Skip to content
BY 4.0 license Open Access Published by De Gruyter July 10, 2023

The effect of hormonal contraceptive therapy on clinical laboratory parameters: a literature review

  • Ömer Özcan , Wendy P.J. den Elzen , Jacquelien J. Hillebrand , Martin den Heijer , Laura L. van Loendersloot , Johan Fischer , Henrike Hamer , Robert de Jonge and Annemieke C. Heijboer EMAIL logo

Abstract

Hormonal contraceptives (HC) are widely used among women in reproductive ages. In this review, the effects of HCs on 91 routine chemistry tests, metabolic tests, and tests for liver function, hemostatic system, renal function, hormones, vitamins and minerals were evaluated. Test parameters were differently affected by the dosage, duration, composition of HCs and route of administration. Most studies concerned the effects of combined oral contraceptives (COC) on the metabolic, hemostatic and (sex) steroids test results. Although the majority of the effects were minor, a major increase was seen in angiotensinogen levels (90–375 %) and the concentrations of the binding proteins (SHBG [∼200 %], CBG [∼100 %], TBG [∼90 %], VDBP [∼30 %], and IGFBPs [∼40 %]). Also, there were significant changes in levels of their bound molecules (testosterone, T3, T4, cortisol, vitamin D, IGF1 and GH). Data about the effects of all kinds of HCs on all test results are limited and sometimes inconclusive due to the large variety in HC, administration routes and dosages. Still, it can be concluded that HC use in women mainly stimulates the liver production of binding proteins. All biochemical test results of women using HC should be assessed carefully and unexpected test results should be further evaluated for both methodological and pre-analytical reasons. As HCs change over time, future studies are needed to learn more about the effects of other types, routes and combinations of HCs on clinical chemistry tests.

Introduction

The importance of the oral contraceptive pill, also known as “the pill,” cannot be overstated. It was developed at the beginning of the 1960s, and for the first time in human history, women had the power to regulate their own fertility. The pill was referred to as “one of the seven wonders of the modern world” and “the one invention that historians will look back on and say: That characterized the 20th century” [1].

The combined oral contraceptive pill (COC) was developed more or less by chance. The first progestin drugs were tainted with mestranol, a synthetic estrogen, during the first human study in Puerto Rico in 1956 [2]. After breakthrough bleeding occurred as a result of the products’ purification and lower estrogen content, it was decided to keep the estrogen for cycle control, establishing the idea behind combined estrogen-progestin oral contraceptives [3].

Immediately following the pill’s debut, a sizable number of women started using it; however, over time, many health issues emerged, along with evidence of additional health benefits. The first COCs (first generation) on the market contained high doses (100–150 µg) of ethinylestradiol (EE) which were reduced since the mid-1960s due to the association with increased risks of venous thromboembolisms (VTE) and cardiovascular events. While in de 1970s 50 µg was the most prescribed dose, the switch to <50 µg was made from the 1980s since no loss of potency besides less clinical side effects were reported. Currently, the EE concentration in prescribed COC can be variable in which 20 µg is determined as the lowest effective dose.

In order to increase safety, in addition to the decrease of EE levels, other estrogen molecules, new progestins, new treatment regimens, and different delivery methods were added to hormonal contraceptive (HC) formulations. For decades, only the estrogenic molecule EE was utilized in HC. Recently, the novel compounds estradiol (E2), its valerate derivative (E2V), and estetrol (E4) have entered the market. New progestins derived from different compounds were also added to HC formulations, as three generations of progestins derived from testosterone were created. Progestins can be categorized by their activity in addition to the molecule from which they originate as (anti-)estrogenic, (anti-)androgenic, glucocorticoid, or anti-mineralocorticoid. The types and classifications of sex steroids that are being used in HC formulations are listed in Table 1.

Table 1:

Types and classifications of sex steroids used in hormonal contraception formulations. HC formulations consist of synthetic estrogens or natural estrogens and progestins (the androgenic progestins and the anti-androgenic progestins).

Estrogens Ethinyl estradiol (EE) Synthetic estrogen
Estradiol (E2) Natural estrogens
Estradiol valerate (E2V)
Estetrol (E4)

Progestins Estranes Norethisterone (NET) Androgenic effects, Structurally related to testosterone
Norethisterone acetate (NETA)
Norethynodrol
Dienogest
Gonanes Levonorgestrel (LNG)
Desogestrel (DSG)
Etenogestrel (ENG)
Gestedone
Norgestimate (Norelgestromine)
Pregnanes and nor-pregnanes Medroxyprogesterone acetate (MPA) Anti-androgenic effects, structurally related to progesterone
Cyproterone acetate (CPA)
Chlormadinone acetate (CMA)
19-Norprogesterones Trimegestone (TMG)
Nestorone (NES)
Nomegestrol acetate (NOMAC)
Spironolactone derivative Drospirenone (DRSP)

In 2019, HCs were used by about 250 million women globally [4]. The use varies greatly over the world, with Northern and Western Europe, and the United States, having the highest rates. In the US, 17.5 % of women between the ages of 15 and 45 y use COC, which equates to 27.5 % of all contraceptive users. Similarly in the Netherlands nearly 35 % of contraceptive methods are COCs.

There are countless potential COC combinations for both chemicals and dosages. According to Stridham Hall and Trussell’s analysis of COC use in the US, there were over 80 different COC formulations in use [5]. Between nations as well as between different regions of the world, the prescription pattern varies. Different safety data interpretations are a contributing factor, but traditions can have an impact as well. The most popular COCs in the US are those containing EE and norgestimate or drospirenone (DRSP); however, triphasic tablets are also widely used [3]. Due to data showing a decreased risk of VTE in this group of users compared to those on third-generation pills, in 2001, the European Medicines Agency (EMA) suggested second-generation COCs as the first choice (COC containing EE and levonorgestrel [LNG], desogestrel [DSG] or gestodene).

The use of an implant, an intrauterine device (IUD), injections, pills, vaginal rings, and skin patches are other examples of hormonal approaches. HC could be classified as combined (estrogen and progestin) or progestin only according to their contents. They can also be divided as long acting (depot), medium acting (monthly) and short acting (mini-pills).

Because of its wide usage among fertile women, its effects on biochemical test results should strongly be considered while assessing patients’ laboratory results. In the 1970s reviews were published focusing on the effects of COCs on laboratory tests, but the dosages of hormonal contraceptives used at that time were much higher than currently used [6], [7], [8]. In addition to oral HC, vaginal, subdermal, transdermal and injectable HC are on the market now and their use is growing. Therefore, a literature review is needed to understand the effects of modern HCs on clinical chemistry test results. The aim of this review is to create a reference for physicians and laboratory specialists in case of a woman using HC and showing deviating biochemical test results.

Metabolism of hormonal contraceptives

Figure 1 illustrates the metabolism of hormonal contraceptives (HC) in the female body, which varies depending on the administration method. Orally taken HCs undergo significant first-pass metabolism in the intestinal wall and liver, resulting in a 40–60 % loss of the oral dose [9]. This results in the synthesis of their metabolites [10]. Enterohepatic circulation plays an important role in the metabolism of contraceptive steroids like ethinyl estradiol (EE) and estradiol (E2). Most EE is metabolized before entering the bloodstream, while the majority of E2 is excreted in urine [10]. E2 has a plasma half-life of 2.5 h, but its ultimate half-life, including metabolites, is 13–20 h. However, there is considerable inter-subject variability in pharmacokinetics [11]. Drug interactions may also influence the metabolism and efficacy of oral contraceptives.

Figure 1: 
Schematic illustration of metabolism of hormonal contraceptives.
Figure 1:

Schematic illustration of metabolism of hormonal contraceptives.

Progestins are generally well absorbed orally, reaching peak concentration in 1–3 h after first-pass metabolism. Nestorone (NES) is inactive orally [12] but effective in non-oral forms like implants, gels, and rings [13]. Dienogest is suitable for oral use due to its high bioavailability (>90 %) [14], while desogestrel (DSG) is converted to its active metabolite etonogestrel (ETG) in the liver and gut wall [15].

In the circulation, contraceptive steroids are mainly bound (>95 %) to binding proteins, making them vulnerable to influences that might just slightly alter the binding. Therefore, a negligible 2 % reduction in the amount bound could result in an important 50 % increase in the unbound fraction, which is what determines the biologic activity of the steroids. EE exclusively binds to albumin. Despite the existence of albumin variants or deviating albumin concentrations in specific populations, no abnormalities in interaction between HC and albumin have been reported in literature. However, the gestagens have a complex equilibrium between the various fractions and bind to both albumin and sex hormone binding globulin (SHBG), with the extent and tightness of the binding to SHBG depending on their structure [9].

Effects on clinical chemistry tests

Metabolic parameters

COCs are suggested to have a possible deleterious impact on metabolic markers like lipids and carbohydrates [16]. Clinical investigations have demonstrated that the doses, ratios, and potencies of the estrogens and progestogens found in COC have a variety of effects on blood lipid and lipoprotein concentrations [17], [18], [19], [20], [21], [22], [23]; see Table 2 for a summary of all published effects.

Table 2:

Effects of hormonal contraceptives on lipids and lipoproteins.

Total cholesterol

(3–10 %)
2nd and 3rd gen OCs [24]

EE + DSG [18, 25]

EE + DSG or EE + GSD [26]

EE + CMA, EE + DSG [27]

Vaginal ring (EE + ENG)-1 year use [28]

EE + DRSP [29]

EE + DRSP or E4 + DRSP [30]

ENG implant (3 year) [31]
One-rod LNG implant [32]

EE + CMA and EE + DSG [23]

Progestin-only pill [24]

DRSP-only pill [33]

ENG implant [34]

Skin patch (EE + norelgestromine) [35]

EE + LNG, EE + norethindrone or EE + DSG [19]

Progestin only pill (LNG or norethindrone) [19]

EE + any type progestins [36]

Vaginal ring (EE + ENG) [37]

E4 + DRSP, EE + LNG [29]

EE + LNG [27]


(5–20 %)
LNG-IUD [24]

ENG implant [35, 38]

E4 + LNG [30]
Triglycerides

(5–50 %)
EE + CMA and EE + DSG [23]

EE + CMA, EE + DSG [27]

2nd and 3rd gen OCs [24]

EE + LNG, EE + norethindrone, EE + DSG [19]

EE + DSG [18, 25]

EE + DSG or EE + gestodene [26]

E4 + DRSP, EE + LNG, EE + DRSP [29]

EE + DRSP and E4 + DRSP [30]

EE + any type PG [36, 39]

Vaginal ring (EE + etonogestrel)-1 year use [28]

Etonogestrel implant (3 year) [31]
One-rod levonorgestrel implant [32]

LNG-IUD and progestin-only pill [24]

DRSP only pill [33]

Etonogestrel (ENG) implant [34]

Skin patch (EE + norelgestromine) [35]

Progestin only pill (LNG or norethindrone) [19]

Vaginal ring (EE + ENG) [37]

EE + LNG [27]


(5–30 %)
ENG implant [35, 38]

E4 + LNG [30]
LDL-C

(4–10 %)
DMPA injection [18]

EE + any type progestins [39]

EE + LNG [27]
EE + DSG [18, 23, 25]

EE + CMA, EE + DSG [27]

EE + DSG or EE + GSD [26]

EE + any type progestins [36]

E4 + DRSP, EE + LNG, EE + DRSP [29]

EE + DRSP, E4 + LNG and E4 + DRSP [30]

LNG-IUD and progestin-only pill [24]

DRSP-only pill [33]

ENG implant [34]

ENG implant (3 year) [31]

Skin patch (EE + norelgestromine) [35]

Progestin only (LNG or norethindrone) [19]

Vaginal ring (EE + ENG) [37]

Vaginal ring (EE + ENG)-1 year use [28]


(5–30 %)
One-rod LNG implant [32]

EE + CMA [23]

2nd and 3rd gen OCs [24]

ENG implant [35, 38]

EE + LNG, norethindrone or DSG [19]
HDL-C

(8–15 %)
One-rod LNG implant [32]

EE + CMA [23]

EE + CMA, EE + DSG [27]

2nd and 3rd gen OCs [24]

EE + DSG or EE + low dose norethindrone [19]

EE + DSG [18, 25]

EE + DSG or EE + GSD [26]

EE + DRSP [29]

EE + DRSP and E4 + DRSP [30]

EE + any type PG [36, 39]

Vaginal ring (EE + ENG)-1 year use [28]
EE + DSG [23]

E4 + DRSP [29]

Progestin-only pill [24]

DRSP-only pill [33]

ENG implant [34, 35, 38]

ENG implant (3 year) [31]

Skin patch (EE + norelgestromine) [35]

Progestin only pill (LNG or norethindrone) [19]

EE + high dose norethindrone [19]

Vaginal ring (EE + ENG) [37]


(8–20 %)
LNG-IUD [24]

EE + LNG [19, 27, 29]

E4 + LNG [30]

DMPA injection [18]
Apo-B

(11–23 %)
E4 + DRSP, EE + LNG or EE + DRSP [29]
EE + CMA or EE + DSG [23]

Progestin only pill (LNG or norethindrone) [19]


(5 %)
Vaginal ring (EE + ENG)-1 year use [28]
Apo-A1

(10–20 %)
EE + LNG, EE + norethindrone or EE + DSG [19]

E4 + DRSP, EE + DRSP [29]

EE + CMA, EE + DSG [27]

Vaginal ring (EE + ENG)-1 year use [28]
EE + CMA and EE + DSG [23]

EE + LNG [27]

Progestin only pill (LNG) [19]


(5–15 %)
Progestin only (norethindrone) [19]

EE + high dose LNG [19]

EE + LNG [29]
Apo-A2

(11–24 %)
EE + CMA and EE + DSG [23]

EE + CMA, EE + DSG or EE + LNG [27]

EE + LNG, EE + norethindrone or EE + DSG [19]
Progestin only pill (LNG) [19]


(5 %)
Progestin only pill (norethindrone) [19]
Apo-E EE + CMA and EE + DSG [23]
Lipoprotein(a) EE + CMA and EE + DSG [23]

EE + CMA, EE + DSG, EE + LNG [27]

E4 + DRSP, EE + LNG, EE + DRSP [29]
  1. ENG, etenogestrel; E4, estetrol; LNG, levonorgestrel; DMPA, depot medroxyprogesterone acetate; EE, ethinylestradiol; DSG, desogestrel; CMA: chlormadinone acetate; GSD, gestodene; DRSP, drospirenone.

There is no noticeable difference in the lipid profiles between the cyclic and long period regimens (more than 1 year), according to four randomized controlled trials (RCT) [4043]. Over the course of the one-year study period Wiegratz et al. discovered a rise in total cholesterol, high-density lipoprotein, triglycerides, and very-low-density lipoprotein and a decrease in low-density lipoprotein in both groups [40]. Progestins and estrogens can both have an impact on lipid metabolism. The final impact is influenced by the estrogen-progestin dosage and the relative balance between the progestins’ androgenic activity and estrogenic potency. LNG containing combinations that are more androgenic compared to HCs with less androgenic progestins, such as dienogest or nomegestrol acetate mixed with natural estrogen, are demonstrated to have a favorable effect on the lipid profile [44, 45].

Effects of HCs on other metabolic parameters are summarized in Table 3 en will be discussed in this paragraph. Sitruk-Ware et al. emphasized that changes in carbohydrate metabolism (i.e. lowered glucose tolerance and elevated insulin) in case of COC usage were not significant due to availability of low and ultra-low EE containing COCs. EE specifically has a higher impact on hepatic metabolism, and metabolic alterations, as compared to E2 or its valerate [45]. Although earlier progestins (norethindrone, MPA and CPA) could also affect metabolism [45], the latest progestin generations (NOMAC, DRSP and NES) essentially have no androgenic, estrogenic, or glucocorticoid-related adverse effects and no effects on fasting glucose or insulin levels [16, 43, 46].

Table 3:

Effects of hormonal contraceptives on metabolic parameters.

Glucose

(7–16 %)
ENG implant [34]

EE + any type progestins [39]

EE + CMA, EE + DSG, EE + LNG [27]
LNG-IUD and Progestin-only pill [24]

DRSP-only pill [33]

Skin patch (EE + norelgestromine) [35]

Progestin only pill (norethindrone) [19]

EE + any type progestins [36]

Vaginal ring (EE + ENG) [37]

ENG implant [35, 38]

ENG implant (3 year) [31]


(3–7%)
2nd and 3rd gen OCs [24]

ENG implant [35]

Progestin only pill (LNG) [19]

EE + LNG, EE + norethindrone or EE + DSG [19]
Insulin

(10–30 %)
2nd and 3rd gen OCs [24]

EE + LNG, EE + norethindrone or EE + DSG [19]

EE + any type progestins [36, 39]
LNG-IUD and progestin-only pill [24]

DRSP-only pill [33]

ENG implant [34, 35]

Skin patch (EE + norelgestromine) [35]

Progestin only pill (LNG or norethindrone) [19]

Vaginal ring (EE + ENG) [37]

EE + CMA, EE + DSG or EE + LNG [27]
C-Peptide

(9–23 %)
EE + LNG, EE + norethindrone or EE + DSG [19]

EE + DSG or EE + LNG [27]
DRSP-only pill [33]

ENG implant [35]

Skin patch (EE + norelgestromine) [35]

Progestin only pill (LNG or norethindrone) [19]

EE + CMA [27]
Growth hormone

(30–470 %)
EE + DRSP, E4 + DRSP, E4 + LNG [30]

EE + LNG [47]
EE + DNG [47]
IGF-1

(6–42 %)
EE + DRSP, E4 + DRSP, E4 + LNG [30]

EE + LNG, EE + DNG [47]
IGFBP-1

(21–191 %)
EE + DRSP, E4 + DRSP, E4 + LNG [30]
IGFBP-3

(7,5–16 %)
E4 + DRSP, E4 + LNG [30]
EE + DRSP [30]

EE + LNG, EE + DNG [47]
HsCRP or CRP

(100–270 %)
2nd and 3rd gen OCs [24]

EE + DSG or EE + GSD [26]

Skin patch (EE + norelgestromine) [35]

EE + any type progestins [39]
LNG-IUD and progestin-only pill [24]

ENG implant [35]
  1. ENG, etenogestrel; E4, estetrol; LNG, levonorgestrel; DMPA, depot medroxyprogesterone acetate; EE, ethinylestradiol; DSG, desogestrel; CMA, chlormadinone acetate; GSD, gestodene; DRSP, drospirenone.

Contrary to COCs containing EE, COCs containing natural estrogens do not adversely impact the metabolism of glucose and insulin [29, 48].

A previous systematic review showed also no effect of HC use on glucose metabolism [49]. Due to the small number of studies, methodological flaws, and studies that compared the same forms of contraception, no firm conclusions can be made [49].

There is limited data about growth hormone (GH), insulin-like growth factor-1 (IGF-1) and IGF binding proteins concentrations upon HC usage. Some studies showed that GH levels were elevated more than 100 % upon COC (EE/DRSP), whereas IGF-1 levels were 20–50 % reduced. These changes seem to be caused by EE in the COCs, leading to a decrease in liver synthesis of IGF-1 that causes increased GH levels due to feedback interaction [50]. Increased insulin-like growth factor binding protein-1 (IGFBP1) or insulin-like growth factor binding protein-3 (IGFBP3) synthesis in the liver could also be an effect of EE containing COC usage leading to up to 191 and 16 % increase in these levels, respectively [30, 47].

In summary, contraceptives containing less androgenic progestins, i.e. drospirenone (DRSP), lead to improved lipid profiles compared to those containing androgenic progestins, i.e. levonorgestrel (LNG). This also depends on route of administration and dosage. Natural estrogens E2 and E2V cause fewer metabolic alterations than the synthetic EE. Despite statistical significance, except for GH, IGF-1 and IGFBPs changes in laboratory parameters related to metabolism are either small and results stay within the reference interval or data are not consistent (Table 3). Unfortunately, no studies are available to show how fast levels return to baseline after discontinuation of HC usage. Based on the half-life of listed parameters, it could be assumed that all changes might be reversed within 4 weeks after stopping HC. Thus, in case of uncertainty about the influence of HC on metabolic parameters in a woman using HC, these should be re-evaluated at least 4 weeks after HC discontinuation.

Liver function tests

The effect of sex hormones on liver function and the implications for the patient with liver disease are often of concern to obstetricians and gynecologists. The published effects on the liver function tests are shown in Table 4. It is generally known that estrogen receptors exist in the liver [51]. Normal liver tissue, hepatic adenomas, hepatocellular carcinomas, and hepatoblastomas all include estrogen receptors. For sufficient systemic levels to be available after hepatic metabolism, substantial concentrations of EE and a progestin should enter the portal circulation. There have been numerous reports of the hepatic side effects of COC, including cholestasis and the growth of hepatic adenomas. Some estrogen and progestin combinations damage the liver’s ultrastructure and affect a variety of metabolic processes, including protein biosynthesis, energy production, and increased cellular catabolism [52]. Even though these side effects are uncommon and dependent on the dosage, duration and type of OC treatment, the possibility of liver damage makes managing contraception in people with liver illness challenging. Depending on the severity of liver damage due to HC use, effects might not always be represented as elevated alanine aminotransferase (ALT) or aspartate aminotransferase (AST) enzyme levels, since these markers are quite insensitive to small changes.

Table 4:

Effects of hormonal contraceptives on routine chemistry tests.

Uric acid ENG implant or skin patch (EE + norelgestromine) [35]
Total protein ENG implant or skin patch (EE + norelgestromine) [35]
Albumin ENG implant or skin patch (EE + norelgestromine) [35]


(6 %)
EE + DSG or EE + GSD [26]
T. bilirubin EE + DSG or EE + GSD [26]

ENG implant [38]

NOMAC implant [53]

ENG implant or skin patch (EE + norelgestromine) [35]

EE + norgestimate [54]

COC or DMPA [55]
D. bilirubin ENG implant [38]

ENG implant or skin patch (EE + norelgestromine) [35]
AST

(15–20 %)
ENG implant [38]

EE + DRSP [56]
EE + DSG or EE + GSD [26]

One-rod LNG implant [32]

NOMAC implant [53]

ENG implant [57]

ENG implant or Skin patch (EE + norelgestromine) [35]

EE + norgestimate [54]

COC or DMPA [55]
ALT

(30–40 %)
ENG implant [38]

EE + DRSP [56]
EE + DSG or EE + GSD [26]

One-rod LNG implant [32]

NOMAC implant [53]

ENG implant [57]

ENG implant or skin patch (EE + norelgestromine) [35]

EE + norgestimate [54]

COC or DMPA [55]
ALP ENG implant [35]

COC or DMPA [55]


(10–12 %)
EE + DSG or EE + GSD [26]

Skin patch (EE + norelgestromine) [35]
GGT

(25–50 %)
EE + DRSP [56]

ENG implant [35]
EE + DSG or EE + GSD [26]

NOMAC implant [53]

EE + norgestimate [54]
LDH

(7 %)
ENG implant [35]
Skin patch (EE + norelgestromine) [35]
  1. ENG, etenogestrel; LNG, levonorgestrel; DMPA, depot medroxyprogesterone acetate; EE, ethinylestradiol; DSG, desogestrel; GSD, gestodene; DRSP, drospirenone; NOMAC, nomegestrol acetate; COC, combined oral contraceptive.

The effects of HC on liver function are less when the hormones are delivered by parenteral route without liver transit. For instance, it was shown that subdermal implants after three years of use did not significantly impact hepatic enzymes [31].

The unusually elevated liver enzymes and bilirubins related with the development of intrahepatic cholestasis following COC usage is typically attributed to the estrogen component of COC pills [58]. Additionally, bile salt export pump (BSEP) and ATP-binding cassette subfamily B11 (ABC B11) member gene-associated estrogen-induced cholestasis can be found, particularly in women with a history of idiopathic cholestasis of pregnancy or hereditary bilirubin metabolism defects like Dubin Johnson syndrome [59, 60]. Studies on rats have led researchers to hypothesize that canalicular bile transporters (such as multidrug resistance protein 2, which is in charge of biliary secretion of bilirubin glucuronides) may be involved in the estrogen-induced cholestasis [61].

In contrast to estrogens, progestogens rarely contribute to cholestatic liver test changes [62, 63], and can therefore be a good alternative to prevent hepatic test fluctuations, although intrahepatic cholestasis has also been shown after high doses of progesterone treatment in patients with breast cancer [64]. On the other hand, progesterone metabolites may sometimes play a more significant role than estradiol metabolites in the pathogenesis of obstetric cholestasis. Patients with obstetric cholestasis have high concentrations of progesterone metabolites in serum and sulfated progesterone metabolites in urine [65, 66]. Recently, a 49-year-old woman taking progestin only pill (norethisterone) with extremely higher liver parameters related with intrahepatic cholestasis was reported [67]. Therefore, in vulnerable patients with cholangiopathy brought on by steatosis and/or genetic predispositions, progestogens may cause high liver enzymes and bilirubins.

The effects of HCs on the hematology parameters are shown in Table 5. Users of traditionally administered COCs have higher serum transferrin levels than nonusers [68]. Instead of being an indication of iron shortage, the greater transferrin concentration could be the result of the contraceptive pill’s general impact on the levels of transport proteins. Several studies showed that COC users had no differences in hemoglobin vs. non-COC users and that cyclic vs. continuous use of COC also did not influence hemoglobin levels [69, 70]. Thus, it appears that ongoing OC use has little to no effect on the body’s iron status and hemoglobin levels [68].

Table 5:

Effects of hormonal contraceptives on hematology tests.

Hemoglobin

(4.5 %)
ENG implant [38]
EE + DSG or EE + GSD [26]

DRSP or DSG-only pills [33]

LNG-IUD [24]

One-rod LNG implant [32]

ENG implant [57]

EE + DSG [71]
Ferritin EE + DSG [71]
RBC EE + DSG or EE + GSD [26]

DRSP or DSG-only pills [33]

EE + DSG [71]
WBC EE + DSG or EE + GSD [26]

LNG-IUD [24]
Platelets EE + DSG or EE + GSD [26]
  1. ENG, etenogestrel; LNG, levonorgestrel; EE, ethinylestradiol; DSG, desogestrel; GSD, gestodene; DRSP, drospirenone.

In summary, current COC formulations have not been connected to liver enzyme elevations, despite early COC formulations being linked to frequently elevated serum enzyme levels. In people with hereditary types of bilirubin metabolism like Dubin Johnson syndrome, COCs can produce a modest suppression of bilirubin excretion, resulting in jaundice. The ability of COCs to cause aberrant test results with liver damage is more significant in specific cases often having a history of cholestasis of pregnancy (with jaundice and/or pruritus), and genetic variations in bile acid transporter genes (ABC B4, B11 and C2). The effects of HCs on the liver are reversible and most of liver tests could be reassessed 4 weeks after stopping HCs [59, 60].

Hemostasis parameters

The use of COCs has an impact on a number of hemostatic variables that are associated with thrombin formation and procoagulant (Table 6), anticoagulant, or fibrinolytic pathways (Table 7). Despite that their concentrations often stay within the reference range, changes in D-Dimer, antitrombin III, factor II activity and plasminogen levels (Tables 6 and 7) may have additive or antagonistic effects on the risk of venous thromboembolism (VTE) [72]. Activated protein C resistance (APCr) is a relatively new marker found to be important in the pathogenesis of VTE [73, 74]. Increased APCr is seen in COC users, and COCs containing DSG, gestodene (GSD), or DRSP exhibit a higher normalized APC sensitivity ratio (nAPCsr) comparable to those of heterozygous Factor V Leiden carriers [75, 76]. In COC users minimal increases of coagulation factors (such as prothrombin and factor VIII), small decreases of anticoagulant proteins (such as tissue factor pathway inhibitor [TFPI], protein S, protein C and antithrombin), and acquired APCr were reported [77, 78].

Table 6:

Effects of HC on markers of thrombin formation and anticoagulant proteins.

D-Dimer

(30–80 %)
EE + LNG, EE + DSG [79]

EE + LNG [80, 81]

EE + DRSP [82]
EE + DRSP, EE + LNG, E4 + DRSP [83]

E2V + DSG [81]

E2V + DSG, EE + LNG [84]

E2 + NOMAC, EE + LNG [85]


(25–75 %)
E4 + DRSP [82]

EE + NOMAC [80]
PF 1 + 2

(15–63 %)
EE + DRSP, EE + LNG, E4 + DRSP [83]

EE + LNG, EE + DSG [86]

EE + DRSP [82]

EE + LNG [80]
E2V + DSG, EE + LNG [81, 84]

E2 + NOMAC, EE + LNG [85]

E2 + NOMAC [80]

E4 + DRSP [82]
Antithrombin III EE + DRSP, EE + LNG, E4 + DRSP [83]

E2V + DSG, EE + LNG [81, 84]

E2 + NOMAC, EE + LNG [85]

E4 + DRSP [82]

E2 + NOMAC [80]


(5 %)
EE + DRSP [82]

EE + LNG [80]
Free protein S

(5–10 %)
E2 + NOMAC, EE + LNG [80, 85]
EE + DRSP, EE + LNG, E4 + DRSP [83]

LNG-IUD [87]


(12–50 %)
EE + LNG, EE + DSG, EE + DRSP, EE + CPA [87]
Protein S act E4 + DRSP [82]

E2 + NOMAC, EE + LNG [80]


(27 %)
EE + DRSP [82]
Protein C act

(3–11 %)
EE + LNG [85]

EE + DRSP [82]
EE + DRSP, EE + LNG, E4 + DRSP [83]

E2V + DSG, EE + LNG [81, 84]

E2 + NOMAC [85]

E4 + DRSP [82]
ETP-based APC-res.

(150–275 %)
EE + DRSP, EE + LNG, E4 + DRSP [83]

E2 + NOMAC, EE + LNG [85]
aPTT based APC-res.

(20–40 %)
E2 + NOMAC, EE + LNG [80]
E2V + DSG, EE + LNG [81, 84, 88]

E2 + NOMAC, EE + LNG [85]

EE + DRSP [82]


(8 %)
E4 + DRSP [82]
  1. LNG, levonorgestrel; EE, ethinylestradiol; E2, estradiol; DSG, desogestrel; DRSP, drospirenone; E4, estetrol; E2V, estradiol valerate; NOMAC, nomegestrol acetate.

Table 7:

Effects of HC on procoagulant and fibrinolytic factors.

Factor II act.

(4–10 %)
EE + LNG, EE + DSG [86]

E4 + DRSP, EE + DRSP [82]

E2 + NOMAC, EE + LNG [80]
EE + DRSP, EE + LNG, E4 + DRSP [83]

E2V + DSG, EE + LNG [84]

E2 + NOMAC, EE + LNG [85]
Factor VII act.

(12–32 %)
EE + LNG, EE + DSG [86]

EE + LNG [81]
EE + DRSP, EE + LNG, E4 + DRSP [83]

E2V + DSG [81]

E2V + DSG, EE + LNG [84]

E2 + NOMAC, EE + LNG [85]
Factor VIII act. EE + DRSP, EE + LNG, E4 + DRSP [83]

EE + LNG, EE + DSG [86]

E2V + DSG, EE + LNG [81, 84]

E2 + NOMAC, EE + LNG [80, 85]
Fibrinogen

(8–28 %)
EE + LNG [81]

EE + DRSP [82]
EE + DRSP, EE + LNG, E4 + DRSP [83]

E2V + DSG [81]

E2V + DSG, EE + LNG [84]

E4 + DRSP [82]

EE + DRSP, EE + LNG, E4 + DRSP [83]

EE + DRSP, EE + LNG, E4 + DRSP [83]
Plasminogen

(6–30 %)
EE + DRSP, EE + LNG, E4 + DRSP [83]

EE + LNG, EE + DSG [79]

E2 + NOMAC, EE + LNG [80]
TPA antigen

(25–50 %)
EE + DRSP, EE + LNG, E4 + DRSP [83]

EE + LNG, EE + DSG [79]
E4 + DRSP [82]


(48 %)
EE + DRSP [82]
PAI-1 antigen EE + DRSP, EE + LNG, E4 + DRSP [83]

E2V + DSG, EE + LNG [81]


(8–25 %)
EE + LNG, EE + DSG [79]

E2 + NOMAC, EE + LNG [80]
  1. LNG, levonorgestrel; EE, ethinylestradiol; E2, estradiol; DSG, desogestrel; DRSP, drospirenone; E4, estetrol; E2V, estradiol valerate; NOMAC, nomegestrol acetate.

In the seventies, the effects of estrogen on blood coagulation, fibrinolysis, and platelets were compared between COCs containing 50 and 30 µg of EE. With both preparations, higher levels of platelet activity, factor II, VII, VIII, IX, and X, fibrinogen, and soluble fibrin were seen, as well as lower levels of antithrombin (AT) and vessel wall fibrinolytic activator. The effects were EE dose dependent and less pronounced with EE concentrations with 30 µg in COC preparations [8992]. Yet, in a more recent meta-analysis the VTE risk remained higher with the so-called third generation COCs (EE + DSG, GSD, or norgestimate) compared to second generation COCs (EE + LNG or norethisterone) [93]. The difference in VTE risk for each COC could be attributed to a unique modulation of the procoagulant action of EE, exerted by the progestogens, as progestin-only contraceptives do not interfere with the synthesis of coagulation protein [94, 95].

EE combined with DSG, GSD or DRSP cause more alterations in coagulation markers and a greater observed VTE risk than EE/LNG [78, 85]. This is probably due to the less potent antagonistic interactions between the EE-induced alterations in hemostasis variables and DSG, GSD, and DRSP as compared to LNG [80]. COCs that contain E2 seem to have less of an impact on hemostasis parameters [94]. A recent study found that changes in hemostasis measures following six cycles of a natural estrogen (E4) with DRSP were either less pronounced or comparable to those seen with EE/LNG [83]. The observation of larger changes compared to EE/DRSP is consistent with the theory that the estrogenic component of COCs primarily mediates the effect on hemostasis measures.

Regardless of their limited effects on routinely used hemostasis parameters, high estrogenicity is associated with an increased risk of VTE [96]. A COC’s estrogenicity is calculated as the sum of its progesterone and estrogen contributions. The estrogenicity levels of either alone or combined hormonal contraceptives are depicted in Figure 2.

Figure 2: 
Estrogenicity levels of HCs (modified from [96]).
Figure 2:

Estrogenicity levels of HCs (modified from [96]).

The hemostatic measures during traditional COC administration and the values with continuous administration were examined in three RCTs [42, 43, 46]. The pro-coagulatory factors were increased, the coagulation inhibitors were decreased, and the fibrinolysis parameters were enhanced in both regimens. This might mean that there is no difference between the two regimens, but that both traditional and continuous use of OCs have increased coagulation activity.

In summary, the effects of HCs on parameters related to the coagulation system are heterogeneous. While increased APCr is seen in some COCs (containing DSG, GSD or DRSP) exhibiting a higher normalized APC sensitivity ratio (nAPCsr), most changes are within reference intervals and are generally not related with the composition of HC preparations. Although these changes in coagulation are non-explanatory for VTE risk related with HCs, it is still a clinically important issue and estrogen contraception is usually contraindicated in women with a personal history of VTE. The underlying mechanisms should also be further investigated.

Corticosteroids, androgens, estrogens and their binding proteins

The published effects of HC usage on the corticosteroids, androgens, estrogens and their binding proteins are listed in Table 8. Synthetic estrogen- and progesterone-like chemicals in the HCs work by inhibiting the pituitary’s ability to produce follicle stimulating hormone (FSH) and luteinizing hormone (LH). While the LH surge initiates mid-cycle ovulation, FSH promotes the development of folliculogenesis. After ovulation progesterone is secreted by the corpus luteum which inhibits endometrial proliferation, determines endometrial receptivity and gives negative feedback to the pituitary and hypothalamus.

Table 8:

Effects of HCs on the laboratory parameters related with corticosteroids, androgens and estrogens.

SHBG

(200–400 %)
E2V + DSG, EE + LNG [81, 84, 88]

E2 + NOMAC, EE + LNG [80, 85]

E2 + DRSP, EE + DRSP, EE + LNG [83]

EE + LNG, EE + DSG, EE + CPA, EE + DRSP, transtermal patch (EE + NGM), vaginal ring (EE + ENG) [97]

EE + DRSP [82]

E4 + DRSP,EE + LNG, EE + DRSP [29]

EE + LNG [98]

EE + DRSP, E4 + DRSP [30]

EE + CMA, EE + DSG [23]

EE + DNG [99]

EV + DNG, EE + DNG [100]

EE + CPA [101]
LNG-IUD [97]

E4 + DRSP [82]

DMPA, LNG-subdermal implant [102]

E4 + LNG [30]

DNG-only [100]
CBG

(40–170 %)
E2V + DSG, EE + LNG [81]

E4 + DRSP,EE + LNG, EE + DRSP [29]

EE + LNG [98]

EE + DRSP, E4 + DRSP, E4 + LNG [30]

EE + DSG [25]

EE + DNG, EV + DNG, DNG-only [103]
Total testosteronea

(21–37 %)
E4 + DRSP,EE + LNG, EE + DRSP [29]

EE + LNG [98]

EE + DNG [99]

E2V + DNG [100]

EE + CPA [101]
Free testosterone

(34–60 %)
E4 + DRSP,EE + LNG, EE + DRSP [29]

EE + LNG [98]

EE + CMA, EE + DSG [23]

EE + DNG [99]

EE + CPA [101]
Cortisol

(26–109 %)
E4 + DRSP,EE + LNG, EE + DRSP [29]

EE + LNG [98]

EE + DSG [25]

EE + DNG [103],
Free cortisol E4 + DRSP,EE + LNG, EE + DRSP [29]
DHEA-S

(5–10 %)
E4 + DRSP,EE + LNG, EE + DRSP [29]

EE + LNG [98]

EE + CMA, EE + DSG [23]

EE + DNG [103]

EE + CPA [101]
Androstenedione

(31–49 %)
E4 + DRSP,EE + LNG, EE + DRSP [29]

EE + CMA, EE + DSG [23]

EE + CPA [101]
FSH E4 + DRSP [29]

Subdermal implant (ENG) [31]

DNG-only [100]


(13–84 %)
EE + LNG, EE + DRSP [29]

EE + LNG [98]

EE + DSG [25]

EE + CMA, EE + DSG [23]

EV + DNG, EE + DNG [100]

EE + CPA [101]
LH E4 + DRSP [29]

Subdermal implant (ENG) [31]


(51–92 %)
EE + LNG, EE + DRSP [29]

EE + LNG [98]

EE + DSG [25]

EE + CMA, EE + DSG [23]

EE + CPA [101]
Progesterone

(6 %)
EE + LNG [98]


(60–96 %)
E4 + DRSP,EE + LNG, EE + DRSP [29]

EE + DSG [25]

EE + CMA, EE + DSG [23]
Estradiol Subdermal implant (ENG) [31]


(40–92 %)
E4 + DRSP,EE + LNG, EE + DRSP [29]

EE + LNG [98]

EE + DSG [25]

EE + CMA, EE + DSG [23]
Prolactin

(19–35 %)
E4 + DRSP, EE + LNG [29]

Subdermal implant (ENG) [31]
EE + DRSP [29]

EE + LNG [98]

EE + CMA, EE + DSG [23]


(15 %)
EE + DSG [25]
Anti-Müllerian hormone Vaginal ring [104]

EE + LNG, MPA + E2 inj.,DSG, DMPA, LNG-IUD [105]


(7–65 %)
COC, progestin only-pill, LNG-IUD [104]

COC, vaginal ring, LNG-IUD, implant, progestin-only pill [106]

COC, vaginal ring [107]

COC, vaginal ring, DMPA [108]
  1. aAnalyses might be affected by low quality testosterone immunoassay. ENG, etenogestrel; E4, estetrol; LNG, levonorgestrel; DMPA, depot medroxyprogesterone acetate; EE, ethinylestradiol; DSG, desogestrel; CMA, chlormadinone acetate; GSD, gestodene; DRSP, drospirenone; E2V, estradiol valerate; CPA, cyproterone acetate.

LH and FSH testing gives important information about gonadal function. Follicular development and ovulation are stimulated by LH together with FSH resulting proper growth of follicles. These hormones are mostly normally suppressed in women using COCs. HC may also suppress endogenous estradiol and/or progesterone levels. If the HC contain EE, the estradiol measurements are decreased as EE do not cross react in estradiol immunoassays and are not measured using estradiol LC-MS/MS assays. However, when HC contain E2, this exogenous estradiol is measured in the estradiol assays.

EE increases levels of SHBG [109]. As testosterone is bound to SHBG, and the testosterone production in theca cells, that is promoted by LH, is decreased by OCs [98, 110], the serum concentration of free testosterone is often described as being reduced by 20–40 % when low-dose HCs are used [111]. The lowering of free testosterone levels during conventional OC administration is quickly reversed over the 7-day hormone-free period [112, 113]. Free testosterone is said to be returned to baseline levels as a result of this gain in total testosterone and the attenuation of the OC-stimulated rise in SHBG during the pill-free week [99]. The adrenal glands are the primary source of the remaining amounts (25 %) of circulating testosterone, either directly or through conversion of precursors such androstenedione or dehydroepiandrosterone. Dehydroepiandrosterone sulfate (DHEAS) levels are moderately decreased while using OCs (and are partially restored during the hormone-free period), which may be due to a direct interaction between the contraceptive steroids and the production of adrenal steroid hormones [98, 99, 112, 114]. Recent studies have shown that COCs containing E4 have milder effects on SHBG, CBG, and gonadotropins (60–90 %) compared to those containing EE (200–400 %) [29, 30].

Anti-androgen progestins combined with EE are not only used for contraception but also for the treatment of hirsutism and polycystic ovary syndrome. Levels of free and total testosterone, androstenedione and DHEA-S as well as FSH and LH were found lower in women using COCs containing EE and anti-androgen progestin CPA [101, 115].

One of the drawbacks and shortcomings of testosterone immunoassays revealed that these methods are not accurate in the low testosterone concentrations range and are analytically influenced by high SHBG concentrations. The high SHBG concentration in women using HCs might therefore lead to falsely low testosterone concentrations measured using these immunoassays [116, 117]. Thus, the lower testosterone levels commonly seen in hormonal contraceptive usage, should be confirmed and studied by LC-MS/MS methods, which are not analytically influenced by SHBG concentrations, in future studies.

As estrogens regulate cortisol equilibrium by promoting hepatic CBG synthesis, both CBG and total cortisol levels rise when COCs are used [29, 85, 111]. Increased CBG levels lead to a new, altered CBG-cortisol equilibrium followed by concurrent increases in cortisol production. EE-based COCs have more significant impact on adrenal steroids and CBG levels (∼two-fold higher than baseline) whereas different E2/EV/E4 combinations seem to be of less significance [29, 81, 85].

Adrenal reactivity to adrenocorticotrophic hormone (ACTH) is similarly increased by COC usage due to an increase in CBG concentrations [118]. After exogenous ACTH administration (synacthen test) higher cortisol levels in serum, but not in saliva, were found in women using three months low-dose COC [119]. Moreover, COC use is also associated with inadequate suppression of cortisol by overnight-1 mg dexamethasone [120]. Recently, a two-day low dose dexamethasone suppression test was found to be a better screening test for hypercortisolism in women using HC [121]. Alternatively, the 1 mg dexamethasone suppression tests could be performed after a 1 month HC free period [121]. The free cortisol index or salivary cortisol which are said not to be affected by EE might be an option for hypercortisolism screening in women using HC [122].

Anti-Mullerian hormone (AMH), which is a widely used parameter for the assessment of ovarian reserve, was found to be decreased by long term (>6 months) HC usage in studies having a larger number of participants while smaller studies did not find any change [104, 105, 107, 108, 123, 124]. This decline in AMH levels returned to baseline levels after two months of HC discontinuation [124]. Although the exact reason why AMH levels were suppressed during HC treatment is currently not known, suppression of FSH and LH levels, and diminished formation of antral follicles by HC were suggested as a possible causative explanation [125].

In summary, HCs suppress LH and FSH and the endogenous estradiol production due to feedback on the pituitary gland and hypothalamus. Furthermore, lower levels of AMH during HC usage could reflect suppression of ovarian functions. HCs including EE have a major impact on SHBG and CBG concentrations. The impact varies and depend on the kind of progestin present in the COC, i.e. those having progestins with antiandrogenic qualities had the greatest impact, leading to increases between 200 and 400 %. Natural estrogens appear to have less effect on those hepatic proteins, i.e. an increase of only 60–90 %. All preparations of HCs have the ability to decrease androgen secretion since they inhibit the hypothalamus-pituitary-ovarian axis. Yet, the weaknesses of widely used immunoassay methods for total testosterone levels (and thereby the calculated free testosterone) should also be considered as an important analytical error source leading to falsely low androgen levels.

Thyroid hormones and related proteins

The published effects of HC usage on thyroid hormones and related proteins are listed in Table 9. It is widely accepted that thyroxine-binding globulin (TBG) concentration is increased during OC therapy [110, 126]. Initial studies suggest that estrogen can induce hepatic TBG protein synthesis [127]. Other studies showed that natural estrogen induces sialylation of TBG. Sialylation prolongs TBG half-life due to a decreased clearance rate resulting in higher serum TBG concentrations upon COC therapy [128]. The amount of COC induced TBG elevation is dependent on the prescribed type and concentration of the synthetic estrogens and progestogens [111]. Since TBG is the major binding protein for thyroid hormones, its elevation is directly related to a rise in total T3 and T4. This is the reason why nowadays most laboratories changed thyroid functions test from total T4 and T3 into fT4 and fT3 as these are not affected by COC use, despite transient changes (depending on COC type)_ during the first cycle of COC use [111]. The elevations in TBG, T3 and T4 are more prominent in dienogest-containing COC compared to LNG which can be explained by the lower estrogenic activity of LNG [99, 110, 111]. rT3 remains unaffected after LNG or DSG-containing COC administration [110]. TSH levels are also not affected, except following EE/E2V/DNG use which is suggested to result in higher TSH levels [111].

Table 9:

Effects of HCs on thyroid hormones and related proteins.

TBG

(30–75 %)
EE + DNG, EE + LNG, EE + EV + DNG [109]

EE + LNG,EE + DSG,EE + DRSP, EE + CPA [129]

EE + DSG [24]

EE + DNG [111]

E2 + NOMAC, EE + LNG [95]
LNG-IUD [129]
Total T3

(20–70 %)
EE + DNG, EE + LNG, EE + EV + DNG [109]

EE + DSG [24]

Skin patch (EE + NGM) [34]

EE + DNG [111]
Subdermal implant (ENG) [34]
fT3

(6 %)
EE + LNG [29]
EE + DNG, EE + LNG, EE + EV + DNG [109]

DMPA [130]

Subdermal implant (ENG) [28]

EE + DNG [111]

E4 + DRSP, EE + DRSP [29]
Total T4

(8–35 %)
EE + DNG, EE + LNG, EE + EV + DNG [109]

EE + DSG [24]

EE + DNG [111]
EE + DRSP, EE + CMA [131]

Skin patch (EE + NGM), subdermal implant (ENG) [34]
fT4

(6 %)
E4 + DRSP, EE + LNG, EE + DRSP [29]
EE + DNG, EE + E2V + DNG [109]

LNG-IUD, EE + LNG,EE + DSG,EE + DRSP, EE + CPA [129]

DMPA [130]

Subdermal implant (ENG) [28]

EE + DNG [111]

E2 + NOMAC, EE + LNG [95]
TSH

(25–40 %)
EE + LNG,EE + DSG,EE + DRSP, EE + CPA [129]
EE + DRSP, EE + CMA [131]

DMPA [130]

Subdermal implant (ENG) [28]

Skin patch (EE + NGM) [34]

E2 + NOMAC, EE + LNG [95]


(20 %)
Subdermal implant (ENG) [34]
  1. ENG, etenogestrel; E2, estradiol; LNG, levonorgestrel; DMPA, depot medroxyprogesterone acetate; EE, ethinylestradiol; DSG, desogestrel; CMA, chlormadinone acetate; DRSP, drospirenone; E2V, estradiol valerate; CPA, cyproterone acetate; NOMAC, nomegestrol acetate.

In addition to studies on the effects of COC therapy on thyroid parameters, other studies focused on the effect of different manners of administration of estrogens and progestogens on thyroid function parameters. Remarkably, transdermal administration of E2 did not have an effect on TBG concentration [130]. A more recent study showed that the use of a contraceptive (transdermal) patch that delivers EE and LNG results in a significant TBG increase. This rise was even more prominent compared to the elevation upon COC administration [132]. Another study showed that a single intramuscular progesterone administration has not been associated with elevation of TBG and T4 levels [129]. Usage of depot medroxyprogesterone acetate as injectable contraceptive caused a minor but statistically significant increase in fT4 levels, which was not observed in patients using copper IUD [131]. In accordance, administration of micronized progesterone increases fT4 concentration, which is still in the reference range, without affecting fT3 and TSH levels [133].

In conclusion, HC, depending on the type and the administration route, can lead to elevated TBG, T4 and T3 concentrations. It is therefore recommended not to measure total T4 and total T3 levels in women using HCs, but instead rely on fT4 and fT3 measurements which show at most minor changes during the usage of some of HCs.

Renin angiotensin system and renal function

The renin-angiotensin-aldosterone system is known to be affected by HC in two ways. The published data on the effects are shown in Table 10. The increased (95–375 %) formation of angiotensinogen is first and foremost highly stimulated by estrogens, which results in higher levels of angiotensin and aldosterone as well as sodium retention. Second, progesterone inhibits sodium retention by acting as a strong aldosterone antagonist on the mineralocorticoid receptor. Progestogens lacking anti-androgenic and anti-mineralocorticoid properties are unable to offset the sodium-retention impact of the EE component in COC. In turn, this could lead to increased fluid retention and the promotion of symptoms like edema and increased body weight [134, 135].

Table 10:

Effects of HCs on the parameters related with renal functions and renin-angiotensin-aldosterone system.

Creatinin Clearence

(20–30 %)
EE + CPA, EE + DSG, EE + GSD [136]
EE + LNG, EE + DSG [137]
Serum Na EE + DRSP, Subdermal implant (ENG) [138]

EE + CPA, EE + DSG, EE + GSD [136]
Serum K EE + DRSP, Subdermal implant (ENG) [138]

EE + CPA, EE + DSG, EE + GSD [136]
K excretion rate

(29–57 %)
EE + CPA, EE + DSG [136]
EE + GSD [136, 139]
Na excretion rate

(30 %)
EE + GSD [136]
EE + CPA, EE + DSG [136, 139]
Albumin excretion rate

(5–21 %)
EE + LNG [139, 140]
EE + CPA, EE + DSG, EE + GSD [136]

EE + LNG, EE + DSG [137]
Angiotensin II

(23–36 %)
EE + DNG, EE/E2V + DNG, EE + LNG [111]
Renin

(100 %)
DRSP-only [141]
GSD-only, DSG-only [141]

Subdermal implant (ENG) [138]


(40 %)
EE + DRSP [138]
Plasma renin activity

(25–245 %)
EE + DRSP [138, 139, 142]

DRSP-only, EE + DRSP [143]

DRSP-only [141]
EE + LNG [142]

EE + DSG [143]

GSD-only, DSG-only [141]

Subdermal implant (ENG) [138]
Aldosterone

(30–400 %)
EE + DRSP [138, 142]

DRSP-only, EE + DRSP, EE + DSG [143]

DRSP-only [141]
EE + LNG [139, 142]

GSD-only, DSG-only [141]

Subdermal implant (ENG) [138]

EE + DNG, EE/E2V + DNG, EE + LNG [111]
Angiotensinogen

(95–375 %)
EE + DRSP, EE + LNG [142]

EE + DRSP, EE + DSG [143]
  1. ENG, etenogestrel; GSD, gestodene; LNG, levonorgestrel; EE, ethinylestradiol; DSG, desogestrel; DRSP, drospirenone; E2V, estradiol valerate; CPA, cyproterone acetate.

Due to the significant hepatic effect of EE, oral contraceptives are also known to raise the liver derived plasma renin substrate angiotensinogen [144]. Angiotensin II (the renin-angiotensin system [RAS] effector substance), renin and aldosterone levels may also increase in conjunction in this way [145].

The impact of synthetic estrogens on RAS is also shown by exogenous [146] estrogen injections increasing plasma, hepatic, and renal concentrations of angiotensinogen, which has the ability to increase plasma concentrations of angiotensin II. This can be explained by the angiotensinogen gene having a promoter region responding to estrogen [147]. When taken as part of a COC, EE (15 µg/day) causes a rise in plasma angiotensinogen that is only marginally smaller than that observed during pregnancy [148]. Studies on exogenous estrogen administration have shown both stimulation [149, 150] and inhibition of the circulating RAS [151]. Studies evaluating the effects of OCs on aldosterone levels have also shown conflicting results [139, 145, 152, 153].

The generation of angiotensinogen in the liver will not be stimulated by the contraceptive patch because of the absence of first-pass metabolism [154]. Therefore, the transdermal application does not result in an increase in circulating RAS components [130, 146, 155] and similarly, the use of injectable contraceptives was not found to be linked to an increase in blood pressure [156].

The endogenous creatinine clearance was significantly raised in groups using COC. Also significantly higher rates of potassium salt excretion were found [136]. This study demonstrated that oral contraceptives could raise glomerular filtration rate and some types have a protein catabolic effect, in addition to their different effects on renal tubular function. Numerous short-term investigations and cross-sectional studies have revealed a relationship between HC usage and urine albumin loss with regard to the effects of HC on urinary albumin excretion [139, 140]. In contrast to women who did not use HC, women using HC had a 90 % higher risk of microalbuminuria (30–300 mg/day) [140]. These effects account for both normotensive and hypertensive women who used oral contraceptives [139]. A significant reduction on albumin excretion rates was found after discontinuation of HC usage [137].

In summary, angiotensinogen levels can be extremely (even 4-fold) elevated by HC use. The RAS system could also be affected depending on the composition of HCs. Because of pre-analytical effects and also analytical variations, aldosterone levels show significant variations independent from the compositions of OCs.

Also, measurement techniques in aldosterone and renin concentrations might cause inappropriate results [157159]. Therefore, the diagnosis of primary hyperaldosteronism in women using HCs could be interfered by false positive aldosterone renin ratios due to affected parameters of renin-angiotensin-aldosterone system. Despite insufficient evidence that using HC makes women more likely to develop renal illness, HCs may raise the risk of developing microalbuminuria regardless of blood pressure. The disturbances in RAS and renal functions due to HC usage are therefore advised to be re-evaluated after at least a 4 week treatment free period.

Vitamins and minerals

Studies have shown that the widespread use of oral contraceptives may have a negative impact on the body’s levels of several vital vitamins and minerals [160163]. The published effects of HC usage on vitamins and minerals are listed in Table 11. According to the World Health Organization (WHO), the impact of nutritional depletion brought on by using oral contraceptives is of high clinical relevance. Folate, vitamins B1, B2, B6, B12, C, and E, as well as the minerals magnesium, selenium, and zinc, may be affected by oral contraceptives. Additionally, they may lower levels of the amino acid tyrosine and the antioxidant Coenzyme Q10.

Table 11:

Effects of HCs on vitamin and mineral parameters.

Vitamin B12

(13–59 %)
EE + DSG, DMPA [164]

EE + DSG [165]

EE + DSG, EE + LNG [166]

EE + LNG, EE + DRSP [167]

EE + LNG, EE + DSG, EE + norgestimate [168]
Folate EE + DSG [165]

EE + DSG, EE + LNG [166]

EE + LNG, EE + DRSP [167]

EE + LNG, EE + DSG, EE + norgestimate [168]
Homocysteine EE + DSG [165]

EE + LNG, EE + DRSP [167]

EE + LNG [169]

EE + LNG, EE + DSG, EE + norgestimate [168]
Vitamin B6 EE + LNG, DMPA [170]


(25 %)
EE + DSG [165]
Vitamin B2 EE + LNG, DMPA [170]
Vitamin E

(24–29 %)
EE + LNG, vaginal ring (EE + ENG), skin patch (EE + Norelgestromin) [171]
Selenium EE + LNG [172]
Magnesium DMPA, Norethisterone injection [173]


(26 %)
EE + norgestrel [173]

EE + LNG [174]
Zinc EE + LNG [172]


(40–45 %)
EE + Norgestrel, DMPA, NET [175]
Coenzyme Q10

(33–56 %)
EE + LNG, vaginal ring (EE + ENG), Skin patch (EE + norelgestromin) [171]
25-hydroxy vitamin D

(20–41 %)
COC [176180]
EE + NETA, EE + LNG, LNG-only [181]
Vitamin DBP

(25–35 %)
HCs [179]

CHCs [180]
EE + NETA, LNG-only [181]


(19 %)
EE + LNG [181]
  1. ENG, etenogestrel; LNG, levonorgestrel; DMPA, depot medroxyprogesterone acetate; EE, ethinylestradiol; DSG, desogestrel; DRSP, drospirenone; NET, norethisterone; NETA, norethisterone acetate.

Vitamin B6, B12, or folate levels were found to be lower in OC users compared to controls [164166, 182]. According to a study by Green et al. [168], total vitamin B12 levels were 33 % lower in OC users than in non-users. Given that OCs affect the quantities of circulating proteins, it is probable that they change the transcobalamins (TCs), which are circulating B12 binding proteins. In the circulation, approximately 20 % of B12 is bound to TC2 and is available for tissues (holo-TC or active B12); 80 % is bound to TC1 (Haptocorrin) and is not available for tissues. Haptocorrin was found to be lower in OC users compared to nonusers, suggesting a decreased vitamin B12 binding ability rather than a functional deficiency [167, 183].

There was no difference between OC users and controls in plasma MMA and plasma total homocysteine (tHcy) suggesting that OC does not lead to a functional B12 deficiency [167]. It is still unclear whether the lower serum B12 levels associated with OC usage genuinely point to a biochemical B12 shortage, despite the fact that there is consistent data to support this association. The levels of active vitamin B12 during HC use should be further investigated. It is also unknown how people already at risk for B12 insufficiency, including vegetarians, may be impacted by a possible alteration in blood B12 binding ability brought on by OC usage.

The use of COCs has a detrimental effect on folate status by affecting the absorption pathway and thereby lowering serum levels. In comparison to a control group, women taking OCs had significantly decreased serum folate levels [184, 185]. Other investigations supported these findings revealing either a decrease in intestinal folate absorption in OC users [186] or increased folate metabolism and urine excretion [185]. While other studies conducted during the same time period found no difference in the folate status of OC users and controls, a recent meta-analysis that included case-control, cohort studies, and clinical trials from 1970 to 2013 came to the conclusion that OC use is, in fact, associated with lower serum folate status [187]. Confounding factors, such as dietary folate intake not being adjusted for, supplement use, smoking, and alcohol consumption may be the underlying explanation for this variation [188, 189]. The variable results even excluding dietary intake of folate [168, 190, 191] may be caused by inter-individual genetic variations in how folate is processed [188].

To investigate the effects of OCs of vitamin B6, one should take other metabolites of vitamin B6 into account because estrogens may affect tryptophan metabolism independently of vitamin B6 [188, 192]. They are urinary 4-pyridoxic acid (4-PA), plasma pyridoxal 5-phosphate (PLP), urinary B6, and erythrocyte aminotransferase or transaminase activity. Interestingly, PLP is considerably lower in OC users than non-users in both fasting and non-fasting plasma [165, 193], supporting the presence of lower vitamin B6 status in these women.

Moreover, COCs also cause a vitamin E deficiency. In fact, OC users experience more oxidative stress, particularly in terms of lipid peroxidation, which is linked to reduced amounts of vitamin E [194196] in the blood. The injection of contraceptive drugs dramatically decreased plasma levels of tocopherol and increased dietary needs for vitamin E [197], according to preclinical research in rats. In agreement with these findings, a clinical study found that COCs also lower plasma tocopherol levels in healthy women [198]. OC users have also greater blood-clotting activity, which may be related to the participants’ lower levels of tocopherol [199].

COC-based treatments, especially containing estrogens, have an impact on vitamin C status as indicated by lower levels of ascorbate in plasma leukocytes, platelets, and whole blood entities due to the increased rate of vitamin C metabolism [200202]. This effect is particularly amplified in women who have a disease of malabsorption, a poor diet, bad lifestyle, or both. In agreement with this, research has shown that a sufficient diet rich in ascorbic acid can prevent depletion brought on by OC usage over a seven-year period [203].

The usage of HC has been linked to statistically higher total 25-hydroxyvitamin D (25(OH)D) levels in adults and adolescents [176180] which might be due to higher levels of vitamin D-binding protein (VDBP) [179, 180, 204, 205]. HC use did not change free 25(OH)D (if possible to measure accurately). Vitamin D status of HC users may therefore be overestimated [205]. As a result, in a population that uses HC, free or bioavailable 25(OH)D might be a more helpful indicator of vitamin D status in theory. VDBP-bound 25(OH)D can however be used by several tissues, including the parathyroid gland, which may change the relevance of free 25(OH)D levels [206]. However, as the majority of immunoassays produce 25(OH)D results that could be inaccurate due to being affected by VDBP concentration, further studies should be performed using LC-MS/MS methods before any conclusions can be drawn [207, 208].

Women using HC usually show altered mineral status, particularly at the levels of magnesium, zinc, and selenium [198]. OC users have lower serum magnesium levels than either noncontraceptive-users or women using alternative methods of contraception [160, 173, 209, 210]. In early studies, women using OCs had lower zinc levels than non-users [211, 212]. These findings were supported by other studies [172, 175, 213, 214], which suggested that reduced serum zinc levels might be a reflection of a decline in the status of zinc in tissues as a result of changes in zinc absorption, excretion, or tissue turnover. Numerous studies have shown that OCs may hinder the absorption of selenium and result in insufficiency. According to a clinical investigation in 200 young women, taking low-dose OCs for at least three months causes a statistically significant drop in mean serum selenium levels when compared to control subjects [172, 215].

In summary, HCs can affect blood levels of vitamins or minerals but probably without affecting their physiological functions. When assessing vitamin state in women using HCs, determination of functional markers (like homocysteine, and methylmalonic acid) could be beneficial to determine whether there is an actual vitamin deficiency or not. However, women who get HCs are at risk for a number of adverse effects linked to deficiency in a number of vitamins and minerals, including zinc, magnesium, selenium, vitamins B, C, and E which may request further investigations. 25OH Vitamin D levels may be increased in women using HC due to increased VDBP concentrations, however similar to testosterone, analytical errors may lead to falsely low 25OH Vitamin D levels.

Conclusions and recommendations

In this review, the impact of currently used HCs on clinical chemistry test results was summarized. HCs were defined as all hormonal contraceptive types and combinations. The effects of HCs differ by type of contraceptives, administration route and duration. The results of studies that included the definition of the composition of HC (76 studies) were summarized in tables. Unfortunately, a lot of studies provided no information about the HC composition used by their patient groups, consequently these studies were not included in the tables of this review. In addition, the number of publications about newer (e.g., those containing naturel estrogens) and non-oral HCs (vaginal ring, IUD, subcutaneous, transdermal and parenteral) and their effects on biochemical test results are limited. Despite the scarce information, the data about binding proteins suggest that natural estrogens have less impact than EE.

Even though the changes shown in the tables were statistically significant, most of them were rather small and concentrations therefore mostly did not end up outside the reference interval. Although HCs itself did not interfere the laboratory analyses as an analytical bias, increased binding protein concentrations like SHBG or VDBP did analytically influence the determination of the total levels (like testosterone and 25(OH)D) leading to mostly falsely low total hormone concentrations using several immunoassays. Thus, we suggest to measure these total hormones using more reliable methods like LC-MS/MS in order not to be analytically influenced by the concentration of the binding proteins. This phenomenon is especially important as HCs strongly affect the synthesis of transport or binding proteins produced in the liver (like TBG, SHBG, CBG and VDBP). In addition to the analytical consequences this might have, the hormones bound to these proteins (T4, T3, testosterone, cortisol and 25(OH)D) can show concentrations that need to be interpreted with care.

An important question is: what to do with women on HCs and affected clinical chemistry tests? One possibility is stopping with the HC and performing the tests again later. This possibility leads to the next question: if a woman would stop, after what time frame can she be tested again? This is especially important for dynamic endocrine tests as hormones like cortisol are heavily influenced. Although there are not enough data available for a firm conclusion, based on the half-life of measured analytes (i.e., for SHBG, TBG and CBG biological half-life is about 1 week) it might be suggested discontinuation of HC for at least 1 month could be enough for measuring clinical chemistry laboratory parameters reliably. Future studies need to focus on when levels return to baseline after discontinuation of HCs. Discontinuation of HC, however, has a drawback on contraception and physicians and patients should be well aware of this. An alternative for stopping HC could theoretically be establishing specific reference intervals for women using HCs. However, the different effects between HC types, dosages and in-cycle dose on clinical chemistry parameters make it impossible to set up specific reference intervals for HC users.

A third option for some of these tests, are free hormone measurements, such as 24 h urine cortisol or late night salivary cortisol instead of a total serum cortisol after dexamethasone or to use functional markers (i.e., tHcy and MMA) instead of vitamins and minerals to understand the existence of an actual deficiency.

In conclusion, many clinical chemistry tests show small changes upon HC use. We found that only some of these tests require special attention when measured in women using HCs: the binding proteins TBG, SHBG, CBG and VDBP and total measurements of the hormones binding to these proteins; the RAS system, aldosterone, renin and angiotensinogen; AMH; and some vitamins and minerals. Natural estrogens seem to have less effects on these clinical chemistry tests yet effects of non-oral HCs need to be studied. Alternative measurements such as free hormones or functional markers are preferred for optimal interpretation, otherwise HCs should be stopped for at least one month while taking care for anticonception in another way.


Corresponding author: Annemieke C. Heijboer, Department of Laboratory Medicine, Amsterdam UMC Location University of Amsterdam, Endocrine Laboratory, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam, The Netherlands; Department of Laboratory Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands; and Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands, E-mail:

  1. Research funding: None declared.

  2. Author contributions: The authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Competing interests: The authors state no conflict of interest.

  4. Informed consent: Not applicable.

  5. Research ethics: Not applicable.

  6. Data availability: Not applicable.

References

1. Oral contraceptives: the liberator. Economics. 1999.Search in Google Scholar

2. Junod, SW, Marks, L. Women’s trials: the approval of the first oral contraceptive pill in the United States and Great Britain. J Hist Med Allied Sci 2002;57:117–60. https://doi.org/10.1093/jhmas/57.2.117.Search in Google Scholar PubMed

3. Darney, PD. Family planning and the future. Am J Obstet Gynecol 2011;205(4 Suppl):S26–8. https://doi.org/10.1016/j.ajog.2011.04.032.Search in Google Scholar PubMed

4. United Nations DoEaSA, Population Division. Contraceptive use by method 2019: data booklet (ST/ESA/SER.A/435) 2019.Search in Google Scholar

5. Hall, KS, Trussell, J. Types of combined oral contraceptives used by US women. Contraception 2012;86:659–65. https://doi.org/10.1016/j.contraception.2012.05.017.Search in Google Scholar PubMed PubMed Central

6. Miale, JB, Kent, JW. The effects of oral contraceptives on the results of laboratory tests. Am J Obstet Gynecol 1974;120:264–72. https://doi.org/10.1016/0002-9378(74)90374-3.Search in Google Scholar PubMed

7. Briggs, MH. Human metabolism and steroid contraceptives. Part 2. Aust Fam Physician 1977:23–8.Search in Google Scholar

8. Briggs, MH. Human metabolism and steroid contraceptives. Part 1. Aust Fam Physician 1977:12–5.Search in Google Scholar

9. Fotherby, K. Interactions with oral contraceptives. Am J Obstet Gynecol 1990;163:2153–9. https://doi.org/10.1016/0002-9378(90)90556-m.Search in Google Scholar PubMed

10. Hoy, SM, Scott, LJ. Estradiol valerate/dienogest: in oral contraception. Drugs 2009;69:1635–46. https://doi.org/10.2165/11202820-000000000-00000.Search in Google Scholar PubMed

11. Stanczyk, FZ. Pharmacokinetics and potency of progestins used for hormone replacement therapy and contraception. Rev Endocr Metab Disord 2002;3:211–24. https://doi.org/10.1023/a:1020072325818.10.1023/A:1020072325818Search in Google Scholar PubMed

12. Kumar, N, Koide, SS, Tsong, Y, Sundaram, K. Nestorone: a progestin with a unique pharmacological profile. Steroids 2000;65:629–36. https://doi.org/10.1016/s0039-128x(00)00119-7.Search in Google Scholar PubMed

13. Sitruk-Ware, R, Small, M, Kumar, N, Tsong, YY, Sundaram, K, Jackanicz, T. Nestorone: clinical applications for contraception and HRT. Steroids 2003;68:907–13. https://doi.org/10.1016/s0039-128x(03)00140-5.Search in Google Scholar PubMed

14. Schindler, AE, Campagnoli, C, Druckmann, R, Huber, J, Pasqualini, JR, Schweppe, KW, et al.. Classification and pharmacology of progestins. Maturitas 2008;61:171–80. https://doi.org/10.1016/j.maturitas.2003.09.014.Search in Google Scholar PubMed

15. Phillips, A, Hahn, DW, McGuire, JL. Preclinical evaluation of norgestimate, a progestin with minimal androgenic activity. Am J Obstet Gynecol 1992;167:1191–6. https://doi.org/10.1016/s0002-9378(12)90410-x.Search in Google Scholar PubMed

16. Wiegratz, I, Stahlberg, S, Manthey, T, Sanger, N, Mittmann, K, Palombo-Kinne, E, et al.. Effects of combined oral contraceptive ethinylestradiol (30 microg) and dienogest (2 mg) on carbohydrate metabolism during 1 year of conventional or extended-cycle use. Horm Metab Res 2010;42:358–63. https://doi.org/10.1055/s-0030-1248263.Search in Google Scholar PubMed

17. Newton, JR. Classification and comparison of oral contraceptives containing new generation progestogens. Hum Reprod Update 1995;1:231–63. https://doi.org/10.1093/humupd/1.3.231.Search in Google Scholar PubMed

18. Berenson, AB, Rahman, M, Wilkinson, G. Effect of injectable and oral contraceptives on serum lipids. Obstet Gynecol 2009;114:786–94. https://doi.org/10.1097/aog.0b013e3181b76bea.Search in Google Scholar

19. Godsland, IF, Crook, D, Simpson, R, Proudler, T, Felton, C, Lees, B, et al.. The effects of different formulations of oral contraceptive agents on lipid and carbohydrate metabolism. N Engl J Med 1990;323:1375–81. https://doi.org/10.1056/nejm199011153232003.Search in Google Scholar PubMed

20. Kakis, G, Powell, M, Marshall, A, Woutersz, TB, Steiner, G. A two-year clinical study of the effects of two triphasic oral contraceptives on plasma lipids. Int J Fertil Menopausal Stud 1994;39:283–91.Search in Google Scholar

21. Sondheimer, SJ. Update on the metabolic effects of steroidal contraceptives. Endocrinol Metab Clin N Am 1991;20:911–23. https://doi.org/10.1016/s0889-8529(18)30250-0.Search in Google Scholar

22. Teichmann, A. Metabolic profile of six oral contraceptives containing norgestimate, gestodene, and desogestrel. Int J Fertil Menopausal Stud 1995;40(Suppl 2):98–104.Search in Google Scholar

23. Winkler, UH, Sudik, R. The effects of two monophasic oral contraceptives containing 30 mcg of ethinyl estradiol and either 2 mg of chlormadinone acetate or 0.15 mg of desogestrel on lipid, hormone and metabolic parameters. Contraception 2009;79:15–23. https://doi.org/10.1016/j.contraception.2008.08.011.Search in Google Scholar PubMed

24. Morin-Papunen, L, Martikainen, H, McCarthy, MI, Franks, S, Sovio, U, Hartikainen, AL, et al.. Comparison of metabolic and inflammatory outcomes in women who used oral contraceptives and the levonorgestrel-releasing intrauterine device in a general population. Am J Obstet Gynecol 2008;199:529 e1–10. https://doi.org/10.1016/j.ajog.2008.04.013.Search in Google Scholar PubMed

25. Crook, D. Multicenter study of endocrine function and plasma lipids and lipoproteins in women using oral contraceptives containing desogestrel progestin. UK Desogen Study Group. Contraception 1997;55:219–24. https://doi.org/10.1016/s0010-7824(97)00010-3.Search in Google Scholar PubMed

26. Cauci, S, Di Santolo, M, Culhane, JF, Stel, G, Gonano, F, Guaschino, S. Effects of third-generation oral contraceptives on high-sensitivity C-reactive protein and homocysteine in young women. Obstet Gynecol 2008;111:857–64. https://doi.org/10.1097/aog.0b013e31816a2476.Search in Google Scholar PubMed

27. Winkler, UH, Rohm, P, Hoschen, K. An open-label, comparative study of the effects of a dose-reduced oral contraceptive containing 0.02 mg ethinylestradiol/2 mg chlormadinone acetate on hemostatic parameters and lipid and carbohydrate metabolism variables. Contraception 2010;81:391–400. https://doi.org/10.1016/j.contraception.2009.12.005.Search in Google Scholar PubMed

28. Barreiros, FA, Guazzelli, CA, Barbosa, R, Torloni, MR, Barbieri, M, Araujo, FF. Extended regimens of the combined contraceptive vaginal ring containing etonogestrel and ethinyl estradiol: effects on lipid metabolism. Contraception 2011;84:155–9. https://doi.org/10.1016/j.contraception.2010.11.002.Search in Google Scholar PubMed

29. Klipping, C, Duijkers, I, Mawet, M, Maillard, C, Bastidas, A, Jost, M, et al.. Endocrine and metabolic effects of an oral contraceptive containing estetrol and drospirenone. Contraception 2021;103:213–21. https://doi.org/10.1016/j.contraception.2021.01.001.Search in Google Scholar PubMed

30. Mawet, M, Maillard, C, Klipping, C, Zimmerman, Y, Foidart, JM, Coelingh Bennink, HJ. Unique effects on hepatic function, lipid metabolism, bone and growth endocrine parameters of estetrol in combined oral contraceptives. Eur J Contracept Reprod Health Care 2015;20:463–75. https://doi.org/10.3109/13625187.2015.1068934.Search in Google Scholar PubMed PubMed Central

31. Inal, MM, Yildirim, Y, Ertopcu, K, Avci, ME, Ozelmas, I, Tinar, S. Effect of the subdermal contraceptive etonogestrel implant (Implanon) on biochemical and hormonal parameters (three years follow-up). Eur J Contracept Reprod Health Care 2008;13:238–42. https://doi.org/10.1080/13625180802075315.Search in Google Scholar PubMed

32. Gunardi, ER, Surya, R, Syafitri, I, Pasidri, Y. Impact of one-rod levonorgestrel implant on the blood chemistry profile. Sci Rep 2021;11:20141. https://doi.org/10.1038/s41598-021-99801-z.Search in Google Scholar PubMed PubMed Central

33. Palacios, S, Colli, E, Regidor, PA. Metabolic and laboratory effects of a progestin-only pill containing drospirenone 4 mg in comparison to desogestrel 75 microg: a double-blind, double-dummy, prospective, randomised study. Eur J Contracept Reprod Health Care 2021;26:454–61. https://doi.org/10.1080/13625187.2021.1957094.Search in Google Scholar PubMed

34. Bender, NM, Segall-Gutierrez, P, Najera, SO, Stanczyk, FZ, Montoro, M, Mishell, DRJr. Effects of progestin-only long-acting contraception on metabolic markers in obese women. Contraception 2013;88:418–25. https://doi.org/10.1016/j.contraception.2012.12.007.Search in Google Scholar PubMed

35. Hernandez-Juarez, J, Garcia-Latorre, EA, Moreno-Hernandez, M, Moran-Perez, JF, Rodriguez-Escobedo, MA, Cogque-Hernandez, G, et al.. Metabolic effects of the contraceptive skin patch and subdermal contraceptive implant in Mexican women: a prospective study. Reprod Health 2014;11:33. https://doi.org/10.1186/1742-4755-11-33.Search in Google Scholar PubMed PubMed Central

36. Frempong, BA, Ricks, M, Sen, S, Sumner, AE. Effect of low-dose oral contraceptives on metabolic risk factors in African-American women. J Clin Endocrinol Metab 2008;93:2097–103. https://doi.org/10.1210/jc.2007-2599.Search in Google Scholar PubMed PubMed Central

37. Elkind-Hirsch, KE, Darensbourg, C, Ogden, B, Ogden, LF, Hindelang, P. Contraceptive vaginal ring use for women has less adverse metabolic effects than an oral contraceptive. Contraception 2007;76:348–56. https://doi.org/10.1016/j.contraception.2007.08.001.Search in Google Scholar PubMed

38. Guazzelli, CA, de Queiroz, FT, Barbieri, M, Barreiros, FA, Torloni, MR, Araujo, FF. Metabolic effects of contraceptive implants in adolescents. Contraception 2011;84:409–12. https://doi.org/10.1016/j.contraception.2011.02.006.Search in Google Scholar PubMed

39. Haarala, A, Eklund, C, Pessi, T, Lehtimaki, T, Huupponen, R, Jula, A, et al.. Use of combined oral contraceptives alters metabolic determinants and genetic regulation of C-reactive protein. The Cardiovascular Risk in Young Finns Study. Scand J Clin Lab Invest 2009;69:168–74. https://doi.org/10.1080/00365510802449642.Search in Google Scholar PubMed

40. Wiegratz, I, Stahlberg, S, Manthey, T, Sanger, N, Mittmann, K, Palombo-Kinne, E, et al.. Effects of an oral contraceptive containing 30 mcg ethinyl estradiol and 2 mg dienogest on lipid metabolism during 1 year of conventional or extended-cycle use. Contraception 2010;81:57–61. https://doi.org/10.1016/j.contraception.2009.07.011.Search in Google Scholar PubMed

41. Anderson, FD, Hait, H. A multicenter, randomized study of an extended cycle oral contraceptive. Contraception 2003;68:89–96. https://doi.org/10.1016/s0010-7824(03)00141-0.Search in Google Scholar PubMed

42. Cachrimanidou, AC, Hellberg, D, Nilsson, S, von Schoulz, B, Crona, N, Siegbahn, A. Hemostasis profile and lipid metabolism with long-interval use of a desogestrel-containing oral contraceptive. Contraception 1994;50:153–65. https://doi.org/10.1016/0010-7824(94)90051-5.Search in Google Scholar PubMed

43. Machado, RB, Fabrini, P, Cruz, AM, Maia, E, da Cunha Bastos, A. Clinical and metabolic aspects of the continuous use of a contraceptive association of ethinyl estradiol (30 microg) and gestodene (75 microg). Contraception 2004;70:365–70. https://doi.org/10.1016/j.contraception.2004.06.001.Search in Google Scholar PubMed

44. Cagnacci, A. Hormonal contraception: venous and arterial disease. Eur J Contracept Reprod Health Care 2017;22:191–9. https://doi.org/10.1080/13625187.2017.1305349.Search in Google Scholar PubMed

45. Sitruk-Ware, R, Nath, A. Characteristics and metabolic effects of estrogen and progestins contained in oral contraceptive pills. Best Pract Res Clin Endocrinol Metabol 2013;27:13–24. https://doi.org/10.1016/j.beem.2012.09.004.Search in Google Scholar PubMed

46. Wiegratz, I, Stahlberg, S, Manthey, T, Sanger, N, Mittmann, K, Lange, E, et al.. Effects of conventional or extended-cycle regimen of an oral contraceptive containing 30 mcg ethinylestradiol and 2 mg dienogest on various hemostasis parameters. Contraception 2008;78:384–91. https://doi.org/10.1016/j.contraception.2008.06.015.Search in Google Scholar PubMed

47. Balogh, A, Kauf, E, Vollanth, R, Graser, G, Klinger, G, Oettel, M. Effects of two oral contraceptives on plasma levels of insulin-like growth factor I (IGF-I) and growth hormone (hGH). Contraception 2000;62:259–69. https://doi.org/10.1016/s0010-7824(00)00176-1.Search in Google Scholar PubMed

48. Grandi, G, Napolitano, A, Cagnacci, A. Metabolic impact of combined hormonal contraceptives containing estradiol. Expet Opin Drug Metabol Toxicol 2016;12:779–87. https://doi.org/10.1080/17425255.2016.1190832.Search in Google Scholar PubMed

49. Lopez, LM, Grimes, DA, Schulz, KF. Steroidal contraceptives: effect on carbohydrate metabolism in women without diabetes mellitus. Cochrane Database Syst Rev 2014;4:CD006133. https://doi.org/10.1002/14651858.cd006133.pub5.Search in Google Scholar

50. Weissberger, AJ, Ho, KK, Lazarus, L. Contrasting effects of oral and transdermal routes of estrogen replacement therapy on 24-hour growth hormone (GH) secretion, insulin-like growth factor I, and GH-binding protein in postmenopausal women. J Clin Endocrinol Metab 1991;72:374–81. https://doi.org/10.1210/jcem-72-2-374.Search in Google Scholar PubMed

51. Soria-Jasso, LE, Carino-Cortes, R, Munoz-Perez, VM, Perez-Hernandez, E, Perez-Hernandez, N, Fernandez-Martinez, E. Beneficial and deleterious effects of female sex hormones, oral contraceptives, and phytoestrogens by immunomodulation on the liver. Int J Mol Sci 2019;20:4694. https://doi.org/10.3390/ijms20194694.Search in Google Scholar PubMed PubMed Central

52. Czekaj, P, Nowaczyk-Dura, G, Ciszkowa, V. Effects of ethinylestradiol-norethisterone acetate combinations on the ultrastructure of liver, kidney, ovary and endometrium cells. Folia Biol (Praha) 1998;44:207–11.Search in Google Scholar

53. Barbosa, I, Coutinho, E, Athayde, C, Ladipo, O, Olsson, SE, Ulmsten, U. The effects of nomegestrol acetate subdermal implant (Uniplant) on carbohydrate metabolism, serum lipoproteins and on hepatic function in women. Contraception 1995;52:111–4. https://doi.org/10.1016/s0010-7824(95)00144-1.Search in Google Scholar PubMed

54. Paseka, J, Unzeitig, V, Cibula, D, Chroust, K. Liver function tests during administration of triphasic contraceptives containing Norgestimate. Ceska Gynekol 2000;65:420–4.Search in Google Scholar

55. Tagy, AH, Saker, ME, Moussa, AA, Kolgah, A. The effect of low-dose combined oral contraceptive pills versus injectable contraceptive (Depot Provera) on liver function tests of women with compensated bilharzial liver fibrosis. Contraception 2001;64:173–6. https://doi.org/10.1016/s0010-7824(01)00248-7.Search in Google Scholar PubMed

56. Kowalska, K, Sciskalska, M, Bizon, A, Sliwinska-Mosson, M, Milnerowicz, H. Influence of oral contraceptives on lipid profile and paraoxonase and commonly hepatic enzymes activities. J Clin Lab Anal 2018;32:e22194. https://doi.org/10.1002/jcla.22194.Search in Google Scholar PubMed PubMed Central

57. Dilbaz, B, Ozdegirmenci, O, Caliskan, E, Dilbaz, S, Haberal, A. Effect of etonogestrel implant on serum lipids, liver function tests and hemoglobin levels. Contraception 2010;81:510–4. https://doi.org/10.1016/j.contraception.2010.01.014.Search in Google Scholar PubMed

58. Lindberg, MC. Hepatobiliary complications of oral contraceptives. J Gen Intern Med 1992;7:199–209. https://doi.org/10.1007/bf02598014.Search in Google Scholar

59. Gijbels, E, Vinken, M. Mechanisms of drug-induced cholestasis. Methods Mol Biol 2019;1981:1–14. https://doi.org/10.1007/978-1-4939-9420-5_1.Search in Google Scholar PubMed

60. Hardikar, W, Kansal, S, Oude Elferink, RP, Angus, P. Intrahepatic cholestasis of pregnancy: when should you look further? World J Gastroenterol 2009;15:1126–9. https://doi.org/10.3748/wjg.15.1126.Search in Google Scholar PubMed PubMed Central

61. Huang, L, Smit, JW, Meijer, DK, Vore, M. Mrp2 is essential for estradiol-17beta(beta-D-glucuronide)-induced cholestasis in rats. Hepatology 2000;32:66–72. https://doi.org/10.1053/jhep.2000.8263.Search in Google Scholar PubMed

62. Koh, K, Kathirvel, R, Mathur, M. Rare case of obstetric cholestasis presenting in the first trimester following in vitro fertilisation. BMJ Case Rep 2021;14:e244254. https://doi.org/10.1136/bcr-2021-244254.Search in Google Scholar PubMed PubMed Central

63. Sabel, GH, Kirk, RF. Biliary stasis after mestranol-norethindrone ingestion. Report of a case. Obstet Gynecol 1968;31:375–7. https://doi.org/10.1097/00006250-196803000-00013.Search in Google Scholar PubMed

64. Zaki, K, Rizk, M, Kira, L, Nour, H, Guirguis, R. Studies on the effects of ethinyl estradiol and norethisterone acetate on the adrenal cortex and some other tissues in the rat. Endokrinologie 1979;73:66–76.Search in Google Scholar

65. Abu-Hayyeh, S, Papacleovoulou, G, Lovgren-Sandblom, A, Tahir, M, Oduwole, O, Jamaludin, NA, et al.. Intrahepatic cholestasis of pregnancy levels of sulfated progesterone metabolites inhibit farnesoid X receptor resulting in a cholestatic phenotype. Hepatology 2013;57:716–26. https://doi.org/10.1002/hep.26055.Search in Google Scholar PubMed PubMed Central

66. Smith, DD, Rood, KM. Intrahepatic cholestasis of pregnancy. Clin Obstet Gynecol 2020;63:134–51. https://doi.org/10.1097/grf.0000000000000495.Search in Google Scholar

67. Moussaoui, S, Abdelwahed, M, Ben Chaaben, N, Bellalah, A, Ben Fadhel, N, Guediche, A, et al.. Norethisterone-induced cholestasis: a case report. Clin Case Rep 2022;10:e05522. https://doi.org/10.1002/ccr3.5522.Search in Google Scholar PubMed PubMed Central

68. Hee, L, Kettner, LO, Vejtorp, M. Continuous use of oral contraceptives: an overview of effects and side-effects. Acta Obstet Gynecol Scand 2013;92:125–36. https://doi.org/10.1111/aogs.12036.Search in Google Scholar PubMed

69. Burton, JL. Effect of oral contraceptives on haemoglobin, packed-cell volume, serum-iron, and total iron-binding capacity in healthy women. Lancet 1967;1:978–80. https://doi.org/10.1016/s0140-6736(67)92359-8.Search in Google Scholar PubMed

70. Miller, L, Hughes, JP. Continuous combination oral contraceptive pills to eliminate withdrawal bleeding: a randomized trial. Obstet Gynecol 2003;101:653–61. https://doi.org/10.1016/s0029-7844(03)00014-0.Search in Google Scholar PubMed

71. Larsson, G, Milsom, I, Lindstedt, G, Rybo, G. The influence of a low-dose combined oral contraceptive on menstrual blood loss and iron status. Contraception 1992;46:327–34. https://doi.org/10.1016/0010-7824(92)90095-b.Search in Google Scholar PubMed

72. Martinelli, I, Bucciarelli, P, Mannucci, PM. Thrombotic risk factors: basic pathophysiology. Crit Care Med 2010;38(2 Suppl):S3–9. https://doi.org/10.1097/ccm.0b013e3181c9cbd9.Search in Google Scholar PubMed

73. Dahlback, B, Carlsson, M, Svensson, PJ. Familial thrombophilia due to a previously unrecognized mechanism characterized by poor anticoagulant response to activated protein C: prediction of a cofactor to activated protein C. Proc Natl Acad Sci U S A 1993;90:1004–8. https://doi.org/10.1073/pnas.90.3.1004.Search in Google Scholar PubMed PubMed Central

74. Wu, O, Robertson, L, Langhorne, P, Twaddle, S, Lowe, GD, Clark, P, et al.. Oral contraceptives, hormone replacement therapy, thrombophilias and risk of venous thromboembolism: a systematic review. The Thrombosis: Risk and Economic Assessment of Thrombophilia Screening (TREATS) Study. Thromb Haemostasis 2005;94:17–25. https://doi.org/10.1160/th04-11-0759.Search in Google Scholar

75. Lidegaard, O, Nielsen, LH, Skovlund, CW, Skjeldestad, FE, Lokkegaard, E. Risk of venous thromboembolism from use of oral contraceptives containing different progestogens and oestrogen doses: Danish cohort study, 2001–9. BMJ 2011;343:d6423. https://doi.org/10.1136/bmj.d6423.Search in Google Scholar PubMed PubMed Central

76. Tchaikovski, SN, Rosing, J. Mechanisms of estrogen-induced venous thromboembolism. Thromb Res 2010;126:5–11. https://doi.org/10.1016/j.thromres.2010.01.045.Search in Google Scholar PubMed

77. Westhoff, CL, Pike, MC, Cremers, S, Eisenberger, A, Thomassen, S, Rosing, J. Endogenous thrombin potential changes during the first cycle of oral contraceptive use. Contraception 2017;95:456–63. https://doi.org/10.1016/j.contraception.2017.01.001.Search in Google Scholar PubMed PubMed Central

78. Farris, M, Bastianelli, C, Rosato, E, Brosens, I, Benagiano, G. Pharmacodynamics of combined estrogen-progestin oral contraceptives: 2. Effects on hemostasis. Expet Rev Clin Pharmacol 2017;10:1129–44. https://doi.org/10.1080/17512433.2017.1356718.Search in Google Scholar PubMed

79. Meijers, JC, Middeldorp, S, Tekelenburg, W, van den Ende, AE, Tans, G, Prins, MH, et al.. Increased fibrinolytic activity during use of oral contraceptives is counteracted by an enhanced factor XI-independent down regulation of fibrinolysis: a randomized cross-over study of two low-dose oral contraceptives. Thromb Haemostasis 2000;84:9–14. https://doi.org/10.1055/s-0037-1613959.Search in Google Scholar

80. Gaussem, P, Alhenc-Gelas, M, Thomas, JL, Bachelot-Loza, C, Remones, V, Ali, FD, et al.. Haemostatic effects of a new combined oral contraceptive, nomegestrol acetate/17beta-estradiol, compared with those of levonorgestrel/ethinyl estradiol. A double-blind, randomised study. Thromb Haemostasis 2011;105:560–7. https://doi.org/10.1160/th10-05-0327.Search in Google Scholar

81. Junge, W, Mellinger, U, Parke, S, Serrani, M. Metabolic and haemostatic effects of estradiol valerate/dienogest, a novel oral contraceptive: a randomized, open-label, single-centre study. Clin Drug Invest 2011;31:573–84. https://doi.org/10.2165/11590220-000000000-00000.Search in Google Scholar PubMed

82. Kluft, C, Zimmerman, Y, Mawet, M, Klipping, C, Duijkers, IJM, Neuteboom, J, et al.. Reduced hemostatic effects with drospirenone-based oral contraceptives containing estetrol vs. ethinyl estradiol. Contraception 2017;95:140–7. https://doi.org/10.1016/j.contraception.2016.08.018.Search in Google Scholar PubMed

83. Douxfils, J, Morimont, L, Bouvy, C. Oral contraceptives and venous thromboembolism: focus on testing that may enable prediction and assessment of the risk. Semin Thromb Hemost 2020;46:872–86. https://doi.org/10.1055/s-0040-1714140.Search in Google Scholar PubMed

84. Klipping, C, Duijkers, I, Parke, S, Mellinger, U, Serrani, M, Junge, W. Hemostatic effects of a novel estradiol-based oral contraceptive: an open-label, randomized, crossover study of estradiol valerate/dienogest versus ethinylestradiol/levonorgestrel. Drugs R D 2011;11:159–70. https://doi.org/10.2165/11591200-000000000-00000.Search in Google Scholar PubMed PubMed Central

85. Agren, UM, Anttila, M, Maenpaa-Liukko, K, Rantala, ML, Rautiainen, H, Sommer, WF, et al.. Effects of a monophasic combined oral contraceptive containing nomegestrol acetate and 17beta-oestradiol compared with one containing levonorgestrel and ethinylestradiol on haemostasis, lipids and carbohydrate metabolism. Eur J Contracept Reprod Health Care 2011;16:444–57. https://doi.org/10.3109/13625187.2011.604450.Search in Google Scholar PubMed PubMed Central

86. Middeldorp, S, Meijers, JC, van den Ende, AE, van Enk, A, Bouma, BN, Tans, G, et al.. Effects on coagulation of levonorgestrel- and desogestrel-containing low dose oral contraceptives: a cross-over study. Thromb Haemostasis 2000;84:4–8. https://doi.org/10.1055/s-0037-1613958.Search in Google Scholar

87. Raps, M, Helmerhorst, FM, Fleischer, K, Dahm, AE, Rosendaal, FR, Rosing, J, et al.. The effect of different hormonal contraceptives on plasma levels of free protein S and free TFPI. Thromb Haemostasis 2013;109:606–13. https://doi.org/10.1160/th12-10-0771.Search in Google Scholar

88. Raps, M, Rosendaal, F, Ballieux, B, Rosing, J, Thomassen, S, Helmerhorst, F, et al.. Resistance to APC and SHBG levels during use of a four-phasic oral contraceptive containing dienogest and estradiol valerate: a randomized controlled trial. J Thromb Haemostasis 2013;11:855–61. https://doi.org/10.1111/jth.12172.Search in Google Scholar PubMed

89. Bonnar, J. Coagulation effects of oral contraception. Am J Obstet Gynecol 1987;157:1042–8. https://doi.org/10.1016/s0002-9378(87)80129-1.Search in Google Scholar PubMed

90. Meade, TW, Haines, AP, North, WR, Chakrabarti, R, Howarth, DJ, Stirling, Y. Haemostatic, lipid, and blood-pressure profiles of women on oral contraceptives containing 50 microgram or 30 microgram oestrogen. Lancet 1977;2:948–51. https://doi.org/10.1016/s0140-6736(77)90888-1.Search in Google Scholar PubMed

91. Stadel, BV. Oral contraceptives and cardiovascular disease (first of two parts). N Engl J Med 1981;305:612–8. https://doi.org/10.1056/nejm198109103051104.Search in Google Scholar

92. Wessler, S, Gitel, SN, Wan, LS, Pasternack, BS. Estrogen-containing oral contraceptive agents. A basis for their thrombogenicity. JAMA 1976;236:2179–82. https://doi.org/10.1001/jama.236.19.2179.Search in Google Scholar

93. Kemmeren, JM, Algra, A, Grobbee, DE. Third generation oral contraceptives and risk of venous thrombosis: meta-analysis. BMJ 2001;323:131–4. https://doi.org/10.1136/bmj.323.7305.131.Search in Google Scholar PubMed PubMed Central

94. Fruzzetti, F, Cagnacci, A. Venous thrombosis and hormonal contraception: what’s new with estradiol-based hormonal contraceptives? Open Access J Contracept 2018;9:75–9. https://doi.org/10.2147/oajc.s179673.Search in Google Scholar

95. Tepper, NK, Whiteman, MK, Marchbanks, PA, James, AH, Curtis, KM. Progestin-only contraception and thromboembolism: a systematic review. Contraception 2016;94:678–700. https://doi.org/10.1016/j.contraception.2016.04.014.Search in Google Scholar PubMed

96. Morimont, L, Haguet, H, Dogne, JM, Gaspard, U, Douxfils, J. Combined oral contraceptives and venous thromboembolism: review and perspective to mitigate the risk. Front Endocrinol (Lausanne) 2021;12:769187. https://doi.org/10.3389/fendo.2021.769187.Search in Google Scholar PubMed PubMed Central

97. Raps, M, Helmerhorst, F, Fleischer, K, Thomassen, S, Rosendaal, F, Rosing, J, et al.. Sex hormone-binding globulin as a marker for the thrombotic risk of hormonal contraceptives. J Thromb Haemostasis 2012;10:992–7. https://doi.org/10.1111/j.1538-7836.2012.04720.x.Search in Google Scholar PubMed

98. Aden, U, Jung-Hoffmann, C, Kuhl, H. A randomized cross-over study on various hormonal parameters of two triphasic oral contraceptives. Contraception 1998;58:75–81. https://doi.org/10.1016/s0010-7824(98)00071-7.Search in Google Scholar PubMed

99. Sanger, N, Stahlberg, S, Manthey, T, Mittmann, K, Mellinger, U, Lange, E, et al.. Effects of an oral contraceptive containing 30 mcg ethinyl estradiol and 2 mg dienogest on thyroid hormones and androgen parameters: conventional vs. extended-cycle use. Contraception 2008;77:420–5. https://doi.org/10.1016/j.contraception.2008.02.005.Search in Google Scholar PubMed

100. Haverinen, A, Luiro, K, Kangasniemi, MH, Piltonen, TT, Hustad, S, Heikinheimo, O, et al.. Estradiol valerate vs ethinylestradiol in combined oral contraceptives: effects on the pituitary-ovarian axis. J Clin Endocrinol Metab 2022;107:e3008–17. https://doi.org/10.1210/clinem/dgac150.Search in Google Scholar PubMed

101. Falsetti, L, Galbignani, E. Long-term treatment with the combination ethinylestradiol and cyproterone acetate in polycystic ovary syndrome. Contraception 1990;42:611–9. https://doi.org/10.1016/0010-7824(90)90002-d.Search in Google Scholar PubMed

102. Mainwaring, R, Hales, HA, Stevenson, K, Hatasaka, HH, Poulson, AM, Jones, KP, et al.. Metabolic parameter, bleeding, and weight changes in U.S. women using progestin only contraceptives. Contraception 1995;51:149–53. https://doi.org/10.1016/0010-7824(95)00011-x.Search in Google Scholar PubMed

103. Kangasniemi, MH, Arffman, RK, Haverinen, A, Luiro, K, Hustad, S, Heikinheimo, O, et al.. Effects of estradiol- and ethinylestradiol-based contraceptives on adrenal steroids: a randomized trial. Contraception 2022;116:59–65. https://doi.org/10.1016/j.contraception.2022.08.009.Search in Google Scholar PubMed

104. Landersoe, SK, Birch Petersen, K, Sorensen, AL, Larsen, EC, Martinussen, T, Lunding, SA, et al.. Ovarian reserve markers after discontinuing long-term use of combined oral contraceptives. Reprod Biomed Online 2020;40:176–86. https://doi.org/10.1016/j.rbmo.2019.10.004.Search in Google Scholar PubMed

105. Li, HW, Wong, CY, Yeung, WS, Ho, PC, Ng, EH. Serum anti-mullerian hormone level is not altered in women using hormonal contraceptives. Contraception 2011;83:582–5. https://doi.org/10.1016/j.contraception.2010.09.007.Search in Google Scholar PubMed

106. Hariton, E, Shirazi, TN, Douglas, NC, Hershlag, A, Briggs, SF. Anti-Mullerian hormone levels among contraceptive users: evidence from a cross-sectional cohort of 27,125 individuals. Am J Obstet Gynecol 2021;225:515 e1–10. https://doi.org/10.1016/j.ajog.2021.06.052.Search in Google Scholar PubMed

107. Bentzen, JG, Forman, JL, Pinborg, A, Lidegaard, O, Larsen, EC, Friis-Hansen, L, et al.. Ovarian reserve parameters: a comparison between users and non-users of hormonal contraception. Reprod Biomed Online 2012;25:612–9. https://doi.org/10.1016/j.rbmo.2012.09.001.Search in Google Scholar PubMed

108. Bernardi, LA, Weiss, MS, Waldo, A, Harmon, Q, Carnethon, MR, Baird, DD, et al.. Duration, recency, and type of hormonal contraceptive use and antimullerian hormone levels. Fertil Steril 2021;116:208–17. https://doi.org/10.1016/j.fertnstert.2021.02.007.Search in Google Scholar PubMed PubMed Central

109. Mandel, FP, Geola, FL, Lu, JK, Eggena, P, Sambhi, MP, Hershman, JM, et al.. Biologic effects of various doses of ethinyl estradiol in postmenopausal women. Obstet Gynecol 1982;59:673–9.Search in Google Scholar

110. Kuhl, H, Gahn, G, Romberg, G, Althoff, PH, Taubert, HD. A randomized cross-over comparison of two low-dose oral contraceptives upon hormonal and metabolic serum parameters: II. Effects upon thyroid function, gastrin, STH, and glucose tolerance. Contraception 1985;32:97–107. https://doi.org/10.1016/0010-7824(85)90119-2.Search in Google Scholar PubMed

111. Wiegratz, I, Kutschera, E, Lee, JH, Moore, C, Mellinger, U, Winkler, UH, et al.. Effect of four different oral contraceptives on various sex hormones and serum-binding globulins. Contraception 2003;67:25–32. https://doi.org/10.1016/s0010-7824(02)00436-5.Search in Google Scholar PubMed

112. Wiegratz, I, Jung-Hoffmann, C, Kuhl, H. Effect of two oral contraceptives containing ethinylestradiol and gestodene or norgestimate upon androgen parameters and serum binding proteins. Contraception 1995;51:341–6. https://doi.org/10.1016/0010-7824(95)00098-u.Search in Google Scholar PubMed

113. Jung-Hoffmann, C, Heidt, F, Kuhl, H. Effect of two oral contraceptives containing 30 micrograms ethinylestradiol and 75 micrograms gestodene or 150 micrograms desogestrel upon various hormonal parameters. Contraception 1988;38:593–603. https://doi.org/10.1016/0010-7824(88)90044-3.Search in Google Scholar PubMed

114. De Leo, V, Morgante, G, Piomboni, P, Musacchio, MC, Petraglia, F, Cianci, A. Evaluation of effects of an oral contraceptive containing ethinylestradiol combined with drospirenone on adrenal steroidogenesis in hyperandrogenic women with polycystic ovary syndrome. Fertil Steril 2007;88:113–7. https://doi.org/10.1016/j.fertnstert.2006.11.137.Search in Google Scholar PubMed

115. Jing, Z, Liang-Zhi, X, Tai-Xiang, W, Ying, T, Yu-Jian, J. The effects of Diane-35 and metformin in treatment of polycystic ovary syndrome: an updated systematic review. Gynecol Endocrinol 2008;24:590–600. https://doi.org/10.1080/09513590802288242.Search in Google Scholar PubMed

116. Heijboer, AC, Savelkoul, E, Kruit, A, Endert, E, Blankenstein, MA. Inaccurate first-generation testosterone assays are influenced by sex hormone-binding globulin concentrations. J Appl Lab Med 2016;1:194–201. https://doi.org/10.1373/jalm.2016.020065.Search in Google Scholar PubMed

117. Heijboer, AC, Zimmerman, Y, de Boer, T, Coelingh Bennink, H, Blankenstein, MA. Peculiar observations in measuring testosterone in women treated with oral contraceptives supplemented with dehydroepiandrosterone (DHEA). Clin Chim Acta 2014;430:92–5. https://doi.org/10.1016/j.cca.2013.12.042.Search in Google Scholar PubMed

118. Fujimoto, VY, Villanueva, AL, Hopper, B, Moscinski, M, Rebar, RW. Increased adrenocortical responsiveness to exogenous ACTH in oral contraceptive users. Adv Contracept 1986;2:343–53. https://doi.org/10.1007/bf02340051.Search in Google Scholar PubMed

119. Simunkova, K, Starka, L, Hill, M, Kriz, L, Hampl, R, Vondra, K. Comparison of total and salivary cortisol in a low-dose ACTH (Synacthen) test: influence of three-month oral contraceptives administration to healthy women. Physiol Res 2008;57(Suppl 1):S193–9. https://doi.org/10.33549/physiolres.931505.Search in Google Scholar PubMed

120. Vastbinder, M, Kuindersma, M, Mulder, AH, Schuijt, MP, Mudde, AH. The influence of oral contraceptives on overnight 1 mg dexamethasone suppression test. Neth J Med 2016;74:158–61.Search in Google Scholar

121. Carton, T, Mathieu, E, Wolff, F, Bouziotis, J, Corvilain, B, Driessens, N. Two-day low-dose dexamethasone suppression test more accurate than overnight 1-mg in women taking oral contraceptives. Endocrinol Diabetes Metab 2021;4:e00255. https://doi.org/10.1002/edm2.255.Search in Google Scholar PubMed PubMed Central

122. Panton, KK, Mikkelsen, G, Irgens, WO, Hovde, AK, Killingmo, MW, Oien, MA, et al.. New reference intervals for cortisol, cortisol binding globulin and free cortisol index in women using ethinyl estradiol. Scand J Clin Lab Invest 2019;79:314–9. https://doi.org/10.1080/00365513.2019.1622031.Search in Google Scholar PubMed

123. Streuli, I, Fraisse, T, Pillet, C, Ibecheole, V, Bischof, P, de Ziegler, D. Serum antimullerian hormone levels remain stable throughout the menstrual cycle and after oral or vaginal administration of synthetic sex steroids. Fertil Steril 2008;90:395–400. https://doi.org/10.1016/j.fertnstert.2007.06.023.Search in Google Scholar PubMed

124. Amer, S, James, C, Al-Hussaini, TK, Mohamed, AA. Assessment of circulating anti-mullerian hormone in women using hormonal contraception: a systematic review. J Womens Health (Larchmt). 2020;29:100–10. https://doi.org/10.1089/jwh.2019.7733.Search in Google Scholar PubMed

125. Arbo, E, Vetori, DV, Jimenez, MF, Freitas, FM, Lemos, N, Cunha-Filho, JS. Serum anti-mullerian hormone levels and follicular cohort characteristics after pituitary suppression in the late luteal phase with oral contraceptive pills. Hum Reprod 2007;22:3192–6. https://doi.org/10.1093/humrep/dem258.Search in Google Scholar PubMed

126. Glinoer, D. The regulation of thyroid function in pregnancy: pathways of endocrine adaptation from physiology to pathology. Endocr Rev 1997;18:404–33. https://doi.org/10.1210/edrv.18.3.0300.Search in Google Scholar PubMed

127. Glinoer, D, McGuire, RA, Gershengorn, MC, Robbins, J, Berman, M. Effects of estrogen on thyroxine-binding globulin metabolism in rhesus monkeys. Endocrinology 1977;100:9–17. https://doi.org/10.1210/endo-100-1-9.Search in Google Scholar PubMed

128. Ain, KB, Mori, Y, Refetoff, S. Reduced clearance rate of thyroxine-binding globulin (TBG) with increased sialylation: a mechanism for estrogen-induced elevation of serum TBG concentration. J Clin Endocrinol Metab 1987;65:689–96. https://doi.org/10.1210/jcem-65-4-689.Search in Google Scholar PubMed

129. Chen, JJ, Ladenson, PW. Progesterone has no effect on serum thyroxine-binding globulin concentration in men. J Clin Endocrinol Metab 1985;61:983–5. https://doi.org/10.1210/jcem-61-5-983.Search in Google Scholar PubMed

130. Chetkowski, RJ, Meldrum, DR, Steingold, KA, Randle, D, Lu, JK, Eggena, P, et al.. Biologic effects of transdermal estradiol. N Engl J Med 1986;314:1615–20. https://doi.org/10.1097/00006254-198611000-00017.Search in Google Scholar

131. Quintino-Moro, A, Zantut-Wittmann, DE, Silva Dos Santos, PN, Melhado-Kimura, V, da Silva, CA, Bahamondes, L, et al.. Thyroid function during the first year of use of the injectable contraceptive depot medroxyprogesterone acetate. Eur J Contracept Reprod Health Care 2019;24:102–8. https://doi.org/10.1080/13625187.2018.1559284.Search in Google Scholar PubMed

132. White, T, Ozel, B, Jain, JK, Stanczyk, FZ. Effects of transdermal and oral contraceptives on estrogen-sensitive hepatic proteins. Contraception 2006;74:293–6. https://doi.org/10.1016/j.contraception.2006.04.005.Search in Google Scholar PubMed

133. Sathi, P, Kalyan, S, Hitchcock, CL, Pudek, M, Prior, JC. Progesterone therapy increases free thyroxine levels--data from a randomized placebo-controlled 12-week hot flush trial. Clin Endocrinol (Oxf) 2013;79:282–7. https://doi.org/10.1111/cen.12128.Search in Google Scholar PubMed

134. Oelkers, W. Antimineralocorticoid activity of a novel oral contraceptive containing drospirenone, a unique progestogen resembling natural progesterone. Eur J Contracept Reprod Health Care 2002;7(Suppl 3):19–26. discussion 42–3.Search in Google Scholar

135. Smals, AG. Fluid retention with oral contraceptives. Gynecol Endocrinol 2000;14:476–8.10.3109/09513590009167721Search in Google Scholar

136. Brandle, E, Gottwald, E, Melzer, H, Sieberth, HG. Influence of oral contraceptive agents on kidney function and protein metabolism. Eur J Clin Pharmacol 1992;43:643–6. https://doi.org/10.1007/bf02284965.Search in Google Scholar PubMed

137. Atthobari, J, Gansevoort, RT, Visser, ST, de Jong, PE, de Jong-van den Berg, LT, Group, PS. The impact of hormonal contraceptives on blood pressure, urinary albumin excretion and glomerular filtration rate. Br J Clin Pharmacol 2007;63:224–31. https://doi.org/10.1111/j.1365-2125.2006.02747.x.Search in Google Scholar PubMed PubMed Central

138. Ahmed, AH, Gordon, RD, Taylor, PJ, Ward, G, Pimenta, E, Stowasser, M. Effect of contraceptives on aldosterone/renin ratio may vary according to the components of contraceptive, renin assay method, and possibly route of administration. J Clin Endocrinol Metab 2011;96:1797–804. https://doi.org/10.1210/jc.2010-2918.Search in Google Scholar PubMed

139. Ribstein, J, Halimi, JM, du Cailar, G, Mimran, A. Renal characteristics and effect of angiotensin suppression in oral contraceptive users. Hypertension 1999;33:90–5. https://doi.org/10.1161/01.hyp.33.1.90.Search in Google Scholar PubMed

140. Monster, TB, Janssen, WM, de Jong, PE, de Jong-van den Berg, LT, Prevention of, R, Vascular End Stage Disease Study, G. Oral contraceptive use and hormone replacement therapy are associated with microalbuminuria. Arch Intern Med 2001;161:2000–5. https://doi.org/10.1001/archinte.161.16.2000.Search in Google Scholar PubMed

141. Locsei, Z, Horvath, D, Racz, K, Szabolcs, I, Kovacs, GL, Toldy, E. Progestin-dependent effect of oral contraceptives on plasma aldosterone/renin ratio. Clin Biochem 2012;45:1516–8. https://doi.org/10.1016/j.clinbiochem.2012.06.025.Search in Google Scholar PubMed

142. Oelkers, W, Foidart, JM, Dombrovicz, N, Welter, A, Heithecker, R. Effects of a new oral contraceptive containing an antimineralocorticoid progestogen, drospirenone, on the renin-aldosterone system, body weight, blood pressure, glucose tolerance, and lipid metabolism. J Clin Endocrinol Metab 1995;80:1816–21. https://doi.org/10.1210/jcem.80.6.7775629.Search in Google Scholar PubMed

143. Oelkers, W, Helmerhorst, FM, Wuttke, W, Heithecker, R. Effect of an oral contraceptive containing drospirenone on the renin-angiotensin-aldosterone system in healthy female volunteers. Gynecol Endocrinol 2000;14:204–13. https://doi.org/10.3109/09513590009167683.Search in Google Scholar PubMed

144. Beckerhoff, R, Vetter, W, Armbruster, H, Luetscher, JA, Siegenthaler, W. Plasma-aldosterone during oral-contraceptive therapy. Lancet 1973;1:1218–9. https://doi.org/10.1016/s0140-6736(73)90530-8.Search in Google Scholar PubMed

145. Oelkers, WK. Effects of estrogens and progestogens on the renin-aldosterone system and blood pressure. Steroids 1996;61:166–71. https://doi.org/10.1016/0039-128x(96)00007-4.Search in Google Scholar PubMed

146. De Lignieres, B, Basdevant, A, Thomas, G, Thalabard, JC, Mercier-Bodard, C, Conard, J, et al.. Biological effects of estradiol-17 beta in postmenopausal women: oral versus percutaneous administration. J Clin Endocrinol Metab 1986;62:536–41. https://doi.org/10.1210/jcem-62-3-536.Search in Google Scholar PubMed

147. Gordon, MS, Chin, WW, Shupnik, MA. Regulation of angiotensinogen gene expression by estrogen. J Hypertens 1992;10:361–6. https://doi.org/10.1097/00004872-199204000-00007.Search in Google Scholar PubMed

148. Oelkers, W, Blumel, A, Schoneshofer, M, Schwartz, U, Hammerstein, J. Effects of ethinylestradiol on the renin-angiotensin-aldosterone-system and on plasma transcortin in women and men. J Clin Endocrinol Metab 1976;43:1036–40. https://doi.org/10.1210/jcem-43-5-1036.Search in Google Scholar PubMed

149. Derkx, FH, Steunkel, C, Schalekamp, MP, Visser, W, Huisveld, IH, Schalekamp, MA. Immunoreactive renin, prorenin, and enzymatically active renin in plasma during pregnancy and in women taking oral contraceptives. J Clin Endocrinol Metab 1986;63:1008–15. https://doi.org/10.1210/jcem-63-4-1008.Search in Google Scholar PubMed

150. Sealey, JE, Itskovitz-Eldor, J, Rubattu, S, James, GD, August, P, Thaler, I, et al.. Estradiol- and progesterone-related increases in the renin-aldosterone system: studies during ovarian stimulation and early pregnancy. J Clin Endocrinol Metab 1994;79:258–64. https://doi.org/10.1210/jc.79.1.258.Search in Google Scholar

151. Schunkert, H, Danser, AH, Hense, HW, Derkx, FH, Kurzinger, S, Riegger, GA. Effects of estrogen replacement therapy on the renin-angiotensin system in postmenopausal women. Circulation 1997;95:39–45. https://doi.org/10.1161/01.cir.95.1.39.Search in Google Scholar PubMed

152. Khaw, KT, Peart, WS. Blood pressure and contraceptive use. Br Med J (Clin Res Ed) 1982;285:403–7. https://doi.org/10.1136/bmj.285.6339.403.Search in Google Scholar PubMed PubMed Central

153. Woods, JW. Oral contraceptives and hypertension. Hypertension 1988;11:II11–5. https://doi.org/10.1161/01.hyp.11.3_pt_2.ii11.Search in Google Scholar PubMed

154. Henzl, MR, Loomba, PK. Transdermal delivery of sex steroids for hormone replacement therapy and contraception. A review of principles and practice. J Reprod Med 2003;48:525–40.Search in Google Scholar

155. Seely, EW, Walsh, BW, Gerhard, MD, Williams, GH. Estradiol with or without progesterone and ambulatory blood pressure in postmenopausal women. Hypertension 1999;33:1190–4. https://doi.org/10.1161/01.hyp.33.5.1190.Search in Google Scholar PubMed

156. Kalenga, CZ, Dumanski, SM, Metcalfe, A, Robert, M, Nerenberg, KA, MacRae, JM, et al.. The effect of non-oral hormonal contraceptives on hypertension and blood pressure: a systematic review and meta-analysis. Physiol Rep 2022;10:e15267. https://doi.org/10.14814/phy2.15267.Search in Google Scholar PubMed PubMed Central

157. Gordon, RD. The challenge of more robust and reproducible methodology in screening for primary aldosteronism. J Hypertens 2004;22:251–5. https://doi.org/10.1097/00004872-200402000-00006.Search in Google Scholar PubMed

158. Schirpenbach, C, Seiler, L, Maser-Gluth, C, Beuschlein, F, Reincke, M, Bidlingmaier, M. Automated chemiluminescence-immunoassay for aldosterone during dynamic testing: comparison to radioimmunoassays with and without extraction steps. Clin Chem 2006;52:1749–55. https://doi.org/10.1373/clinchem.2006.068502.Search in Google Scholar PubMed

159. Taylor, PJ, Cooper, DP, Gordon, RD, Stowasser, M. Measurement of aldosterone in human plasma by semiautomated HPLC-tandem mass spectrometry. Clin Chem 2009;55:1155–62. https://doi.org/10.1373/clinchem.2008.116004.Search in Google Scholar PubMed

160. Akinloye, O, Adebayo, TO, Oguntibeju, OO, Oparinde, DP, Ogunyemi, EO. Effects of contraceptives on serum trace elements, calcium and phosphorus levels. W Indian Med J 2011;60:308–15.Search in Google Scholar

161. Berg, G, Kohlmeier, L, Brenner, H. Effect of oral contraceptive progestins on serum copper concentration. Eur J Clin Nutr 1998;52:711–5. https://doi.org/10.1038/sj.ejcn.1600631.Search in Google Scholar PubMed

162. Theuer, RC. Effect of oral contraceptive agents on vitamin and mineral needs: a review. J Reprod Med 1972;8:13–9.Search in Google Scholar

163. Seelig, MS. Increased need for magnesium with the use of combined oestrogen and calcium for osteoporosis treatment. Magnes Res 1990;3:197–215.Search in Google Scholar

164. Berenson, AB, Rahman, M. Effect of hormonal contraceptives on vitamin B12 level and the association of the latter with bone mineral density. Contraception 2012;86:481–7. https://doi.org/10.1016/j.contraception.2012.02.015.Search in Google Scholar PubMed PubMed Central

165. Lussana, F, Zighetti, ML, Bucciarelli, P, Cugno, M, Cattaneo, M. Blood levels of homocysteine, folate, vitamin B6 and B12 in women using oral contraceptives compared to non-users. Thromb Res 2003;112:37–41. https://doi.org/10.1016/j.thromres.2003.11.007.Search in Google Scholar PubMed

166. Sutterlin, MW, Bussen, SS, Rieger, L, Dietl, J, Steck, T. Serum folate and Vitamin B12 levels in women using modern oral contraceptives (OC) containing 20 microg ethinyl estradiol. Eur J Obstet Gynecol Reprod Biol 2003;107:57–61. https://doi.org/10.1016/s0301-2115(02)00371-8.Search in Google Scholar PubMed

167. Riedel, B, Bjorke Monsen, AL, Ueland, PM, Schneede, J. Effects of oral contraceptives and hormone replacement therapy on markers of cobalamin status. Clin Chem 2005;51:778–81. https://doi.org/10.1373/clinchem.2004.043828.Search in Google Scholar PubMed

168. Green, TJ, Houghton, LA, Donovan, U, Gibson, RS, O’Connor, DL. Oral contraceptives did not affect biochemical folate indexes and homocysteine concentrations in adolescent females. J Am Diet Assoc 1998;98:49–55. https://doi.org/10.1016/s0002-8223(98)00014-5.Search in Google Scholar PubMed

169. Momeni, Z, Dehghani, A, Fallahzadeh, H, Koohgardi, M, Dafei, M, Hekmatimoghaddam, SH, et al.. The impacts of pill contraceptive low-dose on plasma levels of nitric oxide, homocysteine, and lipid profiles in the exposed vs. non exposed women: as the risk factor for cardiovascular diseases. Contracept Reprod Med 2020;5:7. https://doi.org/10.1186/s40834-020-00110-z.Search in Google Scholar PubMed PubMed Central

170. Joshi, UM, Virkar, KD, Amatayakul, K, Singkamani, R, Bamji, MS, Prema, K, et al.. Impact of hormonal contraceptives vis-a-vis non-hormonal factors on the vitamin status of malnourished women in India and Thailand. World Health Organization: Special Programme of Research, Development and Research Training in Human Reproduction. Task Force on Oral Contraceptives. Hum Nutr Clin Nutr 1986;40:205–20.Search in Google Scholar

171. Palan, PR, Strube, F, Letko, J, Sadikovic, A, Mikhail, MS. Effects of oral, vaginal, and transdermal hormonal contraception on serum levels of coenzyme q(10), vitamin e, and total antioxidant activity. Obstet Gynecol Int 2010;2010. https://doi.org/10.1155/2010/925635.Search in Google Scholar PubMed PubMed Central

172. Fallah, S, Sani, FV, Firoozrai, M. Effect of contraceptive pill on the selenium and zinc status of healthy subjects. Contraception 2009;80:40–3. https://doi.org/10.1016/j.contraception.2009.01.010.Search in Google Scholar PubMed

173. Hameed, A, Majeed, T, Rauf, S, Ashraf, M, Jalil, MA, Nasrullah, M, et al.. Effect of oral and injectable contraceptives on serum calcium, magnesium and phosphorus in women. J Ayub Med Coll Abbottabad 2001;13:24–5.Search in Google Scholar

174. Blum, M, Kitai, E, Ariel, Y, Schnierer, M, Bograd, H. Oral contraceptive lowers serum magnesium. Harefuah 1991;121:363–4.Search in Google Scholar

175. Prema, K, Ramalakshmi, BA, Babu, S. Serum copper and zinc in hormonal contraceptive users. Fertil Steril 1980;33:267–71. https://doi.org/10.1016/s0015-0282(16)44591-7.Search in Google Scholar PubMed

176. Callegari, ET, Garland, SM, Gorelik, A, Reavley, NJ, Wark, JD. Predictors and correlates of serum 25-hydroxyvitamin D concentrations in young women: results from the Safe-D study. Br J Nutr 2017;118:263–72. https://doi.org/10.1017/s0007114517002021.Search in Google Scholar

177. Harmon, QE, Umbach, DM, Baird, DD. Use of estrogen-containing contraception is associated with increased concentrations of 25-hydroxy vitamin D. J Clin Endocrinol Metab 2016;101:3370–7. https://doi.org/10.1210/jc.2016-1658.Search in Google Scholar PubMed PubMed Central

178. Harris, SS, Dawson-Hughes, B. The association of oral contraceptive use with plasma 25-hydroxyvitamin D levels. J Am Coll Nutr 1998;17:282–4. https://doi.org/10.1080/07315724.1998.10718760.Search in Google Scholar PubMed

179. Moller, UK, Streym, S, Jensen, LT, Mosekilde, L, Schoenmakers, I, Nigdikar, S, et al.. Increased plasma concentrations of vitamin D metabolites and vitamin D binding protein in women using hormonal contraceptives: a cross-sectional study. Nutrients 2013;5:3470–80. https://doi.org/10.3390/nu5093470.Search in Google Scholar PubMed PubMed Central

180. Oberg, J, Jorde, R, Figenschau, Y, Thorsby, PM, Dahl, SR, Winther, A, et al.. 100 YEARS OF VITAMIN D: combined hormonal contraceptives and vitamin D metabolism in adolescent girls. Endocr Connect 2022;11:e210395. https://doi.org/10.1530/ec-21-0395.Search in Google Scholar

181. Stanczyk, FZ, Sriprasert, I, Danis, R, Pandian, R, Matharu, H, Bender, N, et al.. Effect of oral contraceptives on total and bioavailable 25-hydroxyvitamin D. J Steroid Biochem Mol Biol 2021;211:105879. https://doi.org/10.1016/j.jsbmb.2021.105879.Search in Google Scholar PubMed

182. Prasad, AS, Oberleas, D, Moghissi, KS, Stryker, JC, Lei, KY. Effect of oral contraceptive agents on nutrients: II. Vitamins. Am J Clin Nutr 1975;28:385–91. https://doi.org/10.1093/ajcn/28.4.385.Search in Google Scholar PubMed

183. Shojania, AM, Wylie, B. The effect of oral contraceptives on vitamin B12 metabolism. Am J Obstet Gynecol 1979;135:129–34. https://doi.org/10.1016/s0002-9378(79)80030-7.Search in Google Scholar

184. Shojania, AM. Oral contraceptives: effect of folate and vitamin B12 metabolism. Can Med Assoc J 1982;126:244–7.Search in Google Scholar

185. Shojania, AM, Hornady, G, Barnes, PH. Oral contraceptives and serum-folate level. Lancet 1968;1:1376–7. https://doi.org/10.1016/s0140-6736(68)92081-3.Search in Google Scholar PubMed

186. Streiff, RR. Folate deficiency and oral contraceptives. JAMA 1970;214:105–8. https://doi.org/10.1001/jama.1970.03180010047010.Search in Google Scholar

187. Shere, M, Bapat, P, Nickel, C, Kapur, B, Koren, G. Association between use of oral contraceptives and folate status: a systematic review and meta-analysis. J Obstet Gynaecol Can 2015;37:430–8. https://doi.org/10.1016/s1701-2163(15)30258-9.Search in Google Scholar PubMed

188. Wilson, SM, Bivins, BN, Russell, KA, Bailey, LB. Oral contraceptive use: impact on folate, vitamin B(6), and vitamin B(1)(2) status. Nutr Rev 2011;69:572–83. https://doi.org/10.1111/j.1753-4887.2011.00419.x.Search in Google Scholar PubMed

189. Palmery, M, Saraceno, A, Vaiarelli, A, Carlomagno, G. Oral contraceptives and changes in nutritional requirements. Eur Rev Med Pharmacol Sci 2013;17:1804–13.Search in Google Scholar

190. Pietarinen, GJ, Leichter, J, Pratt, RF. Dietary folate intake and concentration of folate in serum and erythrocytes in women using oral contraceptives. Am J Clin Nutr 1977;30:375–80. https://doi.org/10.1093/ajcn/30.3.375.Search in Google Scholar PubMed

191. Prasad, AS, Lei, KY, Moghissi, KS, Stryker, JC, Oberleas, D. Effect of oral contraceptives on nutrients. III. Vitamins B6, B12, and folic acid. Am J Obstet Gynecol 1976;125:1063–9. https://doi.org/10.1016/0002-9378(76)90809-7.Search in Google Scholar PubMed

192. Lumeng, L, Cleary, RE, Li, TK. Effect of oral contraceptives on the plasma concentration of pyridoxal phosphate. Am J Clin Nutr 1974;27:326–33. https://doi.org/10.1093/ajcn/27.4.326.Search in Google Scholar PubMed

193. Leklem, JE, Brown, RR, Rose, DP, Linkswiler, HM. Vitamin B6 requirements of women using oral contraceptives. Am J Clin Nutr 1975;28:535–41. https://doi.org/10.1093/ajcn/28.5.535.Search in Google Scholar PubMed

194. Kowalska, K, Milnerowicz, H. Pro/antioxidant status in young healthy women using oral contraceptives. Environ Toxicol Pharmacol 2016;43:1–6. https://doi.org/10.1016/j.etap.2016.02.006.Search in Google Scholar PubMed

195. Pincemail, J, Vanbelle, S, Gaspard, U, Collette, G, Haleng, J, Cheramy-Bien, JP, et al.. Effect of different contraceptive methods on the oxidative stress status in women aged 40 48 years from the ELAN study in the province of Liege, Belgium. Hum Reprod 2007;22:2335–43. https://doi.org/10.1093/humrep/dem146.Search in Google Scholar PubMed

196. Zal, F, Mostafavi-Pour, Z, Amini, F, Heidari, A. Effect of vitamin E and C supplements on lipid peroxidation and GSH-dependent antioxidant enzyme status in the blood of women consuming oral contraceptives. Contraception 2012;86:62–6. https://doi.org/10.1016/j.contraception.2011.11.006.Search in Google Scholar PubMed

197. Durand, P, Blache, D. Enhanced platelet thromboxane synthesis and reduced macrophage-dependent fibrinolytic activity related to oxidative stress in oral contraceptive-treated female rats. Atherosclerosis 1996;121:205–16. https://doi.org/10.1016/0021-9150(95)05720-x.Search in Google Scholar PubMed

198. Basciani, S, Porcaro, G. Counteracting side effects of combined oral contraceptives through the administration of specific micronutrients. Eur Rev Med Pharmacol Sci 2022;26:4846–62. https://doi.org/10.26355/eurrev_202207_29210.Search in Google Scholar PubMed

199. Aftergood, L, Alfin-Slater, RB. Oral contraceptive-alpha-tocopherol interrelationships. Lipids 1974;9:91–6. https://doi.org/10.1007/bf02532132.Search in Google Scholar

200. Matsui, MS, Rozovski, SJ. Drug-nutrient interaction. Clin Therapeut 1982;4:423–40.Search in Google Scholar

201. Veninga, KS. Effects of oral contraceptives on vitamins B6, B12, C, and folacin. J Nurse Midwifery 1984;29:386–90. https://doi.org/10.1016/0091-2182(84)90169-1.Search in Google Scholar PubMed

202. Webb, JL. Nutritional effects of oral contraceptive use: a review. J Reprod Med 1980;25:150–6.Search in Google Scholar

203. Hudiburgh, NK, Milner, AN. Influence of oral contraceptives on ascorbic acid and triglyceride status. J Am Diet Assoc 1979;75:19–22. https://doi.org/10.1016/s0002-8223(21)05275-5.Search in Google Scholar

204. Aarskog, D, Aksnes, L, Markestad, T, Rodland, O. Effect of estrogen on vitamin D metabolism in tall girls. J Clin Endocrinol Metab 1983;57:1155–8. https://doi.org/10.1210/jcem-57-6-1155.Search in Google Scholar PubMed

205. Pilz, S, Obeid, R, Schwetz, V, Trummer, C, Pandis, M, Lerchbaum, E, et al.. Hormonal contraceptive use is associated with higher total but unaltered free 25-hydroxyvitamin D serum concentrations. J Clin Endocrinol Metab 2018;103:2385–91. https://doi.org/10.1210/jc.2018-00336.Search in Google Scholar PubMed

206. Chun, RF, Peercy, BE, Orwoll, ES, Nielson, CM, Adams, JS, Hewison, M. Vitamin D and DBP: the free hormone hypothesis revisited. J Steroid Biochem Mol Biol 2014;144:132–7. https://doi.org/10.1016/j.jsbmb.2013.09.012.Search in Google Scholar PubMed PubMed Central

207. Heijboer, AC, Blankenstein, MA, Kema, IP, Buijs, MM. Accuracy of 6 routine 25-hydroxyvitamin D assays: influence of vitamin D binding protein concentration. Clin Chem 2012;58:543–8. https://doi.org/10.1373/clinchem.2011.176545.Search in Google Scholar PubMed

208. Elsenberg, E, Ten Boekel, E, Huijgen, H, Heijboer, AC. Standardization of automated 25-hydroxyvitamin D assays: how successful is it? Clin Biochem 2017;50:1126–30. https://doi.org/10.1016/j.clinbiochem.2017.06.011.Search in Google Scholar PubMed

209. Olatunbosun, DA, Adeniyi, FA, Adadevoh, BK. Effect of oral contraceptives on Serum magnesium levels. Int J Fertil 1974;19:224–6.Search in Google Scholar

210. Stanton, MF, Lowenstein, FW. Serum magnesium in women during pregnancy, while taking contraceptives, and after menopause. J Am Coll Nutr 1987;6:313–9. https://doi.org/10.1080/07315724.1987.10720193.Search in Google Scholar PubMed

211. Halsted, JA, Hackley, BM, Smith, JCJr. Plasma-zinc and copper in pregnancy and after oral contraceptives. Lancet 1968;2:278–9. https://doi.org/10.1016/s0140-6736(68)92375-1.Search in Google Scholar PubMed

212. King, JC. Do women using oral contraceptive agents require extra zinc? J Nutr 1987;117:217–9. https://doi.org/10.1093/jn/117.1.217.Search in Google Scholar PubMed

213. Briggs, MH, Briggs, M, Austin, J. Effects of steroid pharmaceuticals on plasma zinc. Nature 1971;232:480–1. https://doi.org/10.1038/232480a0.Search in Google Scholar PubMed

214. Prasad, AS, Oberleas, D, Moghissi, KS, Lei, KY, Stryker, JC. Effect of oral contraceptive agents on nutrients: I. Minerals. Am J Clin Nutr 1975;28:377–84. https://doi.org/10.1093/ajcn/28.4.377.Search in Google Scholar PubMed

215. Heese, HD, Lawrence, MA, Dempster, WS, Pocock, F. Reference concentrations of serum selenium and manganese in healthy nulliparas. S Afr Med J 1988;73:163–5.Search in Google Scholar

Received: 2023-04-15
Accepted: 2023-06-14
Published Online: 2023-07-10
Published in Print: 2024-01-26

© 2023 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 9.5.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2023-0384/html
Scroll to top button