 March 2018.
  65. ^ Engorn B, Flerlage J (1 May 2014). The Harriet Lane Handbook E-Book. Elsevier Health Sciences. pp. 846–. ISBN 978-0-323-11246-8.
  66. ^ Tiziani AP (1 June 2013). Havard's Nursing Guide to Drugs. Elsevier Health Sciences. pp. 989–. ISBN 978-0-7295-8162-2.
  67. ^ Leidenberger FA (17 April 2013). Klinische Endokrinologie für Frauenärzte. Springer-Verlag. pp. 528–. ISBN 978-3-662-08110-5.
  68. ^ Stacey, Dawn. Depo Provera: The Birth Control Shot 10 October 2008 at the Wayback Machine Accessed 13 October 2009.
  69. ^ WHO (2004). "Progestogen-only contraceptives". Medical Eligibility Criteria for Contraceptive Use (3rd ed.). Geneva: Reproductive Health and Research, WHO. ISBN 92-4-156266-8. from the original on 31 May 2009.
  70. ^ FFPRHC (2006). (PDF). Archived from the original (PDF) on 19 June 2007. Retrieved 11 January 2007.
  71. ^ a b c d "MedroxyPROGESTERone: Drug Information Provided by Lexi-Comp". Merck Manual. 1 December 2009. from the original on 24 July 2010. Retrieved 8 July 2010.
  72. ^ King SR (9 November 2012). Neurosteroids and the Nervous System. Springer Science & Business Media. pp. 45–. ISBN 978-1-4614-5559-2. from the original on 5 November 2017.
  73. ^ a b Pfizer (October 2004). (PDF). Archived from the original (PDF) on 6 February 2007. Retrieved 21 February 2007.
  74. ^ Amatayakul K, Sivasomboon B, Thanangkul O (September 1978). "Vitamin and trace mineral metabolism in medroxyprogesterone acetate users". Contraception. 18 (3): 253–69. doi:10.1016/s0010-7824(78)80019-5. PMID 720068.
  75. ^ a b Nelson AL (2014). "DMPA: battered and bruised but still needed and used in the USA". Expert Review of Obstetrics & Gynecology. 5 (6): 673–686. doi:10.1586/eog.10.60. ISSN 1747-4108.
  76. ^ a b Aronson JK (15 October 2015). Meyler's Side Effects of Drugs: The International Encyclopedia of Adverse Drug Reactions and Interactions. Elsevier Science. pp. 2–. ISBN 978-0-444-53716-4. Perhaps surprisingly, a consensus seems to be emerging that depot medroxyprogesterone acetate implants do not in fact result in an increase in the incidence of depression or in the severity of pre-existing depression, even after 1 or 2 years, nor do they cause significant weight gain.
  77. ^ "Exposure to DMPA in pregnancy may cause low birth weight". Progress in Human Reproduction Research (23): 2–3. 1992. PMID 12286194.
  78. ^ a b c Lobo RA (5 June 2007). Treatment of the Postmenopausal Woman: Basic and Clinical Aspects. Elsevier. pp. 211–. ISBN 978-0-08-055309-2.
  79. ^ Kaunitz AM (1999). "Long-acting hormonal contraception: assessing impact on bone density, weight, and mood". Int J Fertil Womens Med. 44 (2): 110–7. PMID 10338269. Despite the efficacy and increasing acceptability of these long-term methods, some clinicians and women are reluctant to use them because of concerns regarding reduction in bone density with DMPA, and depressive symptoms and body weight issues with both injectables and implants. Recent multicenter experience showed no increase in depressive symptoms after 1 year's DMPA use and 2 years' Norplant use, even among users with the highest mean depressive symptom scores pre-therapy.
  80. ^ a b c Worly BL, Gur TL, Schaffir J (February 2018). "The relationship between progestin hormonal contraception and depression: a systematic review". Contraception. 97 (6): 478–489. doi:10.1016/j.contraception.2018.01.010. PMID 29496297. S2CID 3644828.
  81. ^ Westhoff C (August 2003). "Depot-medroxyprogesterone acetate injection (Depo-Provera): a highly effective contraceptive option with proven long-term safety". Contraception. 68 (2): 75–87. doi:10.1016/s0010-7824(03)00136-7. PMID 12954518. Another common patient tolerability concern reported with hormonal contraception is the effect on mood [95]. The majority of published reports indicate that DMPA does not cause depressive symptoms. In a large, 1-year, clinical trial of DMPA in 3857 US women, fewer than 2% of users reported depression [15]. Other reports in various settings, including a private practice [96], adolescent clinics [97,98], a psychiatric hospital [99] and inner-city family-planning clinics [100,101], have not found an adverse effect of DMPA on depression. [...] Using a variety of objective indices for depressive symptoms, the overall data for both OCs and DMPA are supportive that these agents have no significant effect on mood. Although history of mood symptoms prior to OC use may predispose a subgroup of women to negative mood changes, the data for DMPA suggest that even women who have depressive symptoms prior to treatment can tolerate therapy with no exacerbation of these symptoms.
  82. ^ a b c d Westhoff C, Truman C, Kalmuss D, Cushman L, Davidson A, Rulin M, Heartwell S (April 1998). "Depressive symptoms and Depo-Provera". Contraception. 57 (4): 237–40. doi:10.1016/s0010-7824(98)00024-9. PMID 9649914.
  83. ^ Fink G, Pfaff DW, Levine J (31 August 2011). Handbook of Neuroendocrinology. Academic Press. pp. 564–. ISBN 978-0-12-378554-1.
  84. ^ Ryden J (2009). Practical Gynecology: A Guide for the Primary Care Physician. ACP Press. pp. 379–. ISBN 978-1-934465-05-9.
  85. ^ Hlatky MA, Boothroyd D, Vittinghoff E, Sharp P, Whooley MA (February 2002). "Quality-of-life and depressive symptoms in postmenopausal women after receiving hormone therapy: results from the Heart and Estrogen/Progestin Replacement Study (HERS) trial". JAMA. 287 (5): 591–7. doi:10.1001/jama.287.5.591. PMID 11829697.
  86. ^ Wiegratz I, Kuhl H (August 2004). "Progestogen therapies: differences in clinical effects?". Trends Endocrinol. Metab. 15 (6): 277–85. doi:10.1016/j.tem.2004.06.006. PMID 15358281. S2CID 35891204.
  87. ^ Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, Kotchen JM, Ockene J (July 2002). "Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial" (PDF). JAMA. 288 (3): 321–33. doi:10.1001/jama.288.3.321. PMID 12117397. S2CID 20149703.
  88. ^ Prentice RL, Anderson GL (2008). "The women's health initiative: lessons learned". Annual Review of Public Health. 29: 131–50. doi:10.1146/annurev.publhealth.29.020907.090947. PMID 18348708.
  89. ^ Buist DS, Newton KM, Miglioretti DL, Beverly K, Connelly MT, Andrade S, Hartsfield CL, Wei F, Chan KA, Kessler L (November 2004). "Hormone therapy prescribing patterns in the United States". Obstetrics and Gynecology. 104 (5 Pt 1): 1042–50. doi:10.1097/01.AOG.0000143826.38439.af. PMID 15516400. S2CID 31593637.
  90. ^ a b Beyer-Westendorf J, Bauersachs R, Hach-Wunderle V, Zotz RB, Rott H (October 2018). "Sex hormones and venous thromboembolism - from contraception to hormone replacement therapy". VASA. Zeitschrift für Gefässkrankheiten. 47 (6): 441–450. doi:10.1024/0301-1526/a000726. PMID 30008249. S2CID 51628832.
  91. ^ a b c d DeLoughery TG (June 2011). "Estrogen and thrombosis: controversies and common sense". Reviews in Endocrine & Metabolic Disorders. 12 (2): 77–84. doi:10.1007/s11154-011-9178-0. PMID 21559819. S2CID 28053690.
  92. ^ a b c Mantha S, Karp R, Raghavan V, Terrin N, Bauer KA, Zwicker JI (August 2012). "Assessing the risk of venous thromboembolic events in women taking progestin-only contraception: a meta-analysis". BMJ. 345 (aug07 2): e4944. doi:10.1136/bmj.e4944. PMC 3413580. PMID 22872710.
  93. ^ a b Tepper NK, Jeng G, Curtis KM, Boutot ME, Boulet SL, Whiteman MK (March 2019). "Venous Thromboembolism Among Women Initiating Depot Medroxyprogesterone Acetate Immediately Postpartum". Obstetrics and Gynecology. 133 (3): 533–540. doi:10.1097/AOG.0000000000003135. PMID 30741807.
  94. ^ van Hylckama Vlieg A, Middeldorp S (February 2011). "Hormone therapies and venous thromboembolism: where are we now?". Journal of Thrombosis and Haemostasis. 9 (2): 257–266. doi:10.1111/j.1538-7836.2010.04148.x. PMID 21114755. S2CID 32394836.
  95. ^ Benagiano G, Primiero FM (June 1983). "Long acting contraceptives. Present status". Drugs. 25 (6): 570–609. doi:10.2165/00003495-198325060-00003. PMID 6223801. S2CID 45898359.
  96. ^ Rott H (February 2019). "Birth Control Pills and Thrombotic Risks: Differences of Contraception Methods with and without Estrogen". Hamostaseologie. 39 (1): 42–48. doi:10.1055/s-0039-1677806. PMID 30669160. S2CID 58947063.
  97. ^ a b Scarabin PY (August 2018). "Progestogens and venous thromboembolism in menopausal women: an updated oral versus transdermal estrogen meta-analysis". Climacteric. 21 (4): 341–345. doi:10.1080/13697137.2018.1446931. PMID 29570359. S2CID 4229701.
  98. ^ a b Sitruk-Ware R, Nath A (June 2011). "Metabolic effects of contraceptive steroids". Reviews in Endocrine & Metabolic Disorders. 12 (2): 63–75. doi:10.1007/s11154-011-9182-4. PMID 21538049. S2CID 23760705.
  99. ^ a b Kuhl H (2011). "Pharmacology of Progestogens" (PDF). J Reproduktionsmed Endokrinol. 8 (1): 157–177. (PDF) from the original on 11 October 2016.
  100. ^ "Depot medroxyprogesterone acetate and bone effects. Committee Opinion #602". June 2014. from the original on 30 April 2015. Retrieved 3 May 2015.
  101. ^ a b FDA (17 November 2004). . Food and Drug Administration. Archived from the original on 21 December 2005. Retrieved 12 May 2006.
  102. ^ World Health Organization (September 2005). . Family Planning. Archived from the original on 14 May 2006. Retrieved 12 May 2006.
  103. ^ Curtis KM, Martins SL (May 2006). "Progestogen-only contraception and bone mineral density: a systematic review". Contraception. 73 (5): 470–87. doi:10.1016/j.contraception.2005.12.010. PMID 16627031.
  104. ^ Cundy T, Cornish J, Evans MC, Roberts H, Reid IR (January 1994). "Recovery of bone density in women who stop using medroxyprogesterone acetate". BMJ. 308 (6923): 247–8. doi:10.1136/bmj.308.6923.247. PMC 2539337. PMID 8111260.
  105. ^ Scholes D, LaCroix AZ, Ichikawa LE, Barlow WE, Ott SM (September 2002). "Injectable hormone contraception and bone density: results from a prospective study". Epidemiology. 13 (5): 581–7. doi:10.1097/00001648-200209000-00015. PMID 12192229.
  106. ^ Scholes D, LaCroix AZ, Ichikawa LE, Barlow WE, Ott SM (February 2005). "Change in bone mineral density among adolescent women using and discontinuing depot medroxyprogesterone acetate contraception". Archives of Pediatrics & Adolescent Medicine. 159 (2): 139–44. doi:10.1001/archpedi.159.2.139. PMID 15699307.
  107. ^ Orr-Walker BJ, Evans MC, Ames RW, Clearwater JM, Cundy T, Reid IR (November 1998). "The effect of past use of the injectable contraceptive depot medroxyprogesterone acetate on bone mineral density in normal post-menopausal women". Clinical Endocrinology. 49 (5): 615–8. doi:10.1046/j.1365-2265.1998.00582.x. PMID 10197077. S2CID 22565912.
  108. ^ Cundy T, Cornish J, Roberts H, Reid IR (May 2002). "Menopausal bone loss in long-term users of depot medroxyprogesterone acetate contraception". American Journal of Obstetrics and Gynecology. 186 (5): 978–83. doi:10.1067/mob.2002.122420. PMID 12015524.
  109. ^ Walsh JS, Eastell R, Peel NF (February 2010). "Depot medroxyprogesterone acetate use after peak bone mass is associated with increased bone turnover but no decrease in bone mineral density". Fertility and Sterility. 93 (3): 697–701. doi:10.1016/j.fertnstert.2008.10.004. PMID 19013564.
  110. ^ American College of Obstetricians Gynecologists Committee on Gynecologic Practice (September 2008). "ACOG Committee Opinion No. 415: Depot medroxyprogesterone acetate and bone effects". Obstetrics and Gynecology. 112 (3): 727–30. doi:10.1097/AOG.0b013e318188d1ec. PMID 18757687.
  111. ^ Polis CB, Phillips SJ, Curtis KM, Westreich DJ, Steyn PS, Raymond E, Hannaford P, Turner AN (October 2014). "Hormonal contraceptive methods and risk of HIV acquisition in women: a systematic review of epidemiological evidence". Contraception. 90 (4): 360–90. doi:10.1016/j.contraception.2014.07.009. PMID 25183264.
  112. ^ WHO Department of Reproductive Health and Research (16 February 2012). . Geneva: World Health Organization. Archived from the original on 30 January 2015.
  113. ^ WHO Department of Reproductive Health and Research (23 July 2014). (PDF). Geneva: World Health Organization. Archived from the original (PDF) on 30 January 2015.
  114. ^ AVAC (27 January 2015). "News from the HC-HIV front: it's raining meta (analyses)!". New York: AIDS Vaccine Advocacy Coalition. from the original on 30 January 2015.
  115. ^ Ralph LJ, McCoy SI, Shiu K, Padian NS (February 2015). "Hormonal contraceptive use and women's risk of HIV acquisition: a meta-analysis of observational studies". The Lancet. Infectious Diseases. 15 (2): 181–9. doi:10.1016/S1473-3099(14)71052-7. PMC 4526270. PMID 25578825.
  116. ^ Morrison CS, Chen PL, Kwok C, Baeten JM, Brown J, Crook AM, Van Damme L, Delany-Moretlwe S, Francis SC, Friedland BA, Hayes RJ, Heffron R, Kapiga S, Karim QA, Karpoff S, Kaul R, McClelland RS, McCormack S, McGrath N, Myer L, Rees H, van der Straten A, Watson-Jones D, van de Wijgert JH, Stalter R, Low N (January 2015). "Hormonal contraception and the risk of HIV acquisition: an individual participant data meta-analysis". PLOS Medicine. 12 (1): e1001778. doi:10.1371/journal.pmed.1001778. PMC 4303292. PMID 25612136.
  117. ^ Faculty of Sexual Reproductive Healthcare (January 2015). (PDF). London: Royal College of Obstetricians and Gynaecologists. Archived from the original (PDF) on 30 January 2015.
  118. ^ Brind J, Condly SJ, Mosher SW, Morse AR, Kimball J (2015). "Risk of HIV Infection in Depot-Medroxyprogesterone Acetate (DMPA) Users: A Systematic Review and Meta-analysis". Issues Law Med. 30 (2): 129–39. PMID 26710371.
  119. ^ ECHO Trial Consortium (13 June 2019). "HIV incidence among women using intramuscular depot medroxyprogesterone acetate, a copper intrauterine device, or a levonorgestrel implant for contraception: a randomised, multicentre, open-label trial". The Lancet. 394 (10195): 303–313. doi:10.1016/S0140-6736(19)31288-7. PMC 6675739. PMID 31204114.
  120. ^ Dahlberg K (February 1982). "Some effects of depo-medroxyprogesterone acetate (DMPA): observations in the nursing infant and in the long-term user". International Journal of Gynaecology and Obstetrics. 20 (1): 43–8. doi:10.1016/0020-7292(82)90044-3. PMID 6126406. S2CID 8295162.
  121. ^ Pardthaisong T, Yenchit C, Gray R (April 1992). "The long-term growth and development of children exposed to Depo-Provera during pregnancy or lactation". Contraception. 45 (4): 313–24. doi:10.1016/0010-7824(92)90053-V. PMID 1387602.
  122. ^ a b Muss HB, Cruz JM (August 1992). "High-dose progestin therapy for metastatic breast cancer". Annals of Oncology. 3 (Suppl 3): 15–20. doi:10.1093/annonc/3.suppl_3.S15. PMID 1390312.
  123. ^ Pannuti F, Martoni A, Lenaz GR, Piana E, Nanni P (April 1978). "A possible new approach to the treatment of metastatic breast cancer: massive doses of medroxyprogesterone acetate". Cancer Treat Rep. 62 (4): 499–504. PMID 350387.
  124. ^ Simons JP, Schols AM, Hoefnagels JM, Westerterp KR, ten Velde GP, Wouters EF (February 1998). "Effects of medroxyprogesterone acetate on food intake, body composition, and resting energy expenditure in patients with advanced, nonhormone-sensitive cancer: a randomized, placebo-controlled trial". Cancer. 82 (3): 553–60. doi:10.1002/(sici)1097-0142(19980201)82:3<553::aid-cncr18>3.0.co;2-0. PMID 9452274.
  125. ^ World Health Organization (2004). Residues of Some Veterinary Drugs in Animals and Food: Monographs Prepared by the Sixty-second Meeting of the Joint FAO/WHO Expert Committee on Food Additives, Rome, 4-12 February 2004. Food & Agriculture Org. p. 49. ISBN 978-92-5-105195-5. from the original on 17 June 2014.
  126. ^ a b c d Kemppainen JA, Langley E, Wong CI, Bobseine K, Kelce WR, Wilson EM (March 1999). "Distinguishing androgen receptor agonists and antagonists: distinct mechanisms of activation by medroxyprogesterone acetate and dihydrotestosterone". Molecular Endocrinology. 13 (3): 440–54. doi:10.1210/mend.13.3.0255. PMID 10077001.
  127. ^ Bentel JM, Birrell SN, Pickering MA, Holds DJ, Horsfall DJ, Tilley WD (August 1999). "Androgen receptor agonist activity of the synthetic progestin, medroxyprogesterone acetate, in human breast cancer cells". Molecular and Cellular Endocrinology. 154 (1–2): 11–20. doi:10.1016/S0303-7207(99)00109-4. PMID 10509795. S2CID 25584803.
  128. ^ a b c Pullen MA, Laping N, Edwards R, Bray J (September 2006). "Determination of conformational changes in the progesterone receptor using ELISA-like assays". Steroids. 71 (9): 792–8. doi:10.1016/j.steroids.2006.05.009. PMID 16784762. S2CID 24703323.
  129. ^ Wikström A, Green B, Johansson ED (1984). "The plasma concentration of medroxyprogesterone acetate and ovarian function during treatment with medroxyprogesterone acetate in 5 and 10 mg doses". Acta Obstetricia et Gynecologica Scandinavica. 63 (2): 163–8. doi:10.3109/00016348409154654. PMID 6233840. S2CID 45767999.
  130. ^ a b Blum RW (22 October 2013). Adolescent Health Care: Clinical Issues. Elsevier Science. pp. 216–. ISBN 978-1-4832-7738-7.
  131. ^ a b Schindler AE, Campagnoli C, Druckmann R, Huber J, Pasqualini JR, Schweppe KW, Thijssen JH (December 2003). "Classification and pharmacology of progestins". Maturitas. 46 (Suppl 1): S7–S16. doi:10.1016/j.maturitas.2003.09.014. PMID 14670641.
  132. ^ Glasier A (2006). "Contraception". In DeGroot LJ, Jameson JL (eds.). Endocrinology (5th ed.). Philadelphia: Elsevier Saunders. pp. 2993–3003. ISBN 978-0-7216-0376-6.
  133. ^ Loose DS, Stancel GM (2006). "Estrogens and Progestins". In Loose DS, Stancel GM (eds.). Goodman & Gilman's The Pharmacological Basis of Therapeutics (11th ed.). New York: McGraw-Hill. pp. 1541–1571. ISBN 0-07-142280-3.
  134. ^ a b Rivera R, Yacobson I, Grimes D (November 1999). "The mechanism of action of hormonal contraceptives and intrauterine contraceptive devices". American Journal of Obstetrics and Gynecology. 181 (5 Pt 1): 1263–9. doi:10.1016/S0002-9378(99)70120-1. PMID 10561657.
  135. ^ Fritz MA, Speroff L (28 March 2012). Clinical Gynecologic Endocrinology and Infertility. Lippincott Williams & Wilkins. pp. 761–. ISBN 978-1-4511-4847-3.
  136. ^ Knörr K, Beller FK, Lauritzen C (17 April 2013). Lehrbuch der Gynäkologie. Springer-Verlag. pp. 214–. ISBN 978-3-662-00942-0.
  137. ^ Knörr K, Knörr-Gärtner H, Beller FK, Lauritzen C (8 March 2013). Geburtshilfe und Gynäkologie: Physiologie und Pathologie der Reproduktion. Springer-Verlag. pp. 583–. ISBN 978-3-642-95583-9.
  138. ^ Labhart A (6 December 2012). Clinical Endocrinology: Theory and Practice. Springer Science & Business Media. pp. 554–. ISBN 978-3-642-96158-8.
  139. ^ Horský J, Presl J (1981). "Hormonal Treatment of Disorders of the Menstrual Cycle". In Horsky J, Presl K (eds.). Ovarian Function and its Disorders: Diagnosis and Therapy. Springer Science & Business Media. pp. 309–332. doi:10.1007/978-94-009-8195-9_11. ISBN 978-94-009-8195-9.
  140. ^ Ufer J (1969). The Principles and Practice of Hormone Therapy in Gynaecology and Obstetrics. de Gruyter. p. 49. ISBN 9783110006148. 17α-Hydroxyprogesterone caproate is a depot progestogen which is entirely free of side actions. The dose required to induce secretory changes in primed endometrium is about 250 mg. per menstrual cycle.
  141. ^ Pschyrembel W (1968). Praktische Gynäkologie: für Studierende und Ärzte. Walter de Gruyter. pp. 598, 601. ISBN 978-3-11-150424-7.
  142. ^ Ferin J (September 1972). "Effects, Duration of Action and Metabolism in Man". In Tausk M (ed.). Pharmacology of the Endocrine System and Related Drugs: Progesterone, Progestational Drugs and Antifertility Agents. Vol. II. Pergamon Press. pp. 13–24. ISBN 978-0080168128. OCLC 278011135.
  143. ^ Henzl MR, Edwards JA (10 November 1999). "Pharmacology of Progestins: 17α-Hydroxyprogesterone Derivatives and Progestins of the First and Second Generation". In Sitruk-Ware R, Mishell DR (eds.). Progestins and Antiprogestins in Clinical Practice. Taylor & Francis. pp. 101–132. ISBN 978-0-8247-8291-7.
  144. ^ Brotherton J (1976). Sex Hormone Pharmacology. Academic Press. p. 114. ISBN 978-0-12-137250-7.
  145. ^ Sang GW (April 1994). "Pharmacodynamic effects of once-a-month combined injectable contraceptives". Contraception. 49 (4): 361–385. doi:10.1016/0010-7824(94)90033-7. PMID 8013220.
  146. ^ Toppozada MK (April 1994). "Existing once-a-month combined injectable contraceptives". Contraception. 49 (4): 293–301. doi:10.1016/0010-7824(94)90029-9. PMID 8013216.
  147. ^ Goebelsmann U (1986). "Pharmacokinetics of Contraceptive Steroids in Humans". In Gregoire AT, Blye RP (eds.). Contraceptive Steroids: Pharmacology and Safety. Springer Science & Business Media. pp. 67–111. doi:10.1007/978-1-4613-2241-2_4. ISBN 978-1-4613-2241-2.
  148. ^ Becker H, Düsterberg B, Klosterhalfen H (1980). "[Bioavailability of cyproterone acetate after oral and intramuscular application in men (author's transl)]" [Bioavailability of Cyproterone Acetate after Oral and Intramuscular Application in Men]. Urologia Internationalis. 35 (6): 381–385. doi:10.1159/000280353. PMID 6452729.
  149. ^ Moltz L, Haase F, Schwartz U, Hammerstein J (May 1983). "[Treatment of virilized women with intramuscular administration of cyproterone acetate]" [Efficacy of Intra muscularly Applied Cyproterone Acetate in Hyperandrogenism]. Geburtshilfe und Frauenheilkunde. 43 (5): 281–287. doi:10.1055/s-2008-1036893. PMID 6223851.
  150. ^ Wright JC, Burgess DJ (29 January 2012). Long Acting Injections and Implants. Springer Science & Business Media. pp. 114–. ISBN 978-1-4614-0554-2.
  151. ^ Chu YH, Li Q, Zhao ZF (April 1986). "Pharmacokinetics of megestrol acetate in women receiving IM injection of estradiol-megestrol long-acting injectable contraceptive". The Chinese Journal of Clinical Pharmacology. The results showed that after injection the concentration of plasma MA increased rapidly. The meantime of peak plasma MA level was 3rd day, there was a linear relationship between log of plasma MA concentration and time (day) after administration in all subjects, elimination phase half-life t1/2β = 14.35 ± 9.1 days.
  152. ^ Runnebaum BC, Rabe T, Kiesel L (6 December 2012). Female Contraception: Update and Trends. Springer Science & Business Media. pp. 429–. ISBN 978-3-642-73790-9.
  153. ^ Artini PG, Genazzani AR, Petraglia F (11 December 2001). Advances in Gynecological Endocrinology. CRC Press. pp. 105–. ISBN 978-1-84214-071-0.
  154. ^ King TL, Brucker MC, Kriebs JM, Fahey JO (21 October 2013). Varney's Midwifery. Jones & Bartlett Publishers. pp. 495–. ISBN 978-1-284-02542-2.
  155. ^ a b Poulin R, Baker D, Poirier D, Labrie F (March 1989). "Androgen and glucocorticoid receptor-mediated inhibition of cell proliferation by medroxyprogesterone acetate in ZR-75-1 human breast cancer cells". Breast Cancer Research and Treatment. 13 (2): 161–72. doi:10.1007/bf01806528. PMID 2525057. S2CID 26116247.
  156. ^ Brady BM, Anderson RA, Kinniburgh D, Baird DT (April 2003). "Demonstration of progesterone receptor-mediated gonadotrophin suppression in the human male". Clinical Endocrinology. 58 (4): 506–12. doi:10.1046/j.1365-2265.2003.01751.x. PMID 12641635. S2CID 12567639.
  157. ^ Saleh FM, Grudzinskas AJ, Bradford JM (11 February 2009). Sex Offenders: Identification, Risk Assessment, Treatment, and Legal Issues. Oxford University Press. p. 44. ISBN 978-0-19-517704-6. from the original on 17 June 2014.
  158. ^ World Health Organization (2009). Stuart MC, Kouimtzi M, Hill SR (eds.). WHO Model Formulary 2008. World Health Organization. p. 368. hdl:10665/44053. ISBN 9789241547659.
  159. ^ a b c Lothstein LM (1996). "Antiandrogen treatment for sexual disorders: Guidelines for establishing a standard of care". Sexual Addiction & Compulsivity. 3 (4): 313–331. doi:10.1080/10720169608400122. ISSN 1072-0162.
  160. ^ a b Dangerous Sex Offenders: A Task Force Report of the American Psychiatric Association. American Psychiatric Pub. 1999. pp. 112–144. ISBN 978-0-89042-280-9.
  161. ^ Kravitz HM, Haywood TW, Kelly J, Liles S, Cavanaugh JL (1996). "Medroxyprogesterone and paraphiles: do testosterone levels matter?". The Bulletin of the American Academy of Psychiatry and the Law. 24 (1): 73–83. PMID 8891323.
  162. ^ Novak E, Hendrix JW, Chen TT, Seckman CE, Royer GL, Pochi PE (October 1980). "Sebum production and plasma testosterone levels in man after high-dose medroxyprogesterone acetate treatment and androgen administration". Acta Endocrinologica. 95 (2): 265–270. doi:10.1530/acta.0.0950265. PMID 6449127.
  163. ^ Kirschner MA, Schneider G (February 1972). "Suppression of the pituitary-Leydig cell axis and sebum production in normal men by medroxyprogesterone acetate (provera)". Acta Endocrinologica. 69 (2): 385–393. doi:10.1530/acta.0.0690385. PMID 5066846.
  164. ^ Asscheman H, Gooren LJ (1993). "Hormone Treatment in Transsexuals". Journal of Psychology & Human Sexuality. 5 (4): 39–54. doi:10.1300/J056v05n04_03. ISSN 0890-7064. S2CID 144580633.
  165. ^ Meriggiola MC, Gava G (November 2015). "Endocrine care of transpeople part II. A review of cross-sex hormonal treatments, outcomes and adverse effects in transwomen". Clinical Endocrinology. 83 (5): 607–615. doi:10.1111/cen.12754. hdl:11585/541921. PMID 25692882. S2CID 39706760.
  166. ^ Gooren LJ, Giltay EJ, Bunck MC (January 2008). "Long-term treatment of transsexuals with cross-sex hormones: extensive personal experience". The Journal of Clinical Endocrinology and Metabolism. 93 (1): 19–25. doi:10.1210/jc.2007-1809. PMID 17986639.
  167. ^ Deutsch M (17 June 2016). "Guidelines for the Primary and Gender-Affirming Care of Transgender and Gender Nonbinary People" (PDF) (2nd ed.). University of California, San Francisco: Center of Excellence for Transgender Health. p. 28.
  168. ^ Dahl M, Feldman JL, Goldberg J, Jaberi A (2015). "Endocrine Therapy for Transgender Adults in British Columbia: Suggested Guidelines" (PDF). Vancouver Coastal Health. Retrieved 15 August 2018.
  169. ^ Leinung MC, Feustel PJ, Joseph J (2018). "Hormonal Treatment of Transgender Women with Oral Estradiol". Transgender Health. 3 (1): 74–81. doi:10.1089/trgh.2017.0035. PMC 5944393. PMID 29756046.
  170. ^ Onu PE (1995). "Depot medroxyprogesterone in the management of benign prostatic hyperplasia". European Urology. 28 (3): 229–235. doi:10.1159/000475056. PMID 8536777.
  171. ^ Meyer WJ, Walker PA, Emory LE, Smith ER (January 1985). "Physical, metabolic, and hormonal effects on men of long-term therapy with medroxyprogesterone acetate". Fertility and Sterility. 43 (1): 102–109. doi:10.1016/S0015-0282(16)48326-3. PMID 3155506.
  172. ^ Birrell SN, Hall RE, Tilley WD (January 1998). "Role of the androgen receptor in human breast cancer". Journal of Mammary Gland Biology and Neoplasia. 3 (1): 95–103. doi:10.1023/A:1018730519839. PMID 10819508. S2CID 12044431.
  173. ^ Buchanan G, Birrell SN, Peters AA, Bianco-Miotto T, Ramsay K, Cops EJ, Yang M, Harris JM, Simila HA, Moore NL, Bentel JM, Ricciardelli C, Horsfall DJ, Butler LM, Tilley WD (September 2005). "Decreased androgen receptor levels and receptor function in breast cancer contribute to the failure of response to medroxyprogesterone acetate". Cancer Research. 65 (18): 8487–96. doi:10.1158/0008-5472.CAN-04-3077. PMID 16166329.
  174. ^ Rees MC, Hope S, Ravnikar V (12 August 2005). The Abnormal Menstrual Cycle. Taylor & Francis. p. 213. ISBN 978-1-84214-212-7. from the original on 31 December 2013.
  175. ^ Aronson JK (20 January 2009). Meyler's Side Effects of Endocrine and Metabolic Drugs. Elsevier. p. 283. ISBN 978-0-444-53271-8. from the original on 31 December 2013.
  176. ^ Ettinger B, Golditch IM (December 1977). "Medroxyprogesterone acetate for the evaluation of hypertestosteronism in hirsute women". Fertility and Sterility. 28 (12): 1285–8. doi:10.1016/S0015-0282(16)42970-5. PMID 590535.
  177. ^ Correa de Oliveira RF, Novaes LP, Lima MB, Rodrigues J, Franco S, Khenaifes AI, Francalanci CP (December 1975). "A new treatment for hirsutism". Annals of Internal Medicine. 83 (6): 817–9. doi:10.7326/0003-4819-83-6-817. PMID 1200527.
  178. ^ Richman RA, Underwood LE, French FS, Van Wyk JJ (December 1971). "Adverse effects of large doses of medroxyprogesterone (MPA) in idiopathic isosexual precocity". The Journal of Pediatrics. 79 (6): 963–71. doi:10.1016/s0022-3476(71)80191-9. PMID 4332067.
  179. ^ a b Luciano, A. A. (1992). "Endometriosis—the role of medroxyprogesterone acetate". Journal of Obstetrics and Gynaecology. 12 (sup2): S38–S44. doi:10.3109/01443619209045611. ISSN 0144-3615.
  180. ^ a b Nachtigall LE, Raju U, Banerjee S, Wan L, Levitz M (2000). "Serum estradiol-binding profiles in postmenopausal women undergoing three common estrogen replacement therapies: associations with sex hormone-binding globulin, estradiol, and estrone levels". Menopause. 7 (4): 243–250. doi:10.1097/00042192-200007040-00006. PMID 10914617. S2CID 3076514.
  181. ^ Lundgren S, Lønning PE, Utaaker E, Aakvaag A, Kvinnsland S (June 1990). "Influence of progestins on serum hormone levels in postmenopausal women with advanced breast cancer--I. General findings". Journal of Steroid Biochemistry. 36 (1–2): 99–104. doi:10.1016/0022-4731(90)90118-c. PMID 2362454.
  182. ^ Lundgren S, Lønning PE (June 1990). "Influence of progestins on serum hormone levels in postmenopausal women with advanced breast cancer--II. A differential effect of megestrol acetate and medroxyprogesterone acetate on serum estrone sulfate and sex hormone binding globulin". Journal of Steroid Biochemistry. 36 (1–2): 105–109. doi:10.1016/0022-4731(90)90119-d. PMID 2141886.
  183. ^ Tomić R, Ljungberg B, Damber JE (1988). "Hormonal effects of high dose medroxyprogesterone acetate treatment in males with renal or prostatic adenocarcinoma". Scandinavian Journal of Urology and Nephrology. 22 (1): 15–18. doi:10.1080/00365599.1988.11690377. PMID 2968646.
  184. ^ Merrin PK, Alexander WD (August 1990). "Cushing's syndrome induced by medroxyprogesterone". BMJ. 301 (6747): 345. doi:10.1136/bmj.301.6747.345-a. PMC 1663616. PMID 2144198.
  185. ^ Systemic Effects of Oral Glucocorticoids, from the original on 28 January 2014
  186. ^ Ishida Y, Ishida Y, Heersche JN (August 2002). "Pharmacologic doses of medroxyprogesterone may cause bone loss through glucocorticoid activity: an hypothesis". Osteoporos Int. 13 (8): 601–5. doi:10.1007/s001980200080. PMID 12181616. S2CID 23343761.
  187. ^ Kuhl H (2005). "Pharmacology of estrogens and progestogens: influence of different routes of administration" (PDF). Climacteric. 8 Suppl 1: 3–63. doi:10.1080/13697130500148875. PMID 16112947. S2CID 24616324.
medroxyprogesterone, acetate, confused, with, progesterone, natural, bioidentical, progestogen, also, known, depot, medroxyprogesterone, acetate, dmpa, injectable, form, sold, under, brand, name, depo, provera, among, others, hormonal, medication, progestin, t. Not to be confused with progesterone a natural and bioidentical progestogen Medroxyprogesterone acetate MPA also known as depot medroxyprogesterone acetate DMPA in injectable form and sold under the brand name Depo Provera among others is a hormonal medication of the progestin type 10 4 It is used as a method of birth control and as a part of menopausal hormone therapy 10 4 It is also used to treat endometriosis abnormal uterine bleeding paraphilia and certain types of cancer 10 The medication is available both alone and in combination with an estrogen 11 12 It is taken by mouth used under the tongue or by injection into a muscle or fat 10 Medroxyprogesterone acetateClinical dataPronunciation m ɛ ˌ d r ɒ k s i p r oʊ ˈ dʒ ɛ s t er oʊ n ˈ ae s ɪ t eɪ t me DROKS ee proh JES ter ohn ASS i tayt 1 Trade namesProvera Depo Provera Depo SubQ Provera 104 Curretab Cycrin Farlutal Gestapuran Perlutex Veramix others 2 Other namesMPA DMPA Methylhydroxy wbr progesterone acetate Methylacetoxy wbr progesterone MAP Methypregnone Metipregnone 6a Methyl 17a hydroxyprogesterone acetate 6a Methyl 17a acetoxyprogesterone 6a Methyl 17a hydroxypregn 4 ene 3 20 dione acetate NSC 26386AHFS Drugs comMonographMedlinePlusa604039Routes ofadministrationBy mouth sublingual intramuscular injection subcutaneous injectionDrug classProgestogen Progestin Progestogen ester Antigonadotropin Steroidal antiandrogenATC codeG03AC06 WHO G03DA02 WHO L02AB02 WHO Legal statusLegal statusAU S4 Prescription only US WARNING 3 Rx only In general Prescription only Pharmacokinetic dataBioavailabilityBy mouth 100 4 5 Protein binding88 to albumin 5 MetabolismLiver hydroxylation CYP3A4 reduction conjugation 6 4 9 Elimination half lifeBy mouth 12 33 hours 6 4 IM aq susp 50 days 7 SC aq susp 40 days 8 ExcretionUrine as conjugates 6 IdentifiersIUPAC name 6S 8R 9S 10R 13S 14S 17R 17 acetyl 6 10 13 trimethyl 3 oxo 2 6 7 8 9 11 12 14 15 16 decahydro 1H cyclopenta a phenanthren 17 yl acetateCAS Number71 58 9PubChem CID6279IUPHAR BPS2879DrugBankDB00603ChemSpider6043UNIIC2QI4IOI2GKEGGC08150 YChEBICHEBI 6716ChEMBLChEMBL717 YCompTox Dashboard EPA DTXSID0025527ECHA InfoCard100 000 689Chemical and physical dataFormulaC 24H 34O 4Molar mass386 532 g mol 13D model JSmol Interactive imageMelting point207 to 209 C 405 to 408 F SMILES C C H 1C C H 2 C H CC C 3 C H 2CC C 3 C O C OC O C C C 4 C1 CC O CC4 CInChI InChI InChI 1S C24H34O4 c1 14 12 18 19 22 4 9 6 17 27 13 21 14 22 7 10 23 5 20 18 8 11 24 23 15 2 25 28 16 3 26 h13 14 18 20H 6 12H2 1 5H3 t14 18 19 20 22 23 24 m0 s1Key PSGAAPLEWMOORI PEINSRQWSA N verify Common side effects include menstrual disturbances such as absence of periods abdominal pain and headaches 10 More serious side effects include bone loss blood clots allergic reactions and liver problems 10 Use is not recommended during pregnancy as it may harm the baby 10 MPA is an artificial progestogen and as such activates the progesterone receptor the biological target of progesterone 4 It also has androgenic activity and weak glucocorticoid activity Due to its progestogenic activity MPA decreases the body s release of gonadotropins and can suppress sex hormone levels 13 It works as a form of birth control by preventing ovulation 10 MPA was discovered in 1956 and was introduced for medical use in the United States in 1959 14 15 10 It is on the World Health Organization s List of Essential Medicines 16 MPA is the most widely used progestin in menopausal hormone therapy and in progestogen only birth control 17 18 DMPA is approved for use as a form of long acting birth control in more than 100 countries 19 20 In 2020 it was the 214th most commonly prescribed medication in the United States with more than 2 million prescriptions 21 22 Contents 1 Medical uses 1 1 Birth control 1 1 1 Effectiveness 1 1 2 Advantages 1 2 Comparison 1 3 Available forms 2 Contraindications 3 Side effects 3 1 Mood changes 3 2 Long term effects 3 2 1 Blood clots 3 2 2 Bone density 3 3 HIV risk 4 Breastfeeding 5 Overdose 6 Interactions 7 Pharmacology 7 1 Pharmacodynamics 7 1 1 Progestogenic activity 7 1 2 Antigonadotropic and anticorticotropic effects 7 1 3 Androgenic activity 7 1 4 Glucocorticoid activity 7 1 5 Steroidogenesis inhibition 7 1 6 GABAA receptor allosteric modulation 7 1 7 Appetite stimulation 7 1 8 Other activity 7 2 Pharmacokinetics 7 2 1 Absorption 7 2 2 Distribution 7 2 3 Metabolism 7 2 4 Elimination 7 2 5 Level effect relationships 7 2 6 Time concentration curves 8 Chemistry 9 History 10 Society and culture 10 1 Generic names 10 2 Brand names 10 3 Availability 10 3 1 United States 10 4 Generation 10 5 Controversy 10 5 1 Outside the United States 10 5 2 United States 11 Research 12 Veterinary use 13 See also 14 References 15 External linksMedical uses editThe most common use of MPA is in the form of DMPA as a long acting progestogen only injectable contraceptive to prevent pregnancy in women It is an extremely effective contraceptive when used with relatively high doses to prevent ovulation MPA is also used in combination with an estrogen in menopausal hormone therapy in postmenopausal women to treat and prevent menopausal symptoms such as hot flashes vaginal atrophy and osteoporosis 4 It is used in menopausal hormone therapy specifically to prevent endometrial hyperplasia and cancer that would otherwise be induced by prolonged unopposed estrogen therapy in women with intact uteruses 4 23 In addition to contraception and menopausal hormone therapy MPA is used in the treatment of gynecological and menstrual disorders such as dysmenorrhea amenorrhea and endometriosis 24 Along with other progestins MPA was developed to allow for oral progestogen therapy as progesterone the progestogen hormone made by the human body could not be taken orally for many decades before the process of micronization was developed and became feasible in terms of pharmaceutical manufacturing 25 DMPA reduces sex drive in men and is used as a form of chemical castration to control inappropriate or unwanted sexual behavior in those with paraphilias or hypersexuality including in convicted sex offenders 26 27 DMPA has also been used to treat benign prostatic hyperplasia as a palliative appetite stimulant for cancer patients and at high doses 800 mg per day to treat certain hormone dependent cancers including endometrial cancer renal cancer and breast cancer 28 29 30 31 32 MPA has also been prescribed in feminizing hormone therapy for transgender women due to its progestogenic and functional antiandrogenic effects 33 It has been used to delay puberty in children with precocious puberty but is not satisfactory for this purpose as it is not able to completely suppress puberty 34 DMPA at high doses has been reported to be definitively effective in the treatment of hirsutism as well 35 Though not used as a treatment for epilepsy MPA has been found to reduce the frequency of seizures and does not interact with antiepileptic medications MPA does not interfere with blood clotting and appears to improve blood parameters for women with sickle cell anemia Similarly MPA does not appear to affect liver metabolism and may improve primary biliary cirrhosis and chronic active hepatitis Women taking MPA may experience spotting shortly after starting the medication but is not usually serious enough to require medical intervention With longer use amenorrhea absence of menstruation can occur as can irregular menstruation which is a major source of dissatisfaction though both can result in improvements with iron deficiency and risk of pelvic inflammatory disease and often do not result in discontinuation of the medication 29 Birth control edit Depot medroxyprogesterone acetate DMPA nbsp BackgroundTypeHormonalFirst use1969 36 Trade namesDepo Provera Depo SubQ Provera 104 othersAHFS Drugs comdepo proveraFailure rates first year Perfect use0 2 37 Typical use6 37 UsageDuration effect3 months 12 14 weeks Reversibility3 18 monthsUser remindersMaximum interval is just under 3 monthsClinic review12 weeksAdvantages and disadvantagesSTI protectionNoPeriod disadvantagesEspecially in first injection may be frequent spottingPeriod advantagesUsually no periods from 2nd injectionBenefitsEspecially good if poor pill compliance Reduced endometrial cancer risk RisksReduced bone density which may reverse after discontinuationMedical notesFor those intending to start family suggest switch 6 months prior to alternative method e g POP allowing more reliable return fertility DMPA under brand names such as Depo Provera and Depo SubQ Provera 104 is used in hormonal birth control as a long lasting progestogen only injectable contraceptive to prevent pregnancy in women 38 39 It is given by intramuscular or subcutaneous injection and forms a long lasting depot from which it is slowly released over a period of several months It takes one week to take effect if given after the first five days of the period cycle and is effective immediately if given during the first five days of the period cycle Estimates of first year failure rates are about 0 3 40 Effectiveness edit Trussell s estimated perfect use first year failure rate for DMPA as the average of failure rates in seven clinical trials at 0 3 40 41 It was considered perfect use because the clinical trials measured efficacy during actual use of DMPA defined as being no longer than 14 or 15 weeks after an injection i e no more than 1 or 2 weeks late for a next injection Prior to 2004 Trussell s typical use failure rate for DMPA was the same as his perfect use failure rate 0 3 42 DMPA estimated typical use first year failure rate 0 3 in Contraceptive Technology 16th revised edition 1994 43 Contraceptive Technology 17th revised edition 1998 44 Adopted in 1998 by the FDA for its current Uniform Contraceptive Labeling guidance 45 In 2004 using the 1995 NSFG failure rate Trussell increased by 10 times his typical use failure rate for DMPA from 0 3 to 3 40 41 DMPA estimated typical use first year failure rate 3 in Contraceptive Technology 18th revised edition 2004 40 Contraceptive Technology 19th revised edition 2007 46 Trussell did not use 1995 NSFG failure rates as typical use failure rates for the other two then newly available long acting contraceptives the Norplant implant 2 3 and the ParaGard copper T 380A IUD 3 7 which were as with DMPA an order of magnitude higher than in clinical trials Since Norplant and ParaGard allow no scope for user error their much higher 1995 NSFG failure rates were attributed by Trussell to contraceptive overreporting at the time of a conception leading to a live birth 40 47 41 Advantages edit DMPA has a number of advantages and benefits 48 49 39 50 Highly effective at preventing pregnancy Injected every 12 weeks The only continuing action is to book subsequent follow up injections every twelve weeks and to monitor side effects to ensure that they do not require medical attention No estrogen No increased risk of deep vein thrombosis pulmonary embolism stroke or myocardial infarction Minimal drug interactions compared to other hormonal contraceptives Decreased risk of endometrial cancer DMPA reduces the risk of endometrial cancer by 80 51 52 53 The reduced risk of endometrial cancer in DMPA users is thought to be due to both the direct anti proliferative effect of progestogen on the endometrium and the indirect reduction of estrogen levels by suppression of ovarian follicular development 54 Decreased risk of iron deficiency anemia pelvic inflammatory disease PID ectopic pregnancy 55 56 and uterine fibroidsNote this is no longer considered valid Progesterones have been found to be the critical mitogen for uterine fibroid tumours 57 Decreased incidence and severity of sickle cell crises in women with sickle cell disease 39 The United Kingdom Department of Health has actively promoted Long Acting Reversible Contraceptive use since 2008 particularly for young people 58 following on from the October 2005 National Institute for Health and Clinical Excellence guidelines 59 Giving advice on these methods of contraception has been included in the 2009 Quality and Outcomes Framework good practice for primary care 60 Comparison edit Proponents of bioidentical hormone therapy believe that progesterone offers fewer side effects and improved quality of life compared to MPA 61 The evidence for this view has been questioned MPA is better absorbed when taken by mouth with a much longer elimination half life leading to more stable blood levels 62 though it may lead to greater breast tenderness and more sporadic vaginal bleeding 61 The two compounds do not differentiate in their ability to suppress endometrial hyperplasia 61 nor does either increase the risk of pulmonary embolism 63 The two medications have not been adequately compared in direct tests to clear conclusions about safety and superiority 25 Available forms edit See also Conjugated estrogens medroxyprogesterone acetate Estradiol medroxyprogesterone acetate and Estradiol cypionate medroxyprogesterone acetate MPA is available alone in the form of 2 5 5 and 10 mg oral tablets as a 150 mg mL 1 mL or 400 mg mL 2 5 mL microcrystalline aqueous suspension for intramuscular injection and as a 104 mg 0 65 mL of 160 mg mL microcrystalline aqueous suspension for subcutaneous injection 64 65 It has also been marketed in the form of 100 200 250 400 and 500 mg oral tablets 500 and 1 000 mg oral suspensions and as a 50 mg mL microcrystalline aqueous suspension for intramuscular injection 66 67 A 100 mg mL microcrystalline aqueous suspension for intramuscular injection was previously available as well 64 In addition to single drug formulations MPA is available in the form of oral tablets in combination with conjugated estrogens CEEs estradiol and estradiol valerate for use in menopausal hormone therapy and is available in combination with estradiol cypionate in a microcrystalline aqueous suspension as a combined injectable contraceptive 11 12 64 19 Depo Provera is the brand name for a 150 mg microcrystalline aqueous suspension of DMPA that is administered by intramuscular injection The shot must be injected into thigh buttock or deltoid muscle four times a year every 11 to 13 weeks and provides pregnancy protection instantaneously after the first injection 68 Depo subQ Provera 104 is a variation of the original intramuscular DMPA that is instead a 104 mg microcrystalline dose in aqueous suspension administered by subcutaneous injection It contains 69 of the MPA found in the original intramuscular DMPA formulation It can be injected using a smaller injection needle inserting the medication just below the skin instead of into the muscle in either the abdomen or thigh This subcutaneous injection claims to reduce the side effects of DMPA while still maintaining all the same benefits of the original intramuscular DMPA Contraindications editMPA is not usually recommended because of unacceptable health risk or because it is not indicated in the following cases 69 70 Conditions where the theoretical or proven risks usually outweigh the advantages of using DMPA Multiple risk factors for arterial cardiovascular disease Current deep vein thrombosis or pulmonary embolus Migraine headache with aura while using DMPA Before evaluation of unexplained vaginal bleeding suspected of being a serious condition A history of breast cancer and no evidence of current disease for five years Active liver disease acute viral hepatitis severe decompensated cirrhosis benign or malignant liver tumours Conditions of concern for estrogen deficiency and reduced HDL levels theoretically increasing cardiovascular risk Hypertension with vascular disease Current and history of ischemic heart disease History of stroke Diabetes for over 20 years or with nephropathy retinopathy neuropathy or vascular diseaseConditions which represent an unacceptable health risk if DMPA is used Current or recent breast cancer a hormonally sensitive tumour Conditions where use is not indicated and should not be initiated PregnancyMPA is not recommended for use prior to menarche or before or during recovery from surgery 71 Side effects editIn women the most common adverse effects of MPA are acne changes in menstrual flow drowsiness and can cause birth defects if taken by pregnant women Other common side effects include breast tenderness increased facial hair decreased scalp hair difficulty falling or remaining asleep stomach pain and weight loss or gain 24 Lowered libido has been reported as a side effect of MPA in women 72 DMPA can affect menstrual bleeding After a year of use 55 of women experience amenorrhea missed periods after two years the rate rises to 68 In the first months of use irregular or unpredictable bleeding or spotting or rarely heavy or continuous bleeding was reported 73 MPA does not appear to be associated with vitamin B12 deficiency 74 Data on weight gain with DMPA likewise are inconsistent 75 76 At high doses for the treatment of breast cancer MPA can cause weight gain and can worsen diabetes mellitus and edema particularly of the face Adverse effects peak at five weeks and are reduced with lower doses Less frequent effects may include thrombosis though it is not clear if this is truly a risk it cannot be ruled out painful urination headache nausea and vomiting When used as a form of androgen deprivation therapy in men more frequent complaints include reduced libido impotence reduced ejaculate volume and within three days chemical castration At extremely high doses used to treat cancer not for contraception MPA may cause adrenal suppression and may interfere with carbohydrate metabolism but does not cause diabetes 29 When used as a form of injected birth control there is a delayed return of fertility The average return to fertility is 9 to 10 months after the last injection taking longer for overweight or obese women By 18 months after the last injection fertility is the same as that in former users of other contraceptive methods 48 49 Fetuses exposed to progestogens have demonstrated higher rates of genital abnormalities low birth weight and increased ectopic pregnancy particularly when MPA is used as an injected form of long term birth control A study of accidental pregnancies among poor women in Thailand found that infants who had been exposed to DMPA during pregnancy had a higher risk of low birth weight and an 80 greater than usual chance of dying in the first year of life 77 Mood changes edit There have been concerns about a possible risk of depression and mood changes with progestins like MPA and this has led to reluctance of some clinicians and women to use them 78 79 However contrary to widely held beliefs most research suggests that progestins do not cause adverse psychological effects such as depression or anxiety 78 A 2018 systematic review of the relationship between progestin based contraception and depression included three large studies of DMPA and reported no association between DMPA and depression 80 According to a 2003 review of DMPA the majority of published clinical studies indicate that DMPA is not associated with depression and the overall data support the notion that the medication does not significantly affect mood 81 In the largest study to have assessed the relationship between MPA and depression to date in which over 3 900 women were treated with DMPA for up to 7 years the incidence of depression was infrequent at 1 5 and the discontinuation rate due to depression was 0 5 80 38 82 This study did not include baseline data on depression 82 and due to the incidence of depression in the study the FDA required package labeling for DMPA stating that women with depression should be observed carefully and that DMPA should be discontinued if depression recurs 80 A subsequent study of 495 women treated with DMPA over the course of 1 year found that the mean depression score slightly decreased in the whole group of continuing users from 7 4 to 6 7 by 9 5 and decreased in the quintile of that group with the highest depression scores at baseline from 15 4 to 9 5 by 38 82 Based on the results of this study and others a consensus began emerging that DMPA does not in fact increase the risk of depression nor worsen the severity of pre existing depression 76 82 38 Similarly to the case of DMPA for hormonal contraception the Heart and Estrogen Progestin Replacement Study HERS a study of 2 763 postmenopausal women treated with 0 625 mg day oral CEEs plus 2 5 mg day oral MPA or placebo for 36 months as a method of menopausal hormone therapy found no change in depressive symptoms 83 84 85 However some small studies have reported that progestins like MPA might counteract beneficial effects of estrogens against depression 78 4 86 Long term effects edit The Women s Health Initiative investigated the use of a combination of oral CEEs and MPA compared to placebo The study was prematurely terminated when previously unexpected risks were discovered specifically the finding that though the all cause mortality was not affected by the hormone therapy the benefits of menopausal hormone therapy reduced risk of hip fracture colorectal and endometrial cancer and all other causes of death were offset by increased risk of coronary heart disease breast cancer strokes and pulmonary embolism 87 When combined with CEEs MPA has been associated with an increased risk of breast cancer dementia and thrombus in the eye In combination with estrogens in general MPA may increase the risk of cardiovascular disease with a stronger association when used by postmenopausal women also taking CEEs It was because of these unexpected interactions that the Women s Health Initiative study was ended early due to the extra risks of menopausal hormone therapy 88 resulting in a dramatic decrease in both new and renewal prescriptions for hormone therapy 89 Long term studies of users of DMPA have found slight or no increased overall risk of breast cancer However the study population did show a slightly increased risk of breast cancer in recent users DMPA use in the last four years under age 35 similar to that seen with the use of combined oral contraceptive pills 73 vte Results of the Women s Health Initiative WHI menopausal hormone therapy randomized controlled trials Clinical outcome Hypothesized effect on risk Estrogen and progestogen CEs 0 625 mg day p o MPA 2 5 mg day p o n 16 608 with uterus 5 2 5 6 years follow up Estrogen alone CEs 0 625 mg day p o n 10 739 no uterus 6 8 7 1 years follow up HR 95 CI AR HR 95 CI ARCoronary heart disease Decreased 1 24 1 00 1 54 6 10 000 PYs 0 95 0 79 1 15 3 10 000 PYsStroke Decreased 1 31 1 02 1 68 8 10 000 PYs 1 37 1 09 1 73 12 10 000 PYsPulmonary embolism Increased 2 13 1 45 3 11 10 10 000 PYs 1 37 0 90 2 07 4 10 000 PYsVenous thromboembolism Increased 2 06 1 57 2 70 18 10 000 PYs 1 32 0 99 1 75 8 10 000 PYsBreast cancer Increased 1 24 1 02 1 50 8 10 000 PYs 0 80 0 62 1 04 6 10 000 PYsColorectal cancer Decreased 0 56 0 38 0 81 7 10 000 PYs 1 08 0 75 1 55 1 10 000 PYsEndometrial cancer 0 81 0 48 1 36 1 10 000 PYs Hip fractures Decreased 0 67 0 47 0 96 5 10 000 PYs 0 65 0 45 0 94 7 10 000 PYsTotal fractures Decreased 0 76 0 69 0 83 47 10 000 PYs 0 71 0 64 0 80 53 10 000 PYsTotal mortality Decreased 0 98 0 82 1 18 1 10 000 PYs 1 04 0 91 1 12 3 10 000 PYsGlobal index 1 15 1 03 1 28 19 10 000 PYs 1 01 1 09 1 12 2 10 000 PYsDiabetes 0 79 0 67 0 93 0 88 0 77 1 01Gallbladder disease Increased 1 59 1 28 1 97 1 67 1 35 2 06Stress incontinence 1 87 1 61 2 18 2 15 1 77 2 82Urge incontinence 1 15 0 99 1 34 1 32 1 10 1 58Peripheral artery disease 0 89 0 63 1 25 1 32 0 99 1 77Probable dementia Decreased 2 05 1 21 3 48 1 49 0 83 2 66Abbreviations CEs conjugated estrogens MPA medroxyprogesterone acetate p o per oral HR hazard ratio AR attributable risk PYs person years CI confidence interval Notes Sample sizes n include placebo recipients which were about half of patients Global index is defined for each woman as the time to earliest diagnosis for coronary heart disease stroke pulmonary embolism breast cancer colorectal cancer endometrial cancer estrogen plus progestogen group only hip fractures and death from other causes Sources See template Blood clots edit DMPA has been associated in multiple studies with a higher risk of venous thromboembolism VTE when used as a form of progestogen only birth control in premenopausal women 90 91 92 93 The increase in incidence of VTE ranges from 2 2 fold to 3 6 fold 90 91 92 93 Elevated risk of VTE with DMPA is unexpected as DMPA has little or no effect on coagulation and fibrinolytic factors 94 95 and progestogens by themselves normally do not increase the risk of thrombosis 91 92 It has been argued that the higher incidence with DMPA has reflected preferential prescription of DMPA to women considered to be at an increased risk of VTE 91 Alternatively it is possible that MPA may be an exception among progestins in terms of VTE risk 96 97 98 A 2018 meta analysis reported that MPA was associated with a 2 8 fold higher risk of VTE than other progestins 97 It is possible that the glucocorticoid activity of MPA may increase the risk of VTE 4 99 98 Bone density edit DMPA may cause reduced bone density in premenopausal women and in men when used without an estrogen particularly at high doses though this appears to be reversible to a normal level even after years of use On 17 November 2004 the United States Food and Drug Administration put a black box warning on the label indicating that there were potential adverse effects of loss of bone mineral density 100 101 While it causes temporary bone loss most women fully regain their bone density after discontinuing use 75 The World Health Organization WHO recommends that the use not be restricted 102 103 The American College of Obstetricians and Gynecologists notes that the potential adverse effects on BMD be balanced against the known negative effects of unintended pregnancy using other birth control methods or no method particularly among adolescents Three studies have suggested that bone loss is reversible after the discontinuation of DMPA 104 105 106 Other studies have suggested that the effect of DMPA use on postmenopausal bone density is minimal 107 perhaps because DMPA users experience less bone loss at menopause 108 Use after peak bone mass is associated with increased bone turnover but no decrease in bone mineral density 109 The FDA recommends that DMPA not be used for longer than two years unless there is no viable alternative method of contraception due to concerns over bone loss 101 However a 2008 Committee Opinion from the American Congress of Obstetricians and Gynecologists ACOG advises healthcare providers that concerns about bone mineral density loss should neither prevent the prescription of or continuation of DMPA beyond two years of use 110 HIV risk edit There is uncertainty regarding the risk of HIV acquisition among DMPA users some observational studies suggest an increased risk of HIV acquisition among women using DMPA while others do not 111 The World Health Organization issued statements in February 2012 and July 2014 saying the data did not warrant changing their recommendation of no restriction Medical Eligibility for Contraception MEC category 1 on the use of DMPA in women at high risk for HIV 112 113 Two meta analyses of observational studies in sub Saharan Africa were published in January 2015 114 They found a 1 4 to 1 5 fold increase risk of HIV acquisition for DMPA users relative to no hormonal contraceptive use 115 116 In January 2015 the Faculty of Sexual amp Reproductive Healthcare of the Royal College of Obstetricians and Gynaecologists issued a statement reaffirming that there is no reason to advise against use of DMPA in the United Kingdom even for women at high risk of HIV infection 117 A systematic review and meta analysis of risk of HIV infection in DMPA users published in fall of 2015 stated that the epidemiological and biological evidence now make a compelling case that DMPA adds significantly to the risk of male to female HIV transmission 118 In 2019 a randomized controlled trial found no significant association between DMPA use and HIV 119 Breastfeeding editMPA may be used by breastfeeding mothers Heavy bleeding is possible if given in the immediate postpartum time and is best delayed until six weeks after birth It may be used within five days if not breast feeding While a study showed no significant difference in birth weights or incidence of birth defects and no significant alternation of immunity to infectious disease caused by breast milk containing DMPA a subgroup of babies whose mothers started DMPA at two days postpartum had a 75 higher incidence of doctor visits for infectious diseases during their first year of life 120 A larger study with longer follow up concluded that use of DMPA during pregnancy or breastfeeding does not adversely affect the long term growth and development of children This study also noted that children with DMPA exposure during pregnancy and lactation had an increased risk of suboptimal growth in height but that after adjustment for socioeconomic factors by multiple logistic regression there was no increased risk of impaired growth among the DMPA exposed children The study also noted that effects of DMPA exposure on puberty require further study as so few children over the age of 10 were observed 121 Overdose editMPA has been studied at massive dosages of up to 5 000 mg per day orally and 2 000 mg per day via intramuscular injection without major tolerability or safety issues described 122 123 124 Overdose is not described in the Food and Drug Administration FDA product labels for injected MPA Depo Provera or Depo SubQ Provera 104 7 8 In the FDA product label for oral MPA Provera it is stated that overdose of an estrogen and progestin may cause nausea and vomiting breast tenderness dizziness abdominal pain drowsiness fatigue and withdrawal bleeding 6 According to the label treatment of overdose should consist of discontinuation of MPA therapy and symptomatic care 6 Interactions editMPA increases the risk of breast cancer dementia and thrombus when used in combination with CEEs to treat menopausal symptoms 71 When used as a contraceptive MPA does not generally interact with other medications The combination of MPA with aminoglutethimide to treat metastases from breast cancer has been associated with an increase in depression 29 St John s wort may decrease the effectiveness of MPA as a contraceptive due to acceleration of its metabolism 71 Pharmacology editPharmacodynamics edit MPA acts as an agonist of the progesterone androgen and glucocorticoid receptors PR AR and GR respectively 5 activating these receptors with EC50 values of approximately 0 01 nM 1 nM and 10 nM respectively 125 It has negligible affinity for the estrogen receptor 5 The medication has relatively high affinity for the mineralocorticoid receptor but in spite of this it has no mineralocorticoid or antimineralocorticoid activity 4 The intrinsic activities of MPA in activating the PR and the AR have been reported to be at least equivalent to those of progesterone and dihydrotestosterone DHT respectively indicating that it is a full agonist of these receptors 126 127 Relative affinities of MPA and related steroids Progestogen PR AR ER GR MRProgesterone 50 0 0 10 100Chlormadinone acetate 67 5 0 8 0Cyproterone acetate 90 6 0 6 8Medroxyprogesterone acetate 115 5 0 29 160Megestrol acetate 65 5 0 30 0Notes Values are percentages Reference ligands 100 were promegestone for the PR metribolone for the AR estradiol for the ER dexamethasone for the GR and aldosterone for the MR Sources 4 Progestogenic activity edit MPA is a potent agonist of the progesterone receptor with similar affinity and efficacy relative to progesterone 128 While both MPA and its deacetylated analogue medroxyprogesterone bind to and agonize the PR MPA has approximately 100 fold higher binding affinity and transactivation potency in comparison 128 As such unlike MPA medroxyprogesterone is not used clinically though it has seen some use in veterinary medicine 2 The oral dosage of MPA required to inhibit ovulation i e the effective contraceptive dosage is 10 mg day whereas 5 mg day was not sufficient to inhibit ovulation in all women 129 In accordance the dosage of MPA used in oral contraceptives in the past was 10 mg per tablet 130 For comparison to MPA the dosage of progesterone required to inhibit ovulation is 300 mg day whereas that of the 19 nortestosterone derivatives norethisterone and norethisterone acetate is only 0 4 to 0 5 mg day 131 The mechanism of action of progestogen only contraceptives like DMPA depends on the progestogen activity and dose High dose progestogen only contraceptives such as DMPA inhibit follicular development and prevent ovulation as their primary mechanism of action 132 133 The progestogen decreases the pulse frequency of gonadotropin releasing hormone GnRH release by the hypothalamus which decreases the release of follicle stimulating hormone FSH and luteinizing hormone LH by the anterior pituitary Decreased levels of FSH inhibit follicular development preventing an increase in estradiol levels Progestogen negative feedback and the lack of estrogen positive feedback on LH release prevent a LH surge Inhibition of follicular development and the absence of a LH surge prevent ovulation 48 49 A secondary mechanism of action of all progestogen containing contraceptives is inhibition of sperm penetration by changes in the cervical mucus 134 Inhibition of ovarian function during DMPA use causes the endometrium to become thin and atrophic These changes in the endometrium could theoretically prevent implantation However because DMPA is highly effective in inhibiting ovulation and sperm penetration the possibility of fertilization is negligible No available data support prevention of implantation as a mechanism of action of DMPA 134 MPA and related steroids at the progesterone receptor Compound Ki nM EC50 nM a EC50 nM bProgesterone 4 3 0 9 25Medroxyprogesterone 241 47 32Medroxyprogesterone acetate 1 2 0 6 0 15Footnotes a Coactivator recruitment b Reporter cell line Sources 128 Oral potencies of MPA and related steroids Progestogen OID mg day TFD mg cycle TFD mg day ODP mg day ECD mg day Progesterone 300 4200 200 300 200Chlormadinone acetate 1 7 20 30 10 2 0 5 10Cyproterone acetate 1 0 20 1 0 2 0 1 0Medroxyprogesterone acetate 10 50 5 10 5 0Megestrol acetate 50 5 0Abbreviations OID ovulation inhibiting dosage without additional estrogen TFD endometrial transformation dosage ODP oral dosage in commercial contraceptive preparations ECD estimated comparable dosage Sources 131 99 135 vte Parenteral potencies and durations of progestogens a b Compound Form Dose for specific uses mg c DOA d TFD e POICD f CICD g Algestone acetophenide Oil soln 75 150 14 32 dGestonorone caproate Oil soln 25 50 8 13 dHydroxyprogest acetate h Aq susp 350 9 16 dHydroxyprogest caproate Oil soln 250 500 i 250 500 5 21 dMedroxyprog acetate Aq susp 50 100 150 25 14 50 dMegestrol acetate Aq susp 25 gt 14 dNorethisterone enanthate Oil soln 100 200 200 50 11 52 dProgesterone Oil soln 200 i 2 6 dAq soln 1 2 dAq susp 50 200 7 14 dNotes and sources Sources 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 All given by intramuscular or subcutaneous injection Progesterone production during the luteal phase is 25 15 50 mg day The OID of OHPC is 250 to 500 mg month Duration of action in days Usually given for 14 days Usually dosed every two to three months Usually dosed once monthly Never marketed or approved by this route a b In divided doses 2 125 or 250 mg for OHPC 10 20 mg for P4 Antigonadotropic and anticorticotropic effects edit MPA suppresses the hypothalamic pituitary adrenal HPA and hypothalamic pituitary gonadal HPG axes at sufficient dosages resulting decreased levels of gonadotropins androgens estrogens adrenocorticotropic hormone ACTH and cortisol as well as levels of sex hormone binding globulin SHBG 13 There is evidence that the suppressive effects of MPA on the HPG axis are mediated by activation of both the PR and the AR in the pituitary gland 155 156 Due to its effects on androgen levels MPA can produce strong functional antiandrogenic effects and is used in the treatment of androgen dependent conditions such as precocious puberty in boys and hypersexuality in men 157 In addition since the medication suppresses estrogen levels as well MPA can produce strong functional antiestrogenic effects similarly and has been used to treat estrogen dependent conditions such as precocious puberty in girls and endometriosis in women Due to low estrogen levels the use of MPA without an estrogen poses a risk of decreased bone mineral density and other symptoms of estrogen deficiency 158 Oral MPA has been found to suppress testosterone levels in men by about 30 from 831 ng dL to 585 ng dL at a dosage of 20 mg day by about 45 to 75 average 60 to 150 400 ng dL at a dosage of 60 mg day 159 160 161 and by about 70 to 75 from 832 862 ng dL to 214 251 ng dL at a dosage of 100 mg day 162 163 Dosages of oral MPA of 2 5 to 30 mg day in combination with estrogens have been used to help suppress testosterone levels in transgender women 164 165 166 167 168 169 One study of injectable MPA in men with benign prostatic hyperplasia reported that a single 150 mg dose suppressed testosterone levels into the defined male castrate range lt 58 ng dL within 7 days and that castration levels of testosterone were maintained for 3 months 170 Very high doses of intramuscular MPA of 150 to 500 mg per week but up to 900 mg per week have similarly been reported to suppress testosterone levels to less than 100 ng dL 159 171 The typical initial dose of intramuscular MPA for testosterone suppression in men with paraphilias is 400 or 500 mg per week 159 Androgenic activity edit MPA is a potent full agonist of the AR Its activation of the AR may play an important and major role in its antigonadotropic effects and in its beneficial effects against breast cancer 155 172 173 However although MPA may produce androgenic side effects such as acne and hirsutism in some women 174 175 In fact likely due to its suppressive actions on androgen levels it has been reported that MPA is generally highly effective in improving pre existing symptoms of hirsutism in women with the condition 176 177 However MPA has been seen to cause androgenic effects in children with precocious puberty 178 The reason for the general lack of virilizing effects with MPA despite it binding to and activating the AR with high affinity and this action potentially playing an important role in many of its physiological and therapeutic effects is not entirely clear However MPA has been found to interact with the AR differently compared to other agonists of the receptor such as dihydrotestosterone DHT 126 The result of this difference appears to be that MPA binds to the AR with a similar affinity and intrinsic activity to that of DHT but requires about 100 fold higher concentrations for a comparable induction of gene transcription while at the same time not antagonizing the transcriptional activity of normal androgens like DHT at any concentration 126 Thus this may explain the low propensity of MPA for producing androgenic side effects 126 MPA shows weak androgenic effects on liver protein synthesis similarly to other weakly androgenic progestins like megestrol acetate and 19 nortestosterone derivatives 4 9 While it does not antagonize estrogen induced increases in levels of triglycerides and HDL cholesterol DMPA every other week may decrease levels of HDL cholesterol 4 In addition MPA has been found to suppress sex hormone binding globulin SHBG production by the liver 9 179 180 At a dosage of 10 mg day oral MPA it has been found to decrease circulating SHBG levels by 14 to 18 in women taking 4 mg day oral estradiol valerate 9 Conversely in a study that combined 2 5 mg day oral MPA with various oral estrogens no influence of MPA on estrogen induced increases in SHBG levels was discerned 180 In another higher dose study SHBG levels were lower by 59 in a group of women treated with 50 mg day oral MPA alone relative to an untreated control group of women 179 In massive dose studies of oral or injectable MPA e g 500 1 000 mg day the medication decreased SHBG levels by about 80 181 182 183 Unlike the related steroids megestrol acetate and cyproterone acetate MPA is not an antagonist of the AR and does not have direct antiandrogenic activity 4 As such although MPA is sometimes described as an antiandrogen it is not a true antiandrogen i e AR antagonist 160 Glucocorticoid activity edit As an agonist of the GR MPA has glucocorticoid activity and as a result can cause symptoms of Cushing s syndrome 184 steroid diabetes and adrenal insufficiency at sufficiently high doses 185 It has been suggested that the glucocorticoid activity of MPA may contribute to bone loss 186 The glucocorticoid activity of MPA may also result in an upregulation of the thrombin receptor in blood vessel walls which may contribute to procoagulant effects of MPA and risk of venous thromboembolism and atherosclerosis 4 The relative glucocorticoid activity of MPA is among the highest of the clinically used progestins 4 vte Glucocorticoid activity of selected steroids in vitro Steroid Class TR a GR bDexamethasone Corticosteroid 100Ethinylestradiol Estrogen 0Etonogestrel Progestin 14Gestodene Progestin 27Levonorgestrel Progestin 1Medroxyprogesterone acetate Progestin 29Norethisterone Progestin 0Norgestimate Progestin 1Progesterone Progestogen 10Footnotes a Thrombin receptor TR upregulation in vascular smooth muscle cells VSMCs b RBA for the glucocorticoid receptor GR Strength No effect Pronounced effect Strong effect Sources 187 Steroidogenesis inhibition edit See also Steroidogenesis inhibitor and Neurosteroidogenesis inhibitor MPA has been found to act as a competitive inhibitor of rat 3a hydroxysteroid dehydrogenase 3a HSD 188 189 190 191 This enzyme is essential for the transformation of progesterone deoxycorticosterone and DHT into inhibitory neurosteroids such as allopregnanolone THDOC and 3a androstanediol respectively 192 MPA has been described as very potent in its inhibition of rat 3a HSD with an IC50 of 0 2 mM and a Ki in rat testicular homogenates of 0 42 mM 188 189 However inhibition of 3a HSD by MPA does not appear to have been confirmed using human proteins yet and the concentrations required with rat proteins are far above typical human therapeutic concentrations 188 189 MPA has been identified as a competitive inhibitor of human 3b hydroxysteroid dehydrogenase D5 4 isomerase II 3b HSD II 193 This enzyme is essential for the biosynthesis of sex steroids and corticosteroids 193 The Ki of MPA for inhibition of 3b HSD II is 3 0 mM and this concentration is reportedly near the circulating levels of the medication that are achieved by very high therapeutic dosages of MPA of 5 to 20 mg kg day dosages of 300 to 1 200 mg day for a 60 kg 132 lb person 193 Aside from 3b HSD II other human steroidogenic enzymes including cholesterol side chain cleavage enzyme P450scc CYP11A1 and 17a hydroxylase 17 20 lyase CYP17A1 were not found to be inhibited by MPA 193 MPA has been found to be effective in the treatment of gonadotropin independent precocious puberty and in breast cancer in postmenopausal women at high dosages and inhibition of 3b HSD II could be responsible for its effectiveness in these conditions 193 GABAA receptor allosteric modulation edit Progesterone via transformation into neurosteroids such as 5a dihydroprogesterone 5b dihydroprogesterone allopregnanolone and pregnanolone catalyzed by the enzymes 5a and 5b reductase and 3a and 3b HSD is a positive allosteric modulator of the GABAA receptor and is associated with a variety of effects mediated by this property including dizziness sedation hypnotic states mood changes anxiolysis and cognitive memory impairment as well as effectiveness as an anticonvulsant in the treatment of catamenial epilepsy 192 194 It has also been found to produce anesthesia via this action in animals when administered at sufficiently high dosages 194 MPA was found to significantly reduce seizure incidence when added to existing anticonvulsant regimens in 11 of 14 women with uncontrolled epilepsy and has also been reported to induce anesthesia in animals raising the possibility that it might modulate the GABAA receptor similarly to progesterone 195 196 MPA shares some of the same metabolic routes of progesterone and analogously can be transformed into metabolites such as 5a dihydro MPA DHMPA and 3a 5a tetrahydro MPA THMPA 195 However unlike the reduced metabolites of progesterone DHMPA and THMPA have been found not to modulate the GABAA receptor 195 Conversely unlike progesterone MPA itself actually modulates the GABAA receptor although notably not at the neurosteroid binding site 195 However rather than act as a potentiator of the receptor MPA appears to act as a negative allosteric modulator 195 Whereas the reduced metabolites of progesterone enhance binding of the benzodiazepine flunitrazepam to the GABAA receptor in vitro MPA can partially inhibit the binding of flunitrazepam by up to 40 with half maximal inhibition at 1 mM 195 However the concentrations of MPA required for inhibition are high relative to therapeutic concentrations and hence this action is probably of little or no clinical relevance 195 The lack of potentiation of the GABAA receptor by MPA or its metabolites is surprising in consideration of the apparent anticonvulsant and anesthetic effects of MPA described above and they remain unexplained 195 Clinical studies using massive dosages of up to 5 000 mg day oral MPA and 2 000 mg day intramuscular MPA for 30 days in women with advanced breast cancer have reported no relevant side effects which suggests that MPA has no meaningful direct action on the GABAA receptor in humans even at extremely high dosages 122 Appetite stimulation edit Although MPA and the closely related medication megestrol acetate are effective appetite stimulants at very high dosages 197 the mechanism of action of their beneficial effects on appetite is not entirely clear However glucocorticoid cytokine and possibly anabolic related mechanisms are all thought to possibly be involved and a number of downstream changes have been implicated including stimulation of the release of neuropeptide Y in the hypothalamus modulation of calcium channels in the ventromedial hypothalamus and inhibition of the secretion of proinflammatory cytokines including IL 1a IL 1b IL 6 and TNF a actions that have all been linked to an increase in appetite 198 Other activity edit MPA weakly stimulates the proliferation of MCF 7 breast cancer cells in vitro an action that is independent of the classical PRs and is instead mediated via the progesterone receptor membrane component 1 PGRMC1 199 Certain other progestins are also active in this assay whereas progesterone acts neutrally 199 It is unclear if these findings may explain the different risks of breast cancer observed with progesterone dydrogesterone and other progestins such as medroxyprogesterone acetate and norethisterone in clinical studies 200 Pharmacokinetics edit Absorption edit Surprisingly few studies have been conducted on the pharmacokinetics of MPA at postmenopausal replacement dosages 201 4 The bioavailability of MPA with oral administration is approximately 100 4 A single oral dose of 10 mg MPA has been found to result in peak MPA levels of 1 2 to 5 2 ng mL within 2 hours of administration using radioimmunoassay 201 202 Following this levels of MPA decreased to 0 09 to 0 35 ng mL 12 hours post administration 201 202 In another study peak levels of MPA were 3 4 to 4 4 ng mL within 1 to 4 hours of administration of 10 mg oral MPA using radioimmunoassay 201 203 Subsequently MPA levels fell to 0 3 to 0 6 ng mL 24 hours after administration 201 203 In a third study MPA levels were 4 2 to 4 4 ng mL after an oral dose of 5 mg MPA and 6 0 ng mL after an oral dose of 10 mg MPA both using radioimmunoassay as well 201 204 Treatment of postmenopausal women with 2 5 or 5 mg day MPA in combination with estradiol valerate for two weeks has been found to rapidly increase circulating MPA levels with steady state concentrations achieved after three days and peak concentrations occurring 1 5 to 2 hours after ingestion 4 205 With 2 5 mg day MPA levels of the medication were 0 3 ng mL 0 8 nmol L in women under 60 years of age and 0 45 ng mL 1 2 nmol L in women 65 years of age or over and with 5 mg day MPA levels were 0 6 ng mL 1 6 nmol L in women under 60 years of age and in women 65 years of age or over 4 205 Hence area under curve levels of the medication were 1 6 to 1 8 times higher in those who were 65 years of age or older relative to those who were 60 years of age or younger 9 205 As such levels of MPA have been found to vary with age and MPA may have an increased risk of side effects in elderly postmenopausal women 9 4 205 This study assessed MPA levels using liquid chromatography tandem mass spectrometry LC MS MS a more accurate method of blood determinations 205 Oral MPA tablets can be administered sublingually instead of orally 206 207 208 Rectal administration of MPA has also been studied 209 With intramuscular administration of 150 mg microcrystalline MPA in aqueous suspension the medication is detectable in the circulation within 30 minutes serum concentrations vary but generally plateau at 1 0 ng mL 2 6 nmol L for 3 months 210 Following this there is a gradual decline in MPA levels and the medication can be detected in the circulation for as long as 6 to 9 months post injection 210 The particle size of MPA crystals significantly influences its rate of absorption into the body from the local tissue depot when used as a microcrystalline aqueous suspension via intramuscular injection 211 212 213 Smaller crystals dissolve faster and are absorbed more rapidly resulting in a shorter duration of action 211 212 213 Particle sizes can differ between different formulations of MPA potentially influencing clinical efficacy and tolerability 211 212 213 214 Distribution edit The plasma protein binding of MPA is 88 4 9 It is weakly bound to albumin and is not bound to sex hormone binding globulin or corticosteroid binding globulin 4 9 Metabolism edit The elimination half life of MPA via oral administration has been reported as both 11 6 to 16 6 hours 6 and 33 hours 4 whereas the elimination half lives with intramuscular and subcutaneous injection of microcrystalline MPA in aqueous suspension are 50 and 40 days respectively 7 8 The metabolism of MPA is mainly via hydroxylation including at positions C6b C21 C2b and C1b mediated primarily via CYP3A4 but 3 and 5 dihydro and 3 5 tetrahydro metabolites of MPA are also formed 4 9 Deacetylation of MPA and its metabolites into e g medroxyprogesterone has been observed to occur in non human primate research to a substantial extent as well 30 to 70 215 MPA and or its metabolites are also metabolized via conjugation 71 The C6a methyl and C17a acetoxy groups of MPA make it more resistant to metabolism and allow for greater bioavailability than oral progesterone 9 Elimination edit MPA is eliminated 20 to 50 in urine and 5 to 10 in feces following intravenous administration 216 Less than 3 of a dose is excreted in unconjugated form 216 Level effect relationships edit With intramuscular administration the high levels of MPA in the blood inhibit luteinizing hormone and ovulation for several months with an accompanying decrease in serum progesterone to below 0 4 ng mL 210 Ovulation resumes when once blood levels of MPA fall below 0 1 ng mL 210 Serum estradiol remains at approximately 50 pg mL for approximately four months post injection with a range of 10 92 pg mL after several years of use rising once MPA levels fall below 0 5 ng mL 210 Hot flashes are rare while MPA is found at significant blood levels in the body and the vaginal lining remains moist and creased The endometrium undergoes atrophy with small straight glands and a stroma that is decidualized Cervical mucus remains viscous Because of its steady blood levels over the long term and multiple effects that prevent fertilization MPA is a very effective means of birth control 210 Time concentration curves edit Hormone levels with medroxyprogesterone acetate nbsp MPA levels with 2 5 or 5 mg day oral MPA in combination with 1 or 2 mg day estradiol valerate Indivina in postmenopausal women 205 nbsp MPA levels after a single 150 mg intramuscular injection of MPA Depo Provera in aqueous suspension in women 217 218 nbsp MPA levels after a single 25 to 150 mg intramuscular injection of MPA Depo Provera in aqueous suspension in women 217 219 nbsp MPA levels after a single 104 mg subcutaneous injection of MPA Depo SubQ Provera in aqueous suspension in women 8 Chemistry editSee also List of progestogens Progestogen ester List of progestogen esters Steroidal antiandrogen and List of steroidal antiandrogens MPA is a synthetic pregnane steroid and a derivative of progesterone and 17a hydroxyprogesterone 220 2 Specifically it is the 17a acetate ester of medroxyprogesterone or the 6a methylated analogue of hydroxyprogesterone acetate 220 2 MPA is known chemically as 6a methyl 17a acetoxyprogesterone or as 6a methyl 17a acetoxypregn 4 ene 3 20 dione and its generic name is a contraction of 6a methyl 17a hydroxyprogesterone acetate 220 2 MPA is closely related to other 17a hydroxyprogesterone derivatives such as chlormadinone acetate cyproterone acetate and megestrol acetate as well as to medrogestone and nomegestrol acetate 220 2 9a fluoromedroxyprogesterone acetate FMPA the C9a fluoro analogue of MPA and an angiogenesis inhibitor with two orders of magnitude greater potency in comparison to MPA was investigated for the potential treatment of cancers but was never marketed 221 222 History editMPA was independently discovered in 1956 by Syntex and the Upjohn Company 14 15 223 224 It was first introduced on 18 June 1959 by Upjohn in the United States under the brand name Provera 2 5 5 and 10 mg tablets for the treatment of amenorrhea metrorrhagia and recurrent miscarriage 225 226 An intramuscular formulation of MPA now known as DMPA 400 mg mL MPA was also introduced under the brand name brand name Depo Provera in 1960 in the U S for the treatment of endometrial and renal cancer 28 MPA in combination with ethinylestradiol was introduced in 1964 by Upjohn in the U S under the brand name Provest 10 mg MPA and 50 mg ethinylestradiol tablets as an oral contraceptive but this formulation was discontinued in 1970 227 228 130 This formulation was marketed by Upjohn outside of the U S under the brand names Provestral and Provestrol while Cyclo Farlutal or Ciclofarlutal and Nogest S 229 were formulations available outside of the U S with a different dosage 5 mg MPA and 50 or 75 mg ethinylestradiol tablets 230 231 Following its development in the late 1950s DMPA was first assessed in clinical trials for use as an injectable contraceptive in 1963 232 Upjohn sought FDA approval of intramuscular DMPA as a long acting contraceptive under the brand name Depo Provera 150 mg mL MPA in 1967 but the application was rejected 233 234 However this formulation was successfully introduced in countries outside of the United States for the first time in 1969 and was available in over 90 countries worldwide by 1992 36 Upjohn attempted to gain FDA approval of DMPA as a contraceptive again in 1978 and yet again in 1983 but both applications failed similarly to the 1967 application 233 234 However in 1992 the medication was finally approved by the FDA under the brand name Depo Provera for use in contraception 233 A subcutaneous formulation of DMPA was introduced in the United States as a contraceptive under the brand name Depo SubQ Provera 104 104 mg 0 65 mL MPA in December 2004 and subsequently was also approved for the treatment of endometriosis related pelvic pain 235 MPA has also been marketed widely throughout the world under numerous other brand names such as Farlutal Perlutex and Gestapuran among others 2 11 Society and culture editGeneric names edit Medroxyprogesterone acetate is the generic name of the drug and its INN USAN BAN and JAN while medrossiprogesterone is the DCIT and medroxyprogesterone the DCF of its free alcohol form 220 12 2 236 11 It is also known as 6a methyl 17a acetoxyprogesterone MAP or 6a methyl 17a hydroxyprogesterone acetate 220 12 2 11 Brand names edit MPA is marketed under a large number of brand names throughout the world 11 12 2 Its most major brand names are Provera as oral tablets and Depo Provera as an aqueous suspension for intramuscular injection 11 12 2 A formulation of MPA as an aqueous suspension for subcutaneous injection is also available in the United States under the brand name Depo SubQ Provera 104 11 12 Other brand names of MPA formulated alone include Farlutal and Sayana for clinical use and Depo Promone Perlutex Promone E and Veramix for veterinary use 11 12 2 In addition to single drug formulations MPA is marketed in combination with the estrogens CEEs estradiol and estradiol valerate 11 12 2 Brand names of MPA in combination with CEEs as oral tablets in different countries include Prempro Premphase Premique Premia and Premelle 11 12 2 Brand names of MPA in combination with estradiol as oral tablets include Indivina and Tridestra 11 12 2 Availability edit Oral MPA and DMPA are widely available throughout the world 11 Oral MPA is available both alone and in combination with the estrogens CEEs estradiol and estradiol valerate 11 DMPA is registered for use as a form of birth control in more than 100 countries worldwide 19 20 11 The combination of injected MPA and estradiol cypionate is approved for use as a form of birth control in 18 countries 19 United States edit See also List of progestogens available in the United States As of November 2016 update MPA is available in the United States in the following formulations 64 Oral pills Amen Curretab Cycrin Provera 2 5 mg 5 mg 10 mg Aqueous suspension for intramuscular injection Depo Provera 150 mg mL for contraception 400 mg mL for cancer Aqueous suspension for subcutaneous injection Depo SubQ Provera 104 104 mg 0 65 mL for contraception It is also available in combination with an estrogen in the following formulations Oral pills CEEs and MPA Prempro Prempro Premarin Cycrin Premphase Premarin Cycrin 14 14 Premphase 14 14 Prempro Premphase 0 3 mg 1 5 mg 0 45 mg 1 5 mg 0 625 mg 2 5 mg 0 625 mg 5 mgWhile the following formulations have been discontinued Oral pills ethinylestradiol and MPA Provest 50 mg 10 mg Aqueous suspension for intramuscular injection estradiol cypionate and MPA Lunelle 5 mg 25 mg for contraception The state of Louisiana permits sex offenders to be given MPA 237 Generation edit Progestins in birth control pills are sometimes grouped by generation 238 239 While the 19 nortestosterone progestins are consistently grouped into generations the pregnane progestins that are or have been used in birth control pills are typically omitted from such classifications or are grouped simply as miscellaneous or pregnanes 238 239 In any case based on its date of introduction in such formulations of 1964 MPA could be considered a first generation progestin 240 Controversy edit Outside the United States edit In 1994 when DMPA was approved in India India s Economic and Political Weekly reported that The FDA finally licensed the drug in 1990 in response to concerns about the population explosion in the third world and the reluctance of third world governments to license a drug not licensed in its originating country 241 Some scientists and women s groups in India continue to oppose DMPA 242 In 2016 India introduced DMPA depo medroxyprogesterone IM preparation in the public health system 243 The Canadian Coalition on Depo Provera a coalition of women s health professional and advocacy groups opposed the approval of DMPA in Canada 244 Since the approval of DMPA in Canada in 1997 a 700 million class action lawsuit has been filed against Pfizer by users of DMPA who developed osteoporosis In response Pfizer argued that it had met its obligation to disclose and discuss the risks of DMPA with the Canadian medical community 245 Clinical trials for this medication regarding women in Zimbabwe were controversial with regard to human rights abuses and Medical Experimentation in Africa A controversy erupted in Israel when the government was accused of giving DMPA to Ethiopian immigrants without their consent Some women claimed they were told it was a vaccination The Israeli government denied the accusations but instructed the four health maintenance organizations to stop administering DMPA injections to women if there is the slightest doubt that they have not understood the implications of the treatment 246 United States edit There was a long controversial history regarding the approval of DMPA by the U S Food and Drug Administration The original manufacturer Upjohn applied repeatedly for approval FDA advisory committees unanimously recommended approval in 1973 1975 and 1992 as did the FDA s professional medical staff but the FDA repeatedly denied approval Ultimately on 29 October 1992 the FDA approved DMPA for birth control which had by then been used by over 30 million women since 1969 and was approved and being used by nearly 9 million women in more than 90 countries including the United Kingdom France Germany Sweden Thailand New Zealand and Indonesia 247 Points in the controversy included Animal testing for carcinogenicity DMPA caused breast cancer tumors in dogs Critics of the study claimed that dogs are more sensitive to artificial progesterone and that the doses were too high to extrapolate to humans The FDA pointed out that all substances carcinogenic to humans are carcinogenic to animals as well and that if a substance is not carcinogenic it does not register as a carcinogen at high doses Levels of DMPA which caused malignant mammary tumors in dogs were equivalent to 25 times the amount of the normal luteal phase progesterone level for dogs This is lower than the pregnancy level of progesterone for dogs and is species specific 248 DMPA caused endometrial cancer in monkeys 2 of 12 monkeys tested the first ever recorded cases of endometrial cancer in rhesus monkeys 249 However subsequent studies have shown that in humans DMPA reduces the risk of endometrial cancer by approximately 80 51 52 53 Speaking in comparative terms regarding animal studies of carcinogenicity for medications a member of the FDA s Bureau of Drugs testified at an agency DMPA hearing Animal data for this drug is more worrisome than any other drug we know of that is to be given to well people Cervical cancer in Upjohn NCI studies Cervical cancer was found to be increased as high as 9 fold in the first human studies recorded by the manufacturer and the National Cancer Institute 250 However numerous larger subsequent studies have shown that DMPA use does not increase the risk of cervical cancer 251 252 253 254 255 Coercion and lack of informed consent Testing or use of DMPA was focused almost exclusively on women in developing countries and poor women in the United States 256 raising serious questions about coercion and lack of informed consent particularly for the illiterate 257 and for mentally disabled people who in some reported cases were given DMPA long term for reasons of menstrual hygiene although they were not sexually active 258 Atlanta Grady Study Upjohn studied the effect of DMPA for 11 years in Atlanta mostly on black women who were receiving public assistance but did not file any of the required follow up reports with the FDA Investigators who eventually visited noted that the studies were disorganized They found that data collection was questionable consent forms and protocol were absent that those women whose consent had been obtained at all were not told of possible side effects Women whose known medical conditions indicated that use of DMPA would endanger their health were given the shot Several of the women in the study died some of cancer but some for other reasons such as suicide due to depression Over half the 13 000 women in the study were lost to followup due to sloppy record keeping Consequently no data from this study was usable 256 WHO Review In 1992 the WHO presented a review of DMPA in four developing countries to the FDA The National Women s Health Network and other women s organizations testified at the hearing that the WHO was not objective as the WHO had already distributed DMPA in developing countries DMPA was approved for use in United States on the basis of the WHO review of previously submitted evidence from countries such as Thailand evidence which the FDA had deemed insufficient and too poorly designed for assessment of cancer risk at a prior hearing The Alan Guttmacher Institute has speculated that United States approval of DMPA may increase its availability and acceptability in developing countries 256 259 In 1995 several women s health groups asked the FDA to put a moratorium on DMPA and to institute standardized informed consent forms 260 Research editDMPA was studied by Upjohn for use as a progestogen only injectable contraceptive in women at a dose of 50 mg once a month but produced poor cycle control and was not marketed for this use at this dosage 261 A combination of DMPA and polyestradiol phosphate an estrogen and long lasting prodrug of estradiol was studied in women as a combined injectable contraceptive for use by intramuscular injection once every three months 262 263 264 High dose oral and intramuscular MPA monotherapy has been studied in the treatment of prostate cancer but was found to be inferior to monotherapy with cyproterone acetate or diethylstilbestrol 265 266 267 High dose oral MPA has been studied in combination with diethylstilbestrol and CEEs as an addition to high dose estrogen therapy for the treatment of prostate cancer in men but was not found to provide better effectiveness than diethylstilbestrol alone 268 DMPA has been studied for use as a potential male hormonal contraceptive in combination with the androgens anabolic steroids testosterone and nandrolone 19 nortestosterone in men 269 However it was never approved for this indication 269 MPA was investigated by InKine Pharmaceutical Salix Pharmaceuticals and the University of Pennsylvania as a potential anti inflammatory medication for the treatment of autoimmune hemolytic anemia Crohn s disease idiopathic thrombocytopenic purpura and ulcerative colitis but did not complete clinical development and was never approved for these indications 270 271 It was formulated as an oral medication at very high dosages and was thought to inhibit the signaling of proinflammatory cytokines such as interleukin 6 and tumor necrosis factor alpha with a mechanism of action that was said to be similar to that of corticosteroids 270 271 The formulation of MPA had the tentative brand names Colirest and Hematrol for these indications 270 MPA has been found to be effective in the treatment of manic symptoms in women with bipolar disorder 272 Veterinary use editMPA has been used to reduce aggression and spraying in male cats 273 It may be particularly useful for controlling such behaviors in neutered male cats 273 The medication can be administered in cats as an injection once per month 273 See also editConjugated estrogens medroxyprogesterone acetate Estradiol medroxyprogesterone acetate Estradiol cypionate medroxyprogesterone acetate Polyestradiol phosphate medroxyprogesterone acetateReferences edit Medroxyprogesterone Uses Dosage amp Side Effects a b c d e f g h i j k l m n o Index Nominum 2000 International Drug Directory Taylor amp Francis January 2000 pp 638 ISBN 978 3 88763 075 1 Archived from the original on 19 June 2013 FDA sourced list of all drugs with black box warnings Use Download Full Results and View Query links nctr crs fda gov FDA Retrieved 22 October 2023 a b c d e f g h i j k l m n o p q r s t u v w x y z Kuhl H 2005 Pharmacology of estrogens and progestogens influence of different routes of administration PDF Climacteric 8 Suppl 1 3 63 doi 10 1080 13697130500148875 PMID 16112947 S2CID 24616324 a b c d Schindler AE Campagnoli C Druckmann R Huber J Pasqualini JR Schweppe KW Thijssen JH 2008 Classification and pharmacology of progestins Maturitas 61 1 2 171 80 doi 10 1016 j maturitas 2008 11 013 PMID 19434889 a b c d e f Provera PDF FDA 2015 Archived from the original PDF on 11 February 2017 Retrieved 31 March 2018 a b c Depo Provera PDF FDA 2016 Retrieved 31 March 2018 a b c d depo subQ Provera PDF FDA 2017 Retrieved 31 March 2018 a b c d e f g h i j Stanczyk FZ Bhavnani BR September 2015 Reprint of Use of medroxyprogesterone acetate for hormone therapy in postmenopausal women Is it safe J Steroid Biochem Mol Biol 153 151 9 doi 10 1016 j jsbmb 2015 08 013 PMID 26291834 S2CID 23985966 a b c d e f g h i Medroxyprogesterone Acetate The American Society of Health System Pharmacists Archived from the original on 24 December 2016 Retrieved 8 December 2016 a b c d e f g h i j k l m n o Medroxyprogesterone Uses Dosage amp Side Effects Drugs com a b c d e f g h i j k Sweetman Sean C ed 2009 Sex hormones and their modulators Martindale The Complete Drug Reference 36th ed London Pharmaceutical Press pp 2113 2114 ISBN 978 0 85369 840 1 a b Genazzani AR 15 January 1993 Frontiers in Gynecologic and Obstetric Investigation Taylor amp Francis p 320 ISBN 978 1 85070 486 7 Archived from the original on 20 May 2016 a b Roberts SM 7 May 2013 Introduction to Biological and Small Molecule Drug Research and Development Chapter 12 Hormone replacement therapy Elsevier Science pp 9 ISBN 978 0 12 806202 9 medroxyprogesterone acetate also known as Provera discovered simultaneously by Searle and Upjohn in 1956 a b Sneader W 2005 Chapter 18 Hormone analogs Drug discovery a history New York Wiley p 204 ISBN 0 471 89980 1 World Health Organization 2019 World Health Organization model list of essential medicines 21st list 2019 Geneva World Health Organization hdl 10665 325771 WHO MVP EMP IAU 2019 06 License CC BY NC SA 3 0 IGO Meikle AW 1 June 1999 Hormone Replacement Therapy Springer Science amp Business Media pp 383 ISBN 978 1 59259 700 0 Special Programme of Research Development and Research Training in Human Reproduction World Health Organization World Health Organization 2002 Research on Reproductive Health at WHO Biennial Report 2000 2001 World Health Organization pp 17 ISBN 978 92 4 156208 9 a href Template Cite book html title Template Cite book cite book a CS1 maint multiple names authors list link a b c d Bagade O Pawar V Patel R Patel B Awasarkar V Diwate S 2014 Increasing use of long acting reversible contraception safe reliable and cost effective birth control PDF World J Pharm Pharm Sci 3 10 364 392 ISSN 2278 4357 Archived from the original PDF on 10 August 2017 Retrieved 2 August 2018 a b Gunasheela S 14 March 2011 Practical Management of Gynecological Problems JP Medical Ltd pp 39 ISBN 978 93 5025 240 6 The Top 300 of 2020 ClinCalc Retrieved 7 October 2022 Medroxyprogesterone Acetate Drug Usage Statistics ClinCalc Archived from the original on 2 July 2020 Retrieved 7 October 2022 Furness S Roberts H Marjoribanks J Lethaby A August 2012 Hormone therapy in postmenopausal women and risk of endometrial hyperplasia The Cochrane Database of Systematic Reviews 2012 8 CD000402 doi 10 1002 14651858 CD000402 pub4 PMC 7039145 PMID 22895916 a b Medroxyprogesterone MedlinePlus 9 January 2008 Archived from the original on 12 July 2010 Retrieved 2 July 2010 a b Panay N Fenton A February 2010 Bioidentical hormones what is all the hype about Climacteric 13 1 1 3 doi 10 3109 13697130903550250 PMID 20067429 S2CID 244295 Light SA Holroyd S March 2006 The use of medroxyprogesterone acetate for the treatment of sexually inappropriate behaviour in patients with dementia PDF Journal of Psychiatry amp Neuroscience 31 2 132 4 PMC 1413960 PMID 16575429 Archived PDF from the original on 7 March 2016 The chemical knife Archived from the original on 7 January 2009 Retrieved 22 January 2009 a b Depo Provera medroxyprogesterone acetate NDA 012541 Drugs FDA FDA Approved Drug Products retrieved 2 April 2018 Original Approvals or Tentative Approvals 09 23 1960 a b c d Meyler L 2009 Meyler s side effects of endocrine and metabolic drugs Amsterdam Elsevier Science pp 281 284 ISBN 978 0 444 53271 8 Archived from the original on 23 October 2014 Ganzina F October 1979 High dose medroxyprogesterone acetate MPA treatment in advanced breast cancer A review Tumori 65 5 563 85 doi 10 1177 030089167906500507 PMID 390798 S2CID 23378403 Kjaer M September 1988 The role of medroxyprogesterone acetate MPA in the treatment of renal adenocarcinoma Cancer Treat Rev 15 3 195 209 doi 10 1016 0305 7372 88 90003 5 PMID 2974757 Vanderstappen D Bonte J 1992 New trends in the use of medroxyprogesterone acetate as a chemotherapeutic agent in gynecologic malignancies Eur J Gynaecol Oncol 13 2 113 23 PMID 1534051 Gooren L 2005 Hormone treatment of the adult transsexual patient Horm Res 64 Suppl 2 31 6 doi 10 1159 000087751 PMID 16286768 S2CID 42507159 Sachdeva A Dutta AK 31 August 2012 Advances in Pediatrics JP Medical Ltd pp 1202 ISBN 978 93 5025 777 7 Hammerstein J 1990 Antiandrogens Clinical Aspects Hair and Hair Diseases pp 827 886 doi 10 1007 978 3 642 74612 3 35 ISBN 978 3 642 74614 7 a b Nadakavukaren A 28 February 2011 Our Global Environment A Health Perspective Seventh Edition Waveland Press pp 63 ISBN 978 1 4786 0976 6 a b Trussell J 2011 Contraceptive efficacy In Hatcher RA Trussell J Nelson AL Cates W Kowal D Policar MS eds Contraceptive technology 20th revised ed New York Ardent Media pp 779 863 ISBN 978 1 59708 004 0 OCLC 781956734 Table 26 1 Table 3 2 Percentage of women experiencing an unintended pregnancy during the first year of typical use and the first year of perfect use of contraception and the percentage continuing use at the end of the first year United States Archived 15 February 2017 at the Wayback Machine a b c Bakry S Merhi ZO Scalise TJ Mahmoud MS Fadiel A Naftolin F July 2008 Depot medroxyprogesterone acetate an update Arch Gynecol Obstet 278 1 1 12 doi 10 1007 s00404 007 0497 z PMID 18470526 S2CID 11340062 a b c Westhoff C August 2003 Depot medroxyprogesterone acetate injection Depo Provera a highly effective contraceptive option with proven long term safety Contraception 68 2 75 87 doi 10 1016 S0010 7824 03 00136 7 PMID 12954518 a b c d e Trussell J 2004 Contraceptive Efficacy In Hatcher RA Trussell J Stewart FH Nelson AL Cates Jr W Guest F Kowal D eds Contraceptive Technology 18th rev ed New York Ardent Media pp 773 845 ISBN 0 9664902 5 8 a b c Trussell J August 2004 Contraceptive failure in the United States Contraception 70 2 89 96 doi 10 1016 j contraception 2004 03 009 PMC 3638209 PMID 15288211 Trussell J Hatcher RA Cates W Stewart FH Kost K September 1990 A guide to interpreting contraceptive efficacy studies Obstetrics and Gynecology 76 3 Pt 2 558 67 PMID 2199875 Trussell J 1994 Contraceptive Failure Rates In Hatcher RA Trussell J Nelson AL Cates W Kowal D Policar MS eds Contraceptive Technology 16th rev ed New York Irvington Publishers pp 637 688 ISBN 0 8290 3171 5 Trussell J 1998 Contraceptive Efficacy In Hatcher RA Trussell J Stewart F Cates W Stewart GK Guest F Kowal D eds Contraceptive Technology 17th rev ed New York Ardent Media pp 779 844 ISBN 0 9664902 0 7 FDA 1998 Guidance for Industry Uniform Contraceptive Labeling PDF Food and Drug Administration Archived from the original PDF on 25 February 2007 Retrieved 21 June 2007 Trussell J 2007 Contraceptive Efficacy In Hatcher RA Trussell J Nelson AL Cates W Stewart FH Kowal D eds Contraceptive Technology 19th rev ed New York Ardent Media Archived from the original on 31 May 2008 Retrieved 21 June 2007 Trussell J Vaughan B 1999 Contraceptive failure method related discontinuation and resumption of use results from the 1995 National Survey of Family Growth PDF Family Planning Perspectives 31 2 64 72 93 doi 10 2307 2991641 JSTOR 2991641 PMID 10224544 Archived PDF from the original on 2 December 2008 a b c Hatcher RA 2004 Depo Provera Injections Implants and Progestin Only Pills Minipills In Hatcher RA Trussell J Stewart F Cates W Stewart GK Guest F Kowal D eds Contraceptive Technology 18th rev ed New York Ardent Media pp 461 494 ISBN 0 9664902 5 8 a b c Speroff L Darney PD 2005 Injectable Contraception A Clinical Guide for Contraception 4th ed Philadelphia Lippincott Williams amp Wilkins pp 201 220 ISBN 0 7817 6488 2 Mishell DR 2004 Contraception In Strauss III JF Barbieri RL eds Yen and Jaffe s Reproductive Endocrinology 5th ed Philadelphia Elsevier Saunders pp 899 938 ISBN 0 7216 9546 9 a b Kaunitz AM December 2001 Current options for injectable contraception in the United States Seminars in Reproductive Medicine 19 4 331 7 doi 10 1055 s 2001 18641 PMID 11727175 S2CID 39556669 a b Bigrigg A Evans M Gbolade B Newton J Pollard L Szarewski A Thomas C Walling M July 1999 Depo Provera Position paper on clinical use effectiveness and side effects The British Journal of Family Planning 25 2 69 76 PMID 10454658 a b Depot medroxyprogesterone acetate DMPA and risk of endometrial cancer The WHO Collaborative Study of Neoplasia and Steroid Contraceptives International Journal of Cancer 49 2 186 90 September 1991 doi 10 1002 ijc 2910490207 PMID 1831802 S2CID 221776781 Santen RJ 2004 Endocrinology of Breast and Endometrial Cancer In Strauss III JF Barbieri RL eds Yen and Jaffe s Reproductive Endocrinology 5th ed Philadelphia Elsevier Saunders pp 787 809 ISBN 0 7216 9546 9 Bartz D Goldberg AB 2011 Injectable contraceptives In Hatcher RA Trussell J Nelson AL Cates W Jr Kowal D Policar MS eds Contraceptive technology 20th revised ed New York Ardent Media pp 212 213 ISBN 978 1 59708 004 0 OCLC 781956734 Advantages of DMPA Injectables 5 Reduced risk of ectopic pregnancy Compared with women who use no contraceptive at all women who use DMPA have a reduced risk for having an ectopic pregnancy Although the overall risk of pregnancy and thus ectopic pregnancy is lowered by DMPA the possibility of an ectopic pregnancy should be excluded if a woman using DMPA becomes pregnant One study showed that 1 5 of women who got pregnant on DMPA had an ectopic pregnancy the same ectopic rate as women who conceived while not using contraception 27 Borgatta L Murthy A Chuang C Beardsley L Burnhill MS September 2002 Pregnancies diagnosed during Depo Provera use Contraception 66 3 169 72 doi 10 1016 S0010 7824 02 00340 2 PMID 12384205 J Julie Kim Takeshi Kurita Serdar E Bulun Progesterone Action in Endometrial Cancer Endometriosis Uterine Fibroids and Breast Cancer Endocrine Reviews Volume 34 Issue 1 1 February 2013 Pages 130 162 https doi org 10 1210 er 2012 1043 lt ref gt Decreased symptoms of endometriosis Decreased incidence of primary dysmenorrhea ovulation pain and functional ovarian cysts Decreased incidence of seizures in women with epilepsy Additionally unlike most other hormonal contraceptives DMPA s contraceptive effectiveness is not affected by enzyme inducing antiepileptic drugs lt ref name obrien gt O Brien MD Guillebaud J September 2006 Contraception for women with epilepsy Epilepsia 47 9 1419 22 doi 10 1111 j 1528 1167 2006 00671 x PMID 16981856 S2CID 22284176 Increasing use of long acting reversible contraception Nursing Times net 21 October 2008 Archived from the original on 26 August 2009 Retrieved 19 June 2009 CG30 Long acting reversible contraception quick reference guide PDF National Institute for Health and Clinical Excellence Archived from the original PDF on 20 September 2009 Retrieved 19 June 2009 Sexual Health Ruleset PDF New GMS Contract Quality and Outcome Framework Implementation Dataset and Business Rules Primary Care Commissioning 1 May 2009 Archived PDF from the original on 10 August 2011 Retrieved 19 June 2009 Sumarized at Contraception Management QOF indicators NHS Clinical Knowledge Summaries NHS Institute for Innovation and Improvement Retrieved 19 June 2009 permanent dead link a b c Holtorf K January 2009 The bioidentical hormone debate are bioidentical hormones estradiol estriol and progesterone safer or more efficacious than commonly used synthetic versions in hormone replacement therapy PDF Postgraduate Medicine 121 1 73 85 doi 10 3810 pgm 2009 01 1949 PMID 19179815 S2CID 2060730 Archived from the original PDF on 8 July 2011 Cirigliano M June 2007 Bioidentical hormone therapy a review of the evidence PDF Journal of Women s Health 16 5 600 31 doi 10 1089 jwh 2006 0311 PMID 17627398 Archived from the original PDF on 6 January 2011 Boothby LA Doering PL August 2008 Bioidentical hormone therapy a panacea that lacks supportive evidence Current Opinion in Obstetrics and Gynecology 20 4 400 7 doi 10 1097 GCO 0b013e3283081ae9 PMID 18660693 S2CID 22449765 a b c d Drugs FDA FDA Approved Drug Products United States Food and Drug Administration Archived from the original on 16 November 2016 Retrieved 31 March 2018 Engorn B Flerlage J 1 May 2014 The Harriet Lane Handbook E Book Elsevier Health Sciences pp 846 ISBN 978 0 323 11246 8 Tiziani AP 1 June 2013 Havard s Nursing Guide to Drugs Elsevier Health Sciences pp 989 ISBN 978 0 7295 8162 2 Leidenberger FA 17 April 2013 Klinische Endokrinologie fur Frauenarzte Springer Verlag pp 528 ISBN 978 3 662 08110 5 Stacey Dawn Depo Provera The Birth Control Shot Archived 10 October 2008 at the Wayback Machine Accessed 13 October 2009 WHO 2004 Progestogen only contraceptives Medical Eligibility Criteria for Contraceptive Use 3rd ed Geneva Reproductive Health and Research WHO ISBN 92 4 156266 8 Archived from the original on 31 May 2009 FFPRHC 2006 The UK Medical Eligibility Criteria for Contraceptive Use 2005 2006 PDF Archived from the original PDF on 19 June 2007 Retrieved 11 January 2007 a b c d MedroxyPROGESTERone Drug Information Provided by Lexi Comp Merck Manual 1 December 2009 Archived from the original on 24 July 2010 Retrieved 8 July 2010 King SR 9 November 2012 Neurosteroids and the Nervous System Springer Science amp Business Media pp 45 ISBN 978 1 4614 5559 2 Archived from the original on 5 November 2017 a b Pfizer October 2004 Depo Provera Contraceptive Injection US patient labeling PDF Archived from the original PDF on 6 February 2007 Retrieved 21 February 2007 Amatayakul K Sivasomboon B Thanangkul O September 1978 Vitamin and trace mineral metabolism in medroxyprogesterone acetate users Contraception 18 3 253 69 doi 10 1016 s0010 7824 78 80019 5 PMID 720068 a b Nelson AL 2014 DMPA battered and bruised but still needed and used in the USA Expert Review of Obstetrics amp Gynecology 5 6 673 686 doi 10 1586 eog 10 60 ISSN 1747 4108 a b Aronson JK 15 October 2015 Meyler s Side Effects of Drugs The International Encyclopedia of Adverse Drug Reactions and Interactions Elsevier Science pp 2 ISBN 978 0 444 53716 4 Perhaps surprisingly a consensus seems to be emerging that depot medroxyprogesterone acetate implants do not in fact result in an increase in the incidence of depression or in the severity of pre existing depression even after 1 or 2 years nor do they cause significant weight gain Exposure to DMPA in pregnancy may cause low birth weight Progress in Human Reproduction Research 23 2 3 1992 PMID 12286194 a b c Lobo RA 5 June 2007 Treatment of the Postmenopausal Woman Basic and Clinical Aspects Elsevier pp 211 ISBN 978 0 08 055309 2 Kaunitz AM 1999 Long acting hormonal contraception assessing impact on bone density weight and mood Int J Fertil Womens Med 44 2 110 7 PMID 10338269 Despite the efficacy and increasing acceptability of these long term methods some clinicians and women are reluctant to use them because of concerns regarding reduction in bone density with DMPA and depressive symptoms and body weight issues with both injectables and implants Recent multicenter experience showed no increase in depressive symptoms after 1 year s DMPA use and 2 years Norplant use even among users with the highest mean depressive symptom scores pre therapy a b c Worly BL Gur TL Schaffir J February 2018 The relationship between progestin hormonal contraception and depression a systematic review Contraception 97 6 478 489 doi 10 1016 j contraception 2018 01 010 PMID 29496297 S2CID 3644828 Westhoff C August 2003 Depot medroxyprogesterone acetate injection Depo Provera a highly effective contraceptive option with proven long term safety Contraception 68 2 75 87 doi 10 1016 s0010 7824 03 00136 7 PMID 12954518 Another common patient tolerability concern reported with hormonal contraception is the effect on mood 95 The majority of published reports indicate that DMPA does not cause depressive symptoms In a large 1 year clinical trial of DMPA in 3857 US women fewer than 2 of users reported depression 15 Other reports in various settings including a private practice 96 adolescent clinics 97 98 a psychiatric hospital 99 and inner city family planning clinics 100 101 have not found an adverse effect of DMPA on depression Using a variety of objective indices for depressive symptoms the overall data for both OCs and DMPA are supportive that these agents have no significant effect on mood Although history of mood symptoms prior to OC use may predispose a subgroup of women to negative mood changes the data for DMPA suggest that even women who have depressive symptoms prior to treatment can tolerate therapy with no exacerbation of these symptoms a b c d Westhoff C Truman C Kalmuss D Cushman L Davidson A Rulin M Heartwell S April 1998 Depressive symptoms and Depo Provera Contraception 57 4 237 40 doi 10 1016 s0010 7824 98 00024 9 PMID 9649914 Fink G Pfaff DW Levine J 31 August 2011 Handbook of Neuroendocrinology Academic Press pp 564 ISBN 978 0 12 378554 1 Ryden J 2009 Practical Gynecology A Guide for the Primary Care Physician ACP Press pp 379 ISBN 978 1 934465 05 9 Hlatky MA Boothroyd D Vittinghoff E Sharp P Whooley MA February 2002 Quality of life and depressive symptoms in postmenopausal women after receiving hormone therapy results from the Heart and Estrogen Progestin Replacement Study HERS trial JAMA 287 5 591 7 doi 10 1001 jama 287 5 591 PMID 11829697 Wiegratz I Kuhl H August 2004 Progestogen therapies differences in clinical effects Trends Endocrinol Metab 15 6 277 85 doi 10 1016 j tem 2004 06 006 PMID 15358281 S2CID 35891204 Rossouw JE Anderson GL Prentice RL LaCroix AZ Kooperberg C Stefanick ML Jackson RD Beresford SA Howard BV Johnson KC Kotchen JM Ockene J July 2002 Risks and benefits of estrogen plus progestin in healthy postmenopausal women principal results From the Women s Health Initiative randomized controlled trial PDF JAMA 288 3 321 33 doi 10 1001 jama 288 3 321 PMID 12117397 S2CID 20149703 Prentice RL Anderson GL 2008 The women s health initiative lessons learned Annual Review of Public Health 29 131 50 doi 10 1146 annurev publhealth 29 020907 090947 PMID 18348708 Buist DS Newton KM Miglioretti DL Beverly K Connelly MT Andrade S Hartsfield CL Wei F Chan KA Kessler L November 2004 Hormone therapy prescribing patterns in the United States Obstetrics and Gynecology 104 5 Pt 1 1042 50 doi 10 1097 01 AOG 0000143826 38439 af PMID 15516400 S2CID 31593637 a b Beyer Westendorf J Bauersachs R Hach Wunderle V Zotz RB Rott H October 2018 Sex hormones and venous thromboembolism from contraception to hormone replacement therapy VASA Zeitschrift fur Gefasskrankheiten 47 6 441 450 doi 10 1024 0301 1526 a000726 PMID 30008249 S2CID 51628832 a b c d DeLoughery TG June 2011 Estrogen and thrombosis controversies and common sense Reviews in Endocrine amp Metabolic Disorders 12 2 77 84 doi 10 1007 s11154 011 9178 0 PMID 21559819 S2CID 28053690 a b c Mantha S Karp R Raghavan V Terrin N Bauer KA Zwicker JI August 2012 Assessing the risk of venous thromboembolic events in women taking progestin only contraception a meta analysis BMJ 345 aug07 2 e4944 doi 10 1136 bmj e4944 PMC 3413580 PMID 22872710 a b Tepper NK Jeng G Curtis KM Boutot ME Boulet SL Whiteman MK March 2019 Venous Thromboembolism Among Women Initiating Depot Medroxyprogesterone Acetate Immediately Postpartum Obstetrics and Gynecology 133 3 533 540 doi 10 1097 AOG 0000000000003135 PMID 30741807 van Hylckama Vlieg A Middeldorp S February 2011 Hormone therapies and venous thromboembolism where are we now Journal of Thrombosis and Haemostasis 9 2 257 266 doi 10 1111 j 1538 7836 2010 04148 x PMID 21114755 S2CID 32394836 Benagiano G Primiero FM June 1983 Long acting contraceptives Present status Drugs 25 6 570 609 doi 10 2165 00003495 198325060 00003 PMID 6223801 S2CID 45898359 Rott H February 2019 Birth Control Pills and Thrombotic Risks Differences of Contraception Methods with and without Estrogen Hamostaseologie 39 1 42 48 doi 10 1055 s 0039 1677806 PMID 30669160 S2CID 58947063 a b Scarabin PY August 2018 Progestogens and venous thromboembolism in menopausal women an updated oral versus transdermal estrogen meta analysis Climacteric 21 4 341 345 doi 10 1080 13697137 2018 1446931 PMID 29570359 S2CID 4229701 a b Sitruk Ware R Nath A June 2011 Metabolic effects of contraceptive steroids Reviews in Endocrine amp Metabolic Disorders 12 2 63 75 doi 10 1007 s11154 011 9182 4 PMID 21538049 S2CID 23760705 a b Kuhl H 2011 Pharmacology of Progestogens PDF J Reproduktionsmed Endokrinol 8 1 157 177 Archived PDF from the original on 11 October 2016 Depot medroxyprogesterone acetate and bone effects Committee Opinion 602 June 2014 Archived from the original on 30 April 2015 Retrieved 3 May 2015 a b FDA 17 November 2004 Black Box Warning Added Concerning Long Term Use of Depo Provera Contraceptive Injection Food and Drug Administration Archived from the original on 21 December 2005 Retrieved 12 May 2006 World Health Organization September 2005 Hormonal contraception and bone health Family Planning Archived from the original on 14 May 2006 Retrieved 12 May 2006 Curtis KM Martins SL May 2006 Progestogen only contraception and bone mineral density a systematic review Contraception 73 5 470 87 doi 10 1016 j contraception 2005 12 010 PMID 16627031 Cundy T Cornish J Evans MC Roberts H Reid IR January 1994 Recovery of bone density in women who stop using medroxyprogesterone acetate BMJ 308 6923 247 8 doi 10 1136 bmj 308 6923 247 PMC 2539337 PMID 8111260 Scholes D LaCroix AZ Ichikawa LE Barlow WE Ott SM September 2002 Injectable hormone contraception and bone density results from a prospective study Epidemiology 13 5 581 7 doi 10 1097 00001648 200209000 00015 PMID 12192229 Scholes D LaCroix AZ Ichikawa LE Barlow WE Ott SM February 2005 Change in bone mineral density among adolescent women using and discontinuing depot medroxyprogesterone acetate contraception Archives of Pediatrics amp Adolescent Medicine 159 2 139 44 doi 10 1001 archpedi 159 2 139 PMID 15699307 Orr Walker BJ Evans MC Ames RW Clearwater JM Cundy T Reid IR November 1998 The effect of past use of the injectable contraceptive depot medroxyprogesterone acetate on bone mineral density in normal post menopausal women Clinical Endocrinology 49 5 615 8 doi 10 1046 j 1365 2265 1998 00582 x PMID 10197077 S2CID 22565912 Cundy T Cornish J Roberts H Reid IR May 2002 Menopausal bone loss in long term users of depot medroxyprogesterone acetate contraception American Journal of Obstetrics and Gynecology 186 5 978 83 doi 10 1067 mob 2002 122420 PMID 12015524 Walsh JS Eastell R Peel NF February 2010 Depot medroxyprogesterone acetate use after peak bone mass is associated with increased bone turnover but no decrease in bone mineral density Fertility and Sterility 93 3 697 701 doi 10 1016 j fertnstert 2008 10 004 PMID 19013564 American College of Obstetricians Gynecologists Committee on Gynecologic Practice September 2008 ACOG Committee Opinion No 415 Depot medroxyprogesterone acetate and bone effects Obstetrics and Gynecology 112 3 727 30 doi 10 1097 AOG 0b013e318188d1ec PMID 18757687 Polis CB Phillips SJ Curtis KM Westreich DJ Steyn PS Raymond E Hannaford P Turner AN October 2014 Hormonal contraceptive methods and risk of HIV acquisition in women a systematic review of epidemiological evidence Contraception 90 4 360 90 doi 10 1016 j contraception 2014 07 009 PMID 25183264 WHO Department of Reproductive Health and Research 16 February 2012 Technical Statement Hormonal contraception and HIV Geneva World Health Organization Archived from the original on 30 January 2015 WHO Department of Reproductive Health and Research 23 July 2014 2014 Guidance Statement Hormonal contraceptive methods for women at high risk of HIV and living with HIV PDF Geneva World Health Organization Archived from the original PDF on 30 January 2015 AVAC 27 January 2015 News from the HC HIV front it s raining meta analyses New York AIDS Vaccine Advocacy Coalition Archived from the original on 30 January 2015 Ralph LJ McCoy SI Shiu K Padian NS February 2015 Hormonal contraceptive use and women s risk of HIV acquisition a meta analysis of observational studies The Lancet Infectious Diseases 15 2 181 9 doi 10 1016 S1473 3099 14 71052 7 PMC 4526270 PMID 25578825 Morrison CS Chen PL Kwok C Baeten JM Brown J Crook AM Van Damme L Delany Moretlwe S Francis SC Friedland BA Hayes RJ Heffron R Kapiga S Karim QA Karpoff S Kaul R McClelland RS McCormack S McGrath N Myer L Rees H van der Straten A Watson Jones D van de Wijgert JH Stalter R Low N January 2015 Hormonal contraception and the risk of HIV acquisition an individual participant data meta analysis PLOS Medicine 12 1 e1001778 doi 10 1371 journal pmed 1001778 PMC 4303292 PMID 25612136 Faculty of Sexual Reproductive Healthcare January 2015 CEU Statement Depot medroxyprogesterone acetate DMPA Depo Provera and risk of HIV acquisition PDF London Royal College of Obstetricians and Gynaecologists Archived from the original PDF on 30 January 2015 Brind J Condly SJ Mosher SW Morse AR Kimball J 2015 Risk of HIV Infection in Depot Medroxyprogesterone Acetate DMPA Users A Systematic Review and Meta analysis Issues Law Med 30 2 129 39 PMID 26710371 ECHO Trial Consortium 13 June 2019 HIV incidence among women using intramuscular depot medroxyprogesterone acetate a copper intrauterine device or a levonorgestrel implant for contraception a randomised multicentre open label trial The Lancet 394 10195 303 313 doi 10 1016 S0140 6736 19 31288 7 PMC 6675739 PMID 31204114 Dahlberg K February 1982 Some effects of depo medroxyprogesterone acetate DMPA observations in the nursing infant and in the long term user International Journal of Gynaecology and Obstetrics 20 1 43 8 doi 10 1016 0020 7292 82 90044 3 PMID 6126406 S2CID 8295162 Pardthaisong T Yenchit C Gray R April 1992 The long term growth and development of children exposed to Depo Provera during pregnancy or lactation Contraception 45 4 313 24 doi 10 1016 0010 7824 92 90053 V PMID 1387602 a b Muss HB Cruz JM August 1992 High dose progestin therapy for metastatic breast cancer Annals of Oncology 3 Suppl 3 15 20 doi 10 1093 annonc 3 suppl 3 S15 PMID 1390312 Pannuti F Martoni A Lenaz GR Piana E Nanni P April 1978 A possible new approach to the treatment of metastatic breast cancer massive doses of medroxyprogesterone acetate Cancer Treat Rep 62 4 499 504 PMID 350387 Simons JP Schols AM Hoefnagels JM Westerterp KR ten Velde GP Wouters EF February 1998 Effects of medroxyprogesterone acetate on food intake body composition and resting energy expenditure in patients with advanced nonhormone sensitive cancer a randomized placebo controlled trial Cancer 82 3 553 60 doi 10 1002 sici 1097 0142 19980201 82 3 lt 553 aid cncr18 gt 3 0 co 2 0 PMID 9452274 World Health Organization 2004 Residues of Some Veterinary Drugs in Animals and Food Monographs Prepared by the Sixty second Meeting of the Joint FAO WHO Expert Committee on Food Additives Rome 4 12 February 2004 Food amp Agriculture Org p 49 ISBN 978 92 5 105195 5 Archived from the original on 17 June 2014 a b c d Kemppainen JA Langley E Wong CI Bobseine K Kelce WR Wilson EM March 1999 Distinguishing androgen receptor agonists and antagonists distinct mechanisms of activation by medroxyprogesterone acetate and dihydrotestosterone Molecular Endocrinology 13 3 440 54 doi 10 1210 mend 13 3 0255 PMID 10077001 Bentel JM Birrell SN Pickering MA Holds DJ Horsfall DJ Tilley WD August 1999 Androgen receptor agonist activity of the synthetic progestin medroxyprogesterone acetate in human breast cancer cells Molecular and Cellular Endocrinology 154 1 2 11 20 doi 10 1016 S0303 7207 99 00109 4 PMID 10509795 S2CID 25584803 a b c Pullen MA Laping N Edwards R Bray J September 2006 Determination of conformational changes in the progesterone receptor using ELISA like assays Steroids 71 9 792 8 doi 10 1016 j steroids 2006 05 009 PMID 16784762 S2CID 24703323 Wikstrom A Green B Johansson ED 1984 The plasma concentration of medroxyprogesterone acetate and ovarian function during treatment with medroxyprogesterone acetate in 5 and 10 mg doses Acta Obstetricia et Gynecologica Scandinavica 63 2 163 8 doi 10 3109 00016348409154654 PMID 6233840 S2CID 45767999 a b Blum RW 22 October 2013 Adolescent Health Care Clinical Issues Elsevier Science pp 216 ISBN 978 1 4832 7738 7 a b Schindler AE Campagnoli C Druckmann R Huber J Pasqualini JR Schweppe KW Thijssen JH December 2003 Classification and pharmacology of progestins Maturitas 46 Suppl 1 S7 S16 doi 10 1016 j maturitas 2003 09 014 PMID 14670641 Glasier A 2006 Contraception In DeGroot LJ Jameson JL eds Endocrinology 5th ed Philadelphia Elsevier Saunders pp 2993 3003 ISBN 978 0 7216 0376 6 Loose DS Stancel GM 2006 Estrogens and Progestins In Loose DS Stancel GM eds Goodman amp Gilman s The Pharmacological Basis of Therapeutics 11th ed New York McGraw Hill pp 1541 1571 ISBN 0 07 142280 3 a b Rivera R Yacobson I Grimes D November 1999 The mechanism of action of hormonal contraceptives and intrauterine contraceptive devices American Journal of Obstetrics and Gynecology 181 5 Pt 1 1263 9 doi 10 1016 S0002 9378 99 70120 1 PMID 10561657 Fritz MA Speroff L 28 March 2012 Clinical Gynecologic Endocrinology and Infertility Lippincott Williams amp Wilkins pp 761 ISBN 978 1 4511 4847 3 Knorr K Beller FK Lauritzen C 17 April 2013 Lehrbuch der Gynakologie Springer Verlag pp 214 ISBN 978 3 662 00942 0 Knorr K Knorr Gartner H Beller FK Lauritzen C 8 March 2013 Geburtshilfe und Gynakologie Physiologie und Pathologie der Reproduktion Springer Verlag pp 583 ISBN 978 3 642 95583 9 Labhart A 6 December 2012 Clinical Endocrinology Theory and Practice Springer Science amp Business Media pp 554 ISBN 978 3 642 96158 8 Horsky J Presl J 1981 Hormonal Treatment of Disorders of the Menstrual Cycle In Horsky J Presl K eds Ovarian Function and its Disorders Diagnosis and Therapy Springer Science amp Business Media pp 309 332 doi 10 1007 978 94 009 8195 9 11 ISBN 978 94 009 8195 9 Ufer J 1969 The Principles and Practice of Hormone Therapy in Gynaecology and Obstetrics de Gruyter p 49 ISBN 9783110006148 17a Hydroxyprogesterone caproate is a depot progestogen which is entirely free of side actions The dose required to induce secretory changes in primed endometrium is about 250 mg per menstrual cycle Pschyrembel W 1968 Praktische Gynakologie fur Studierende und Arzte Walter de Gruyter pp 598 601 ISBN 978 3 11 150424 7 Ferin J September 1972 Effects Duration of Action and Metabolism in Man In Tausk M ed Pharmacology of the Endocrine System and Related Drugs Progesterone Progestational Drugs and Antifertility Agents Vol II Pergamon Press pp 13 24 ISBN 978 0080168128 OCLC 278011135 Henzl MR Edwards JA 10 November 1999 Pharmacology of Progestins 17a Hydroxyprogesterone Derivatives and Progestins of the First and Second Generation In Sitruk Ware R Mishell DR eds Progestins and Antiprogestins in Clinical Practice Taylor amp Francis pp 101 132 ISBN 978 0 8247 8291 7 Brotherton J 1976 Sex Hormone Pharmacology Academic Press p 114 ISBN 978 0 12 137250 7 Sang GW April 1994 Pharmacodynamic effects of once a month combined injectable contraceptives Contraception 49 4 361 385 doi 10 1016 0010 7824 94 90033 7 PMID 8013220 Toppozada MK April 1994 Existing once a month combined injectable contraceptives Contraception 49 4 293 301 doi 10 1016 0010 7824 94 90029 9 PMID 8013216 Goebelsmann U 1986 Pharmacokinetics of Contraceptive Steroids in Humans In Gregoire AT Blye RP eds Contraceptive Steroids Pharmacology and Safety Springer Science amp Business Media pp 67 111 doi 10 1007 978 1 4613 2241 2 4 ISBN 978 1 4613 2241 2 Becker H Dusterberg B Klosterhalfen H 1980 Bioavailability of cyproterone acetate after oral and intramuscular application in men author s transl Bioavailability of Cyproterone Acetate after Oral and Intramuscular Application in Men Urologia Internationalis 35 6 381 385 doi 10 1159 000280353 PMID 6452729 Moltz L Haase F Schwartz U Hammerstein J May 1983 Treatment of virilized women with intramuscular administration of cyproterone acetate Efficacy of Intra muscularly Applied Cyproterone Acetate in Hyperandrogenism Geburtshilfe und Frauenheilkunde 43 5 281 287 doi 10 1055 s 2008 1036893 PMID 6223851 Wright JC Burgess DJ 29 January 2012 Long Acting Injections and Implants Springer Science amp Business Media pp 114 ISBN 978 1 4614 0554 2 Chu YH Li Q Zhao ZF April 1986 Pharmacokinetics of megestrol acetate in women receiving IM injection of estradiol megestrol long acting injectable contraceptive The Chinese Journal of Clinical Pharmacology The results showed that after injection the concentration of plasma MA increased rapidly The meantime of peak plasma MA level was 3rd day there was a linear relationship between log of plasma MA concentration and time day after administration in all subjects elimination phase half life t1 2b 14 35 9 1 days Runnebaum BC Rabe T Kiesel L 6 December 2012 Female Contraception Update and Trends Springer Science amp Business Media pp 429 ISBN 978 3 642 73790 9 Artini PG Genazzani AR Petraglia F 11 December 2001 Advances in Gynecological Endocrinology CRC Press pp 105 ISBN 978 1 84214 071 0 King TL Brucker MC Kriebs JM Fahey JO 21 October 2013 Varney s Midwifery Jones amp Bartlett Publishers pp 495 ISBN 978 1 284 02542 2 a b Poulin R Baker D Poirier D Labrie F March 1989 Androgen and glucocorticoid receptor mediated inhibition of cell proliferation by medroxyprogesterone acetate in ZR 75 1 human breast cancer cells Breast Cancer Research and Treatment 13 2 161 72 doi 10 1007 bf01806528 PMID 2525057 S2CID 26116247 Brady BM Anderson RA Kinniburgh D Baird DT April 2003 Demonstration of progesterone receptor mediated gonadotrophin suppression in the human male Clinical Endocrinology 58 4 506 12 doi 10 1046 j 1365 2265 2003 01751 x PMID 12641635 S2CID 12567639 Saleh FM Grudzinskas AJ Bradford JM 11 February 2009 Sex Offenders Identification Risk Assessment Treatment and Legal Issues Oxford University Press p 44 ISBN 978 0 19 517704 6 Archived from the original on 17 June 2014 World Health Organization 2009 Stuart MC Kouimtzi M Hill SR eds WHO Model Formulary 2008 World Health Organization p 368 hdl 10665 44053 ISBN 9789241547659 a b c Lothstein LM 1996 Antiandrogen treatment for sexual disorders Guidelines for establishing a standard of care Sexual Addiction amp Compulsivity 3 4 313 331 doi 10 1080 10720169608400122 ISSN 1072 0162 a b Dangerous Sex Offenders A Task Force Report of the American Psychiatric Association American Psychiatric Pub 1999 pp 112 144 ISBN 978 0 89042 280 9 Kravitz HM Haywood TW Kelly J Liles S Cavanaugh JL 1996 Medroxyprogesterone and paraphiles do testosterone levels matter The Bulletin of the American Academy of Psychiatry and the Law 24 1 73 83 PMID 8891323 Novak E Hendrix JW Chen TT Seckman CE Royer GL Pochi PE October 1980 Sebum production and plasma testosterone levels in man after high dose medroxyprogesterone acetate treatment and androgen administration Acta Endocrinologica 95 2 265 270 doi 10 1530 acta 0 0950265 PMID 6449127 Kirschner MA Schneider G February 1972 Suppression of the pituitary Leydig cell axis and sebum production in normal men by medroxyprogesterone acetate provera Acta Endocrinologica 69 2 385 393 doi 10 1530 acta 0 0690385 PMID 5066846 Asscheman H Gooren LJ 1993 Hormone Treatment in Transsexuals Journal of Psychology amp Human Sexuality 5 4 39 54 doi 10 1300 J056v05n04 03 ISSN 0890 7064 S2CID 144580633 Meriggiola MC Gava G November 2015 Endocrine care of transpeople part II A review of cross sex hormonal treatments outcomes and adverse effects in transwomen Clinical Endocrinology 83 5 607 615 doi 10 1111 cen 12754 hdl 11585 541921 PMID 25692882 S2CID 39706760 Gooren LJ Giltay EJ Bunck MC January 2008 Long term treatment of transsexuals with cross sex hormones extensive personal experience The Journal of Clinical Endocrinology and Metabolism 93 1 19 25 doi 10 1210 jc 2007 1809 PMID 17986639 Deutsch M 17 June 2016 Guidelines for the Primary and Gender Affirming Care of Transgender and Gender Nonbinary People PDF 2nd ed University of California San Francisco Center of Excellence for Transgender Health p 28 Dahl M Feldman JL Goldberg J Jaberi A 2015 Endocrine Therapy for Transgender Adults in British Columbia Suggested Guidelines PDF Vancouver Coastal Health Retrieved 15 August 2018 Leinung MC Feustel PJ Joseph J 2018 Hormonal Treatment of Transgender Women with Oral Estradiol Transgender Health 3 1 74 81 doi 10 1089 trgh 2017 0035 PMC 5944393 PMID 29756046 Onu PE 1995 Depot medroxyprogesterone in the management of benign prostatic hyperplasia European Urology 28 3 229 235 doi 10 1159 000475056 PMID 8536777 Meyer WJ Walker PA Emory LE Smith ER January 1985 Physical metabolic and hormonal effects on men of long term therapy with medroxyprogesterone acetate Fertility and Sterility 43 1 102 109 doi 10 1016 S0015 0282 16 48326 3 PMID 3155506 Birrell SN Hall RE Tilley WD January 1998 Role of the androgen receptor in human breast cancer Journal of Mammary Gland Biology and Neoplasia 3 1 95 103 doi 10 1023 A 1018730519839 PMID 10819508 S2CID 12044431 Buchanan G Birrell SN Peters AA Bianco Miotto T Ramsay K Cops EJ Yang M Harris JM Simila HA Moore NL Bentel JM Ricciardelli C Horsfall DJ Butler LM Tilley WD September 2005 Decreased androgen receptor levels and receptor function in breast cancer contribute to the failure of response to medroxyprogesterone acetate Cancer Research 65 18 8487 96 doi 10 1158 0008 5472 CAN 04 3077 PMID 16166329 Rees MC Hope S Ravnikar V 12 August 2005 The Abnormal Menstrual Cycle Taylor amp Francis p 213 ISBN 978 1 84214 212 7 Archived from the original on 31 December 2013 Aronson JK 20 January 2009 Meyler s Side Effects of Endocrine and Metabolic Drugs Elsevier p 283 ISBN 978 0 444 53271 8 Archived from the original on 31 December 2013 Ettinger B Golditch IM December 1977 Medroxyprogesterone acetate for the evaluation of hypertestosteronism in hirsute women Fertility and Sterility 28 12 1285 8 doi 10 1016 S0015 0282 16 42970 5 PMID 590535 Correa de Oliveira RF Novaes LP Lima MB Rodrigues J Franco S Khenaifes AI Francalanci CP December 1975 A new treatment for hirsutism Annals of Internal Medicine 83 6 817 9 doi 10 7326 0003 4819 83 6 817 PMID 1200527 Richman RA Underwood LE French FS Van Wyk JJ December 1971 Adverse effects of large doses of medroxyprogesterone MPA in idiopathic isosexual precocity The Journal of Pediatrics 79 6 963 71 doi 10 1016 s0022 3476 71 80191 9 PMID 4332067 a b Luciano A A 1992 Endometriosis the role of medroxyprogesterone acetate Journal of Obstetrics and Gynaecology 12 sup2 S38 S44 doi 10 3109 01443619209045611 ISSN 0144 3615 a b Nachtigall LE Raju U Banerjee S Wan L Levitz M 2000 Serum estradiol binding profiles in postmenopausal women undergoing three common estrogen replacement therapies associations with sex hormone binding globulin estradiol and estrone levels Menopause 7 4 243 250 doi 10 1097 00042192 200007040 00006 PMID 10914617 S2CID 3076514 Lundgren S Lonning PE Utaaker E Aakvaag A Kvinnsland S June 1990 Influence of progestins on serum hormone levels in postmenopausal women with advanced breast cancer I General findings Journal of Steroid Biochemistry 36 1 2 99 104 doi 10 1016 0022 4731 90 90118 c PMID 2362454 Lundgren S Lonning PE June 1990 Influence of progestins on serum hormone levels in postmenopausal women with advanced breast cancer II A differential effect of megestrol acetate and medroxyprogesterone acetate on serum estrone sulfate and sex hormone binding globulin Journal of Steroid Biochemistry 36 1 2 105 109 doi 10 1016 0022 4731 90 90119 d PMID 2141886 Tomic R Ljungberg B Damber JE 1988 Hormonal effects of high dose medroxyprogesterone acetate treatment in males with renal or prostatic adenocarcinoma Scandinavian Journal of Urology and Nephrology 22 1 15 18 doi 10 1080 00365599 1988 11690377 PMID 2968646 Merrin PK Alexander WD August 1990 Cushing s syndrome induced by medroxyprogesterone BMJ 301 6747 345 doi 10 1136 bmj 301 6747 345 a PMC 1663616 PMID 2144198 Systemic Effects of Oral Glucocorticoids archived from the original on 28 January 2014 Ishida Y Ishida Y Heersche JN August 2002 Pharmacologic doses of medroxyprogesterone may cause bone loss through glucocorticoid activity an hypothesis Osteoporos Int 13 8 601 5 doi 10 1007 s001980200080 PMID 12181616 S2CID 23343761 Kuhl H 2005 Pharmacology of estrogens and progestogens influence of different routes of administration PDF Climacteric 8 Suppl 1 3 63 doi 10 1080 13697130500148875 PMID 16112947 S2CID 24616324 span, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.