University of Groningen

The pill and thrombosis van Vlijmen, Elizabeth Femma Willemien

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Document Version Publisher's PDF, also known as Version of record

Publication date: 2016

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA): van Vlijmen, E. F. W. (2016). The pill and thrombosis. Rijksuniversiteit Groningen.

Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

Download date: 23-09-2021 Chapter1 General introduction and outline of thesis

Introduction

Chapter 1

Introduction

Combined oral contraceptives and venous thromboembolism

The combined oral contraceptive, i.e. the ‘Pill’, was the first medicinal product developed that had to be taken every day on a regular basis by persons not suffe- ring from any disease. Today, world-wide more than 100 million women start the day with taking the ‘Pill’.1 Although the Pill is a safe and highly effective contracep- tive method in the vast majority of women, in a small group of women its use is associated with a rare but potentially serious adverse event, i.e. venous throm- boembolism (VTE). Up to now, this association has been the topic of many studies, including the studies presented in this thesis, and probably there are many more to follow, as even after the recent 50th anniversary of the Pill this risk still has not been solved.

Combined oral contraceptives

Composition

Combined oral contraceptives (COCs) contain a combination of a synthetic estro- gen and a synthetic . The component is and recently also , but there are many different types of synthetic proges- togens, e.g. , , , , , , , , , nomegestrol, chlor- madinone, and cyproterone. Initially, the dose of ethinylestradiol was up to 150 micrograms, but since the early seventies the dose is steadily lowered to 35-15 micrograms (so-called sub-50 Pills) that are used today. Also the progestogen doses have decreased substantially. Further, currently several COCs have standard and low-dose versions. Additionally, there are not only oral combined contraceptives, but also non-oral preparations such as a vaginal ring to be applied once a month and a patch to be applied once weekly.

11 Introduction

The reason why there are so many synthetic is that the initially deve- loped progestagens, besides the desired progestogenic effects, also had androge- nic effects. Androgenic properties could result in androgenic side effects like acne, hirsutism, and negative effects on the lipid metabolism. Development therefore focused on progestagens having less androgenic properties.

The consecutive development of new COCs, i.e. with a new synthetic proges- togen, is divided into ‘generations‘, and is related to the time point of their market launch. ‘First generation’ is related to the very first COCs, which were approved around 1960. An overview of several generations of COCs with different synthetic progestogens and their first date of introduction is given in Table 1.

Table 1. Combined hormonal contraceptives divided into generations

Type of progestogen Generation Market introduction Ethinylestradiol/norethisterone-containing COC (Modicon©) 1st generation 1960

Ethinylestradiol/levonorgestrel-containing COC (Microgynon 30©) 2nd generation 1974 Ethinylestradiol/norgestimate-containing COC (Cilest©) 2nd generation 1990

Ethinylestradiol/desogestrel-containing COC (Marvelon©) 3rd generation 1984 Ethinylestradiol/gestodene-containing COC (Femodeen©) 3rd generation 1984

Ethinylestradiol/cyproterone-containing COC (Diane 35©) 4th generation 1987 Ethinylestradiol/chlormadinone-containing COC (Madinelle©)a 4th generation 1995 Ethinylestradiol/drospirenone-containing COC (Yasmin©) 4th generation 2000 Ethinylestradiol/etonogestrel-containing vaginal ring (NuvaRing©) 4th generation 2001 Ethinylestradiol/norelgestromin-containing patch (Evra©) 4th generation 2002

Estradiol/dienogest-containing COC (Qlaira©) ‘5th’ generation 2008 Estradiol/nomegestrol-containing COC (Zoely©) ‘5th’ generation 2011

a: ethinylestradiol/chlormadinone-containing COCs are not available in NL

However, as there are now many different preparations, it is suggested not to use these terms any longer, as they lead to confusion. In practice this has already resulted in studies on COCs and thromboembolism with e.g. COCs being catego-

12 Introduction

rized as a ‘3rd generation’, ‘4th generation’ or as a ‘2nd generation’, which makes- the interpretation of clinical results for the individual progestogens such as risk of VTE, challenging.2

History of combined hormonal contraceptives

Around 1920, several Austrian scientists demonstrated the occurrence of tempo- rary contraception by transplantation of ovaries of a pregnant animal into a non- pregnant animal. These experiments were followed by studies in mice showing that extracts of ovaries could inhibit ovulation. These first studies were performed by Ludwig Haberlandt, an Austrian professor of physiology. He is seen as the Grandfather of the ‘Pill’, as he was the first to perform experiments with the goal to obtain contraception by using female sex hormones. His aim was to develop by mimicking a pregnant state, which would create a ‘temporary hormonal ’. In 1931, he started the production of a hormo- nal extract ‘Infecundin’ in co-operation with the Hungarian pharmaceutical com- pany Gedeon-Richter. However, he was severely criticized by his colleagues and in the lay public press, who accused him of hindering the unborn life. Despite all opposition, he started the first clinical trials. Unfortunately, he died at an early age and his co-worker Otto Fellner lost his life during World War II, which brought an end to this first initiative.3,4

Although Gabriel Fallopinus already in 1561 mentioned the existence of a ‘corpus luteum’, of which the structure is described by the Dutch anatomist Volcher Coiter in 1573,5 its function in regulating the menstrual cycle remained unknown for long. Only at the end of the nineteenth century scientists in Europe and USA discovered the endocrine function of this temporary existing ‘gland’ after ovulation has taken place. Between 1930-1937, scientists unraveled the action of the corpus luteum hormone as well as its chemical structure, and named the hormone ‘’.6,7

But at the time, it was still difficult and very expensive to produce sex hormones, as the only way was to process thousands of animal ovaries to retrieve a small amount of sex hormones. In early 1941, the American chemist Russell Marker solved this problem by the discovery of the Mexican wild Yam plant, which contained the plant steroid diosgenin. This plant was said used by Mexican

13 Introduction

women for treatment of menstrual cramps. This phytoestrogen could be used as a source to produce large amounts of progesterone. Carl Djerassi in Mexico and other chemists in the US improved the potency of the ‘Yam’ progesterone by removing the carbon at the 19-position, which led to the first synthetic proges- togen ‘norethindrone’. Meantime in Europe, chemists at Schering AG in Germany developed the potent synthetic estrogen ‘ethinylestradiol’. The latter became the estrogen compound in future developed combined oral contraceptives.8

In 1944, disappointed by the negative attitude of politics of academic institutions at the time, the physiologists Gregory Pincus, Hudson Hoagland, Min-Chueh Chang, and John Rock, gynecologist, started the ‘Worchester foundation’, a private research center for experimental biology. With financial support and encouragement of biologist Katharine McCormick and Margaret Sanger of the Planned Parenthood association, they focused on development of oral hormonal contraception.9.10 This collaboration led to a first study in women who received oral natural progesterone. In this first study, a 20-day dose regimen starting at day 5 of the menstrual cycle was chosen as it covers the period in the cycle during which ovulation occurs. Discontinuation after these 20 days would result in a withdrawal bleeding thereby mimicking the normal menstrual bleeding, which could reassure women that they are not pregnant.

In 1956, a first study was performed in women who received a synthetic proges- togen and results were published in Science.11 The results indicated that ovulation inhibition could be achieved and that after discontinuation a rapid return to fertility was observed. However, the synthetic progestogen norethindrone used appeared to be contaminated with the estrogen ‘’. When repeating this study using non-contaminated progestogen, the women reported unwanted irregular (breakthrough) bleeding. Based on these results, it was concluded that the progestogen should be combined with a certain amount of estrogen to keep the endometrium stabilized, which avoids occurrence of breakthrough bleeding. The results of these studies showed the usefulness of combining a progestogen with an estrogen, i.e. the concept of the combined contraceptive pill was born. An additional study in which women used norethindrone 10 mg combined with ethinylestradiol 0.15 mg showed a decrease in risk of pregnancy to 2.4%.12 When in 1957 the pharmaceutical company Searle had submitted an application for approval of this combined oral contraceptive pill, Pincus and Rock strongly contri- buted to convince the US food and drug administration to approve Enovid©.

14 Introduction

This first ‘Pill’, with a huge dose of 150 micrograms mestranol and 10 mg norethy- nodrel, was initially approved for ‘treatment of miscarriages and severe menstrual disorders’, with a patient leaflet mentioning ‘temporary infertility’ as an ‘adverse effect’. But in 1960, the indication of contraception was accepted.13

Mechanism of contraceptive action

Combined oral contraceptives contain an estrogen and a progestogen. The progestogen inhibits ovulation, as it primarily suppresses the production of lutei- nizing hormone (LH), thereby preventing the LH-surge needed to start ovulation. The estrogen component suppresses the production of follicle stimulating hormone (FSH), which will prevent follicular growth and development of a domi- nant follicle. The estrogen component also provides stabilization of the endome- trium, which will reduce risk of breakthrough bleeding and it also potentiates the progestogenic activity of the progestogens.10 This potentiating has the advantage that the progestogen dose can be reduced. Initially, a dose regimen was chosen to mimic the menstrual cycle, which included a period of 21 days of tablet intake, followed by a 7-day break in which a withdrawal bleeding occurred due to the drop in hormone levels. Nowadays, several variants are introduced in which the 7-day break is reduced to 4 or even 2 days per month, and recently there are dosage schemes approved in which the ‘Pill’ can be taken continuously for up to 120 days.

Contraceptive efficacy in comparison to other contraceptive methods

Combined oral contraceptives (COCs) are one of the most efficacious contracep- tives available. In Europe, combined oral contraceptives are the most frequently used type of ; about 85% of women in Western Europe have taken the pill during their fertile life. When taken according to the recommended dose regimen (perfect use), the risk of pregnancy, expressed as the Pearl Index, is only about 0.3% (Pearl Index: the number of failures of a contraceptive method [pregnancies] per 100 woman who use the contraceptive for one year). Only those hormonal contraceptive methods which are not dependent of compliance to regu- lar intake, i.e. progestogen-containing IUDs and implants have an even lower pregnancy risk of around 0.1%, as presented in the table of failure rates based on US figures. However, Pearl Index for typical use (actual use which also includes pregnancies due to non-compliance to the dose regimen) is far lower in Europe than in the US, of which the reason is still not elucidated, although it is suggested that less compliance and the higher BMI of US women may play a role.14,15

15 Introduction

Table 2. Percentage of US women experiencing an unintended pregnancy during the first year of use per contraceptive method

% Women experiencing an unintended pregnancy within the first year of use

Method Typical use1 Perfect use2 No method 85 85 29 18 Diaphragm 16 6 : - female 21 5 - male Combined oral contraceptive 8 0.3 Desogestrel-only pill 8 0.3 Combined hormonal patch (Evra©) 8 0.3 Combined hormonal vaginal ring (NuvaRing©) 8 0.3 Medroxyprogesterone injection (Depo-Provera©)* 3 0.3 Levonorgestrel-containing IUD (Mirena©)* 0.2 0.2 Copper-containing IUD* 0.8 0.6 Etonogestrel containing implant (Implanon©)* 0.05 0.05 Adapted from Trussel J. Contraception 2011;83:397-40414 1: typical use shows how effective the different methods are during actual use (including inconsistent or incorrect use). 2: Pregnancy rates during perfect use show how effective methods can be, where perfect use is defined as following the directions for use. * : LARC’s, long-acting reversible contraception

The social, cultural and economic impact of the approval of the Pill

The introduction of the pill caused an earthquake in the Western world, as it rearranged the relationships between men and women completely. For the first time, women were able to plan their own life, by having the opportunity to decide when to get pregnant and how many children she would have. This meant that she could finish an education, have a job and have a life of her own. Up till then, the fear of unexpected pregnancy before marriage and in marriage, and the frequent state of pregnancy were women’s major burden. So there is a straight line between the introduction of the Pill and the changing in family structure. Initially only married women could have a receipt, but when the Pill became also available to unmarried women, this coincided with the increase in the age at first marriage and the increase in women in professional degree programs.16 Its introduction was the basis of the large positive change up to the economic and social status of the women in the world today.

16 Introduction

At its recent 50th anniversary, the Pill was voted by many as the greatest medical advance of the 20th century, as it enabled women to change their domestic posi- tion and plan families around their own careers.

As to the situation in the Netherlands, until the introduction of the ‘Pill’, the Netherlands was prudish society with high birth rates. Contraception was an inde- cent subject, and physicians were ignorant to the subject or didn’t want to have anything to do with it. To sell contraceptives in public was forbidden by law. In 1950, the editors of the ‘Nederlands Tijdschrift voor Geneeskunde’ apologized for having published two articles dealing with different methods for birth control.17,18 With the introduction of the ‘Pill’, a rigorous change in attitude started which was not to be stopped, as in no time a large number of Dutch women started using it. Initially, there was lot of opposition of the church, politicians and doctors, the latter as they were not convinced it would actually work. But in 1963, by wording of the Dutch Bishop Bekkers, the Dutch Catholic church agreed that married couples could decide themselves if they would use the Pill. Within 10 years after its introduction, the Netherlands had highest percentage of Pill users in the world.

Venous thromboembolism

Venous thromboembolism (VTE) is defined as a non-physiologic formation of a thrombus in a vein. In 1846, the German pathologist Virchow hypothesized 3 main mechanisms to cause thrombosis: damage to the vessel wall, stasis of the blood stream, and alterations in the blood composition. The most common location is deep vein thrombosis of the leg and when this thrombus partly deta- ches (embolizes) it can be transported upwards through the right heart into one of the arteries of the lung and cause pulmonary embolism.19,20

VTE is considered a multifactorial disease, based on interactions between genetic and acquired risk factors. In the general population, many risk factors for VTE are present, and often within the same individual, but an interaction of hereditary and acquired risk factors may eventually lead to the development of VTE. Actually, the current view is that the presence of several e.g. more than one risk factors appears to be a requirement for the development of VTE.21-23 There are many factors that contribute to the individual baseline risk of VTE. In this, hereditary thrombophilia and first-degree family history both are established risk factors. The baseline risk

17 Introduction

can be further increased by underlying conditions like malignant disease and obesity, advanced age, and by exogenous risk factors such as surgery, trauma, immobilization, and specifically in women, hormonal exposure due to use of com- bined oral contraceptives (COCs) hormone replacement therapy, hormonal treat- ments in assisted reproduction techniques (in vitro fertilization), and the pregnancy-postpartum period.

Risk of venous thromboembolism during combined oral contraceptive use

Since their introduction in 1960, the first case reports appeared in public literature suggesting that COCs are associated with an increased risk of VTE.24,25 Despite an up to 10-fold reduction in dose of both components and the development of new progestogens, this risk, albeit lower, remained.26

Up to 1995, the risk of VTE was assumed attributable to ethinylestradiol (EE), because this component is identical in all COCs, whereas the type of synthetic progestogen included could differ.

Currently, there is consensus that the type of progestogen can influence this estrogen-mediated VTE risk. Such difference in VTE risk became apparent in 1995 with the simultaneous publication of three epidemiological studies in the Lancet, which observed that the use of 3rd-generation COCs (containing the progestogens desogestrel or gestodene) in healthy women and in women with factor V Leiden mutation resulted in a higher VTE risk than the use of 2nd-generation COCs (containing levonorgestrel).26-28 Many other studies followed that further evalua- ted differences in VTE risk between COCs containing 2nd- and 3rd-generation progestogens.29 In the Netherlands, the results of these studies led to a change in the patient leaflets of all COCs in 1997, by informing that the risk of VTE during use of COCs containing desogestrel- or gestodene-containing COCs (3rd-generation) is 1.5-2 times higher than with levonorgestrel-containing COCs (2nd-generation). Additionally, the Dutch General Practitioners Standard on com- bined oral contraceptives was changed in 1998 to include a statement that second-generation COCs should be considered first choice.30

Based on the results of the most recent epidemiological studies, lowest risk of VTE is observed during use of COCs containing levonorgestrel and norgestimate

18 Introduction

(both 2nd-generation COCs), or norethisterone (a 1st-generation COC), whereas a higher risk is observed during use of COCs containing desogestrel or gestodene, drospirenone, and cyproterone.32-37 The COC containing cyproterone is approved for hormonal treatment in women with -sensitive acne and hirsutism, but has similar contraceptive properties. Recently, also the transdermal patch containing norelgestromin and ethinylestradiol and the vaginal ring containing etonogestrel and ethinylestradiol were shown to have similarly higher VTE risk.34 The relative risk of VTE of recently introduced COCs containing estradiol instead of ethinylestradiol compared to ethinylestradiol-containing COCs is yet unknown. Nevertheless, the absolute risk of VTE is low: varying between 6-9 cases of VTE per 10.000 years of COC-use in comparison to 2-3 cases of VTE per 10.000 women per year who do not use COCs.33,34 Based on these risk profiles, it is now generally recommended in European guidelines,40 including Dutch guidelines (Nederlands Huisartsen Genoodschap already in 1998)30 to preferentially pres- cribe 2nd-generation COCs to women who start using a COC for the first time.

Recently, a re-evaluation of the risk of VTE during COC-use and during use of non-oral combined hormonal contraceptives has been performed by the Euro- pean Medicines Agency based on all published and unpublished data currently available, including those studies discussed above.2 On the basis of this review, also the COCs containing ethinylestradiol/ norgestimate or ethinylestradiol/ nore- thisterone have similar VTE risk as noted in women using ethinylestradiol/ levonorgestrel-containing COCs. This re-evaluation subsequently resulted in adap- ting the information in the patient leaflets regarding the risk of VTE of all COCs available in the EU to include the latest VTE risk estimations. The results of this review of VTE risk in different COCs and non-oral combined hormonal contracep- tives are shown in Table 3.

19 Introduction

Table 3. Estimated incidence of VTE risk with different combined hormonal contraceptives

RR against Incidence per Generation levonorge- 10.000 women/ strel-COCs per year of use

Non-pregnant non-user 2-3 Ethinylestradiol/levonorgestrel-containing COC (Microgynon 30©) Referentie 5-7 2nd

Ethinylestradiol/norethisterone-containing COC (Modicon©) 1 5-7 1st

Ethinylestradiol/norgestimate-containing COC (Cilest©) 1 5-7 2nd

Ethinylestradiol/desogestrel-containing COC (Marvelon©) 1.5-2 9-12 3rd Ethinylestradiol/gestodene-containing COC (Femodeen©) 1.5-2 9-12 3rd Ethinylestradiol/drospirenone-containing COC (Yasmin©) 1.5-2 9-12 4th Ethinylestradiol/cyproterone-containing COCs (Diane 35©) 1.5-2 9-12 4th Ethinylestradiol/etonogestrel-containing vaginal ring (NuvaRing©) 1-2 6-12 4th Ethinylestradiol/norelgestromin-containing patch (Evra©) 1-2 6-12 4th Ethinylestradiol/chlormadinone-containing COC (Madinelle©)a Unknown Unknown 4th Estradiol/dienogest-containing-COC (Qlaira©) Unknown Unknown 4th Estradiol/nomegestrol-containing COCs (Zoely©) Unknown Unknown ‘5th’

RR: relative risk. a: not available in the Netherlands. Adapted from the ‘Assessment report for combined hormonal contraceptives containing medicinal products’ 16 January 2014, EMA/739865/20132

Risk of VTE also changes with duration of COC-use: VTE risk is highest in the first year following initiation of COC-use and when restarting after a break of at least one month. The risk then reduces over the next year and remains stable thereafter.33,34,38-40 Although the risk is high in the first year of COC-use and then falls, it remains higher than in non-users.

Further, risk of VTE is estrogen-dose dependent; Current low dose (30 microgram ethinylestradiol) has lower risk than COCs containing 50 micrograms or more. Limited data suggest risk of VTE to be slightly lower with COCs containing 20 microgram ethinylestradiol than with COCs containing 30 microgram ethinylestradiol.33

20 Introduction

“Estrogenicity” of combined oral contraceptives

Based on the differences in VTE risk with COCs that have the same estrogen dose but different progestogen type, the prothrombotic effect of COCs is considered not only influenced by the estrogen compound, but is based on the “total estroge- nicity” of the specific COC.41 A higher estrogen dose will increase the “total estro- genicity”, but the net estrogenic effect of a COC is also dependent on the anti- estrogenic properties of the synthetic progestogen type. It is assumed that deso- gestrel- and gestodene-containg COCs (3rd-generation progestogens), but also the more recent COCs containing drospirenone, cyproterone, etonogestrel, norelge- stromin (4th-generation COCs), have no or weaker anti-estrogenic activity than levonorgestrel-containing COCs. It is therefore postulated that because these COCs have less or no antagonistic effects on the prothrombotic effect of the estro- gen component, the net thrombotic effects will be higher and therefore could result in a higher risk of VTE.

Sex Hormone Binding Globulin (SHBG), produced in the liver, dose-dependently increases after oral intake of EE alone, whereas progestogen intake results in a SHBG decrease, which is dependent on dose and type of progestogen. In view of this, it is postulated that the SHBG increases with increasing estrogenicity, and it was suggested to use SHBG as a surrogate for predicting VTE risk.42

In addition, an association between SHBG levels with a thrombin generation- based APC-resistance test was reported.43 However, others did not share this opinion. Although estrogen intake and more particularly a pregnancy increases SHBG, SHBG is not involved in the clotting cascade, and various metabolic systems also affect the SHBG level.43 However, SHBG, thrombin-based APC resistance tests (APCsr), or any other postulated tests or coagulation factors are as yet not validated as a surrogate endpoint for the clinical outcome of VTE. Some studies have evaluated whether the level of APCsr could be used to explain risk differences observed between users of 2nd or 3rd generation COCs. One study found no association.44 In another study, the large range in APCsr values observed in the plasma samples of women and the small sample sizes resulted in large confidence intervals for the estimated risk. This precluded a meaningful analysis in the female population with respect to the question of whether venous thrombo- tic risk gradually increased with increasing APCsr.45 This means that it has yet to be demonstrated that such alterations are related to an increase in clinical risk of VTE among COC users, and in particular differences in risks between COCs.

21 Introduction

Biological background of risk of venous thromboembolism during combined oral contraceptive use

The biological background of this association between the use of COCs and the rare adverse event of VTE is complex. The ‘estrogenic’ effect of COCs falls into the category of modifying the blood composition, as its use induces changes in hemostatic balance into a prothrombotic direction by influencing coagulant- and anticoagulant factors and fibrinolysis in Table 4. Results are included which were observed during two months treatment of a 2nd-generation and a 3rd generation COC, with a washout of 2 months between treatments.46

Table 4. Changes in coagulation-, anticoagulation-, and fibrinolytic factors

nd rd 2 generation COCs 3 generation COCs Coagulation factorsa Prothrombin ↑ ↑* Fibrinogen ↑ ↑ Factor V ↓ ↓* Factor VII ↑ ↑* Factor VIII ↑↓ ↑ Factor X ↑ ↑

Anticoagulant systemb Antithrombin ↑↓ ↓

α2-macroglobulin ↑ ↑

α1-antitrypsin ↑ ↑ Protein C inhibitor ↑ ↑ Protein C ↑ ↑*

Protein Stotal and free ↓ ↑* APCsr (aPTT-based assay) ↑↓ ↓* APCsr (EPT-based assay) ↑

Fibrinolytic systemc tPA, antigen, ng/ml ↓ ↓ tPA activity U/ml ↑ ↑ PAI-1 Ag, ng/ml ↓ ↓ PAI-1, act U/ml ↓ ↓ Plasminogen, activity, % ↑ ↑ TAFI Ag, % ↑ ↑*

Significant differences between users of second (2nd gen) and third (3rd gen) generation oral contraceptives (p<0.05). APCsr: activated protein C sensitivity ratio; aPTT: activated partial prothrombin time; ETP: endogenous thrombin potential; PAI: plasminogen activator-inhibitor; TAFI: thrombin-activable fibrinolysis inhibitor; tPA: tissue-type plasminogen activator; Ag: antigen; act: activity; SD: standard deviation. a,47b,48 c,49

22 Introduction

The observed changes are more pronounced during use of 3rd generation COCs than noted during use of 2nd generation COCs, see table above.46-49 It is expected that changes induced by 4th generation COCs will follow the same pattern as noted of 3rd generation COCs. However, in healthy women, these changes in hemostatic variables remain within the normal ranges.21 As stated previously, up to now there are no hemostatic tests that can be used as a validated surrogate for the clinical endpoint of VTE.

Risk of venous thromboembolism during pregnancy and post-partum period

Risk of VTE is substantially higher during pregnancy, especially in the post-partum period, than observed during the use of COCs. In comparison to non-pregnant women, the VTE risk during pregnancy is increased about 9-fold, and up to 50-fold during the postpartum period.50 The main reason of the increased risk of VTE in pregnancy is the hypercoagulable state. This increased coagulation tendency is a physiological adaptation in order to protect the woman at time of delivery from bleeding complications at the placental site of the uterus after placental separa- tion. Like COC-use, a pregnancy is accompanied by changes in sex hormone levels. In comparison to the non-pregnant state, pronounced hemostatic changes are noted, which are presumed due to the huge increase in endogenous estrogen levels already starting from the first trimester in pregnancy; the production of estradiol and estrone is increased 100-fold, and that of estriol a 1000-fold. An accelerated increase of both estradiol and estriol is noted at week 36 of gestation, while at the same time the progesterone level decreases. An increase of coagula- tion factors, a slight decrease in natural anticoagulants and a decreased fibrinoly- tic activity are observed. The most pronounced hemostatic changes towards hypercoagulability are noted in the late third trimester, presumably preparing for the upcoming delivery. Initiated by delivery and uterine contractions, activation of blood coagulation occurs, as after placental separation about more than 100 spiral arteries torn from the uterus wall must thrombose. After delivery, fibrinoly- tic activity rapidly returns to baseline values, while blood coagulation factors mainly normalize during the first 4 to 6 weeks postpartum, although the level of the natural anticoagulant protein S (free fraction) can be decreased for as long as 8 weeks postpartum. Although the increased risk of VTE especially during the postpartum period is firmly established, the exact mechanism of raising sex hormone levels, accompanied with prothrombotic alterations in hemostasis, and the time to return to normal hemostasis is as yet not fully understood.51,52

23 Introduction

Thrombophilia

Inherited thrombophilias are coagulation disorders, leading to a genetic tendency to develop VTE. There are several relevant hereditary thrombophilias identified, which can be divided into rare but severe thrombophilias consisting of a deficiency of the natural anticoagulants protein C, protein S and antithrombin,53-55 of which the prevalence in the general population is about 0.1% each.56-58 These deficiencies increase the risk of VTE more than 10-fold. The absolute risk of VTE in antithrombin-, protein C–, or protein S–deficient persons ranges from 0.7 to 1.7 per 100 person-years. However, these absolute risks are estimated in thrombophi- lic cohort studies only,59-62 which risks cannot be extrapolated directly to the gene- ral population. However, though based on a very low prevalence, some popula- tion based case-control studies showed that antithrombin and protein C levels in the deficient range clearly increased risk of VTE 5- and 7-fold.63 However, protein S levels in the deficient range were not associated with an increased VTE risk, except at extremely low level.64

Additionally, mild but more frequent hereditary thrombophilic defects are a muta- tion in the coagulant factor V Leiden, preventing its inactivation by activated anticoagulant protein C65 and a mutation in prothrombin G20210A, which leads to increased prothrombin, the precursor of thrombin.66 These defects have a preva- lence of 5%67 and 2%,68 respectively, in the general Dutch population. Factor V Leiden mutation is the most common thrombophilic defect in the Caucasian population, but far less common or even rare in populations of other ethnic origin. Prothrombin mutation is only prevalent in the Caucasian population.22 In carriers of these mutations, the risk reported in case-control studies is increased about 2-5 fold,66,70 whereas homozygote carriers and persons with double hetero- zygosity for factor V Leiden and prothrombin mutation have further increased VTE risk.71,72 The absolute risk of VTE in factor V Leiden carriers is estimated being 0.15 per 100 person-years.61,69 Nevertheless, in about 50% of families suffering from VTE, no hereditary thrombophilic defects could be identified. It is possible that some further genetic defects will be identified through genome wide association studies, but if so, such defects are expected to have low risk.

24 Introduction

Hereditary thrombophilia and risk of venous thromboembolism during combined oral contraceptive use

The risk of VTE associated with the use of COCs will increase when the woman’s baseline risk is altered. One of the strongest factors that increase baseline risk is the presence of hereditary thrombophilia. Information on the risk of VTE in COC- users who have severe thrombophilic defects (antithrombin-, protein C, or protein S-deficiency) is very limited,60,73 and only evaluated in thrombophilic cohort studies due to the rarity of these thrombophilic defects. In these families, the risk of VTE in COC-users with severe thrombophilias risk increased risk about 10-fold versus non-thrombophilic COC-users; the observed increased absolute risk varied between 4.3 to 27.5 per 100 pill-years of use.

As to the risk of VTE in COC-users who have mild thrombophilic defects, i.e. factor V Leiden or prothrombin 20210A mutation, a large number of case-control studies have evaluated the risk of VTE in COC-users with factor V Leiden and prothrombin-G20210A, which reported odds ratios varying between a 2 and 26-fold increased risk in comparison to matched control groups.29,74-90 Few subgroup-analyses in family cohort studies presented the absolute risk of VTE in COC-users with factor V Leiden or prothrombin mutation, in which the incidence rate varied between 0.48 and 2.0 per 100 pill-years of use.60,73,91,92

Family history and risk of venous thromboembolism during combined hormonal contraceptive use

Several studies have explored the impact of positive family history and currently it is considered an independent risk factor of VTE, regardless of the presence of other risk factors, including hereditary thrombophilia.93 Several case-control studies reported family history to increase the risk of VTE two to three-fold.93-95 Additionally, two studies reported the VTE risk of a positive family history as higher in first-degree female relatives during fertile age than in male relatives of that age.96,97

However, only one study has considered whether also specific characteristics of patients (probands) with VTE could influence the risk of VTE in their relatives.98 Additionally, it was taken into account whether the proband had experienced a

25 Introduction

provoked or unprovoked VTE. Results of this study indicated that relatives of younger patients (<45 years) with either provoked or unprovoked VTE, have much higher risk for VTE than relatives of older patients. An interesting follow-up ques- tion would be whether these family-conferred risks could also be hormone- or gender specific. In the study described earlier, gender of the patient with VTE was reported as having no influence on the risk of VTE in their first-degree relatives (OR: 0.96 (95% confidence interval: 0.67-1.38).98 However, no study has yet evalu- ated whether a female patient with a hormonally-related VTE could be an additio- nal predictor of (hormonally-related) VTE risk in her first-degree female relatives.

Recurrence rate in women with venous thromboembolism associated with com- bined oral contraceptive use

Several comparative studies evaluated the risk of VTE recurrence in women with COC-associated VTE, in which the recurrence risk varied between 1.8% to 5.6%.99- 103 However, often these women were included as a subgroup, or COC-exposure was combined with exposure to pregnancy and/or hormone replacement, or studies were restricted to women having COC-use as single risk factor.101,103 An additional difficulty is that in several studies not all women discontinued COC-use or categorized COC-associated VTE as unprovoked. The recurrence rate in women with COC-associated VTE who post-VTE have discontinued further COC-use is therefore not firmly established.

Clinical profile of a woman with COC-associated VTE

Up to now, it is not yet fully understood who will develop a COC-associated VTE. Hereditary thrombophilia and first-degree family history, even in the absence of thrombophilia increases the risk of VTE, but studies evaluating the interaction between family history and COC-use are contradictory.104,105 Concomitant presence of other risk factors besides COC-use additionally increases the risk of VTE, like surgery, trauma, immobilization, postpartum period, malignant disease, and obesity. Many of these risk factors are frequently present in COC-users, but only few women actually develop a COC-associated VTE.

26 Introduction

Outline of this thesis

Central theme of this thesis is the association between COC-use and VTE. In this context, the contribution of other risk factors of VTE, including thrombophilia and family history, have been evaluated. Further, comparisons were made against the risk of VTE in women during pregnancy; the condition that is to be prevented by COC-use.

Information on the absolute risk of VTE in COC-users who have thrombophilic defects is limited, whereas this risk in women in combination with other defects was hardly considered in any study. In Chapter 2, we present a thrombophilic family cohort study, in which we assessed the absolute and relative risk of VTE during use of COCs in women with or without protein S-, protein C-, or antithrombin-deficiency and additionally assessed the contribution of other known thrombophilic defects. In Chapter 3 we present a thrombophilic cohort study in which we assessed the absolute and relative risk of first VTE during COC- use and pregnancy-postpartum in women with or without heterozygous, double heterozygous, or homozygous factor V Leiden or prothrombin-G20210A muta- tion. Furthermore, the absolute risk of VTE during COC use in women with or without mild hereditary thrombophilia was put into the perspective of contracep- tive failure of COC and alternative contraceptives.

In Chapter 4, we present a systemic review and meta-analysis of studies, which compared the risk of VTE in COC-users with or without hereditary thrombophilia, in which a case presentation is taken as a starting point.

As shown in cohort studies described in chapter 3 and 4, women from thrombo- philic families have an increased risk of venous thromboembolism (VTE), which increases further during combined oral contraceptive use (COCs) and pregnancy. It is unknown whether this additional risk differs between relatives of male and female patients with VTE, and also whether it matters if the female patient had a hormonally-related VTE (during COC-use or pregnancy). In order to explore this question, we performed a large retrospective family cohort study, which is presented in Chapter 5. In a retrospective thrombophilic family cohort of 1005 first-degree female relatives of reproductive age, we compared VTE risk in relatives of female versus male patients, and between relatives of female patients with and without hormonally-related VTE.

27 Introduction

Although COC-use increases the risk of VTE, it is yet not fully understood which woman will develop COC-associated VTE and evidence on recurrence risk varies. In Chapter 6 we present a prospective cohort of 125 consecutive Dutch women with COC-associated VTE who are, according to Dutch General Practitioner (GP) guidelines, was preferentially prescribed levonorgestrel-containing COCs since 1998.29 In this study, we describe clinical characteristics and post-VTE contracep- tion choices, and have prospectively assessed VTE recurrence after discontinua- tion of further COC-use and thromboprophylaxis in subsequent pregnancies.

References

1. WHO scientific group meeting on cardiovascular disease and steroid hormone contraceptives. Weekly Epidemiol Rec 1997;72:361-363.

2. Assessment report for combined hormonal contraceptives containing medi- cinal products. Procedure under Article 31 of Directive 2001/83/EC. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/Referrals_document/ Combined_hormonal_contraceptives/Recommendation_provided_by_ Pharmacovigilance_Risk_Assessment_Committee/WC500160272.pdf

3. Haberlandt E. Ludwig Haberlandt - A pioneer in hormonal contraception. Wien Klin Wochenschr 2009;121:746-749.

4. Djerassi C. Ludwig Haberlandt - Grandfather of the ’Pill’. Wien Klin Wochen- schr. 2009;121:727-728.

5. The Edinburgh Medical and Surgical Journal. A concise view of the latest important discoveries in medicine, surgery, and pharmacy, volume 55, 1841.

6. Frobemius F. Ludwig Fraenkel: ‘spiritus rector’ of the early progesterone research. Eur J Obstet Gyn Reprod Biol 1999;83:115-119.

7. Morgan TS and Davidson SG. The action of the corpus luteum hormone on the human menstrual cycle. Lancet 1937;April 10:861-64.

8. Innhoffen HH, Hohlweg W. Naturwissenschaften 1938;36(6):96.

9. Bates RW. Pincus G. Recent Prog Horm Res 1968;4:V-VI.

28 Introduction

10. Fritz MA and Speroff L. Clinical gynecologic endocrinology and infertility. 2011; 8th Edition Lippincott, Williams & Wilkins, Philadelphia.

11. Rock J, Pincus G. Garcia CR. Effects of certain 19-nor- steroids on the normal human menstrual cycle. Science 1956;124:891-893.

12. Pincus G, Garcia CR, Rock J, Paniagua M, Pendleton A, Laraque F, Nicolas R, Borno R, Pean V. Effectiveness of a contraceptive; effects of a progestin- estrogen combination upon fertility, menstrual phenomena, and health. Science 1959;130:81-83.

13. White Junod S. FDA's Approval of the First Oral Contraceptive, Enovid. Avai- lable at: http://www.fda.gov/AboutFDA/WhatWeDo/History/ProductRegulation/Selecti- onsFromFDLIUpdateSeriesonFDAHistory/ucm092009.htm

14. Trussel J. Contraceptive failure in the United States. Contraception 2011;83:397-404.

15. Trussel J. and Portman D. The creeping pearl: why has the rate of contracep- tive failure increased in clinical trials of combined hormonal contraceptive pills? Contraception 2013;88:604-610.

16. Goldin, C, and Katz LF. The Power of the Pill: Oral Contraceptives and Women's Career and Marriage Decisions". J Political Economy 2002;110: 730-770.

17. Wijnberg R. Voorbehoedmiddelen tegen zwangerschap. Ned Tijdschr Geneeskd 1949;93:4242-4245.

18. Kouwenaar W. De taak van het Tijdschrift. Ned Tijdschr Geneeskd 1950;94:290.

19. Virchow R. Die verstopfung der Lungenarterie und ihre Folgen. Beitr Exper Path Physiol 1846;2(1).

20. Virchow R. Phlogose und Thrombose im GeFäβsystem. Gesammelte Abhandlungen Zur Wissenschaftlichen Medizin Frankfurt-am-Main: Von Meidinger Sohn & Comp. 1856. P. 458-636.

29 Introduction

21. Rosendaal FR. Venous thrombosis: a multicausal disease. Lancet 1993;353:1167-1173.

22. Rosendaal FR, Reitsma PH. Genetics of venous thrombosis. J Thromb Haemost 2009; 7(S1): 301-304.

23. Rosendaal FR. Risk factors for venous thrombosis: prevalence, risk and interaction. Semin Hematol 1997;34:171-187.

24. Jordan W. Pulmonary embolism. Lancet 1961;18:1146-1147.

25. Tyler ET. Oral contraception and venous thrombosis. JAMA 1963;185:131-132.

26. Gerstman BB, Piper JM, Freiman JP, Tomita DK, Kennedy DL, Ferguson WJ, Bennett RC. Oral contraceptive oestrogen and progestin potencies and the incidence of deep venous thromboembolism. Int J Epidemiol 1990;19:931- 936.

27. Effect of different progestagens in low oestrogen oral contraceptives on venous thromboembolic disease: World Health Organization Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Lancet 1995;346:1582-1588.

28. Jick H, Jick S, Gurewich V, Wald Myers M, Vasilakis C. Risk of idiopathic venous thromboembolism in women using oral contraceptives with differing progestagen components. Lancet. 1995;346:1589-1593.

29. Bloemenkamp KW, Rosendaal FR, Helmerhorst FM, Büller HR, Vanden- broucke JP. Enhancement by factor V Leiden mutation of risk of deep-vein thrombosis associated with oral contraceptives containing a third-generation progestagen. Lancet 1995;346:1593-1596.

30. Kemmeren JM, Algra A, Meijers JC, Bouma BN, Grobbee DE. Effect of second- and third-generation oral contraceptives on the protein C system in the absence or presence of the factorVLeiden mutation: a randomized trial. Blood 2004;103:927–933.

30 Introduction

31. Sips AJBI, Beijderwellen L, Kardolus GJ, Burgers JS. Dutch General Practitioner’s Standard on hormonal contraception, first revision (Nederlands Huisartsen Genootschap-Standaard Hormonale Anticonceptie (eerste revisie). Huisarts Wet 1998;41:385-394.

32. van Hylckama V, Helmerhorst FM, Vandenbroucke JP, Doggen CJ, Rosen- daal FR. The venous thrombotic risk of oral contraceptives, effects of oestrogen dose and progestogen type: results of the MEGA case-control study. BMJ 2009;339:b2921.

33. Lidegaard Ø, Løkkegaard E, Svendsen AL, Agger C. Hormonal contraception and risk of venous thromboembolism: national follow-up study. BMJ 2009;33:b2890.

34. Lidegaard Ø, Nielsen LH, Skovlund CW, Løkkegaard E. Venous thrombosis in users of non-oral hormonal contraception: follow-up study, Denmark 2001-10. BMJ 2012;344:e2990.

35. Stegeman BH, de Bastos M, Rosendaal FR, van Hylckama Vlieg A, Helmer- horst FM, Stijnen T, Dekkers OM. Different combined oral contraceptives and the risk of venous thrombosis: systematic review and network meta- analysis. BMJ 2013;347:f5298.

36. de Bastos M, Stegeman BH, Rosendaal FR, Van Hylckama Vlieg A, Helmer- horst FM, Stijnen T, Dekkers OM. Combined oral contraceptives: venous thrombosis. Cochrane Database of Systematic Reviews 2014, Issue 3. Art. No.: CD010813. DOI:10.1002/14651858.CD010813.pub2.

37. Vinogradova Y, Coupland C, Hippisley-Cox J. Use of combined oral contra- ceptives and risk of venous thromboembolism: nested case-control studies using the QResearch and CPRD databases. BMJ 2015;350:h2135.

38. Dinger JC, Heinemann LAJ, and Kuhl-Habichl, D. The safety of drospirenone- containing oral contraceptive: final results from the European Active Surveil lance study on Oral Contraceptives based on 142,475 women-years of obser vation. Contraception 2007;75:344-354.

31 Introduction

39. Sidney S, Cheethamb TC, Connell FA, Ouellet-Hellstrom R, Ouellet- Hellstrom R, Graham DJ, Davis D, Sorela M, Quesenberry CP, Cooper WO. Recent combined hormonal contraceptives (CHCs) and the risk of throm- boembolism and other cardiovascular events in new users. Contraception 2013;87:93-100.

40. RCOGs’ Faculty of sexual & reproductive health care (2014) statement on Venous thromboembolism and hormonal contraception, November 2014. Available at: http://www.fsrh.org/pdfs/FSRHStatementVTEandHormonalContraception.pdf

41. Odlind V, Milsom I, Persson I, Victor A. Can changes in sex hormone binding globulin predict the risk of venous thromboembolism with com- bined oral contraceptive pills? Acta Obstet Gynecol Scand 2002;81:482-490.

42. van Vliet HAAM, Frolich M, Thomassen MCLGD, Doggen CJ, Rosendaal FR, Rosing J, Helmerhorst FM. Association between sex hormone-binding globu- lin levels and activated protein C resistance in explaining the risk of throm- bosis in users of oral contraceptives containing different progestogens. Hum Reprod 2005;20:563–568.

43. Stanczyk FZ, Grimes DA. Sex hormone-binding globulin: not a surrogate marker for venous thromboembolism in women using oral contraceptives. Contraception 2008;78:201-203.

44. Heinemann LAJ, Assmann A, Spannagl M, Schramm W, Dick A, Kluft C, de Maat MPM. Normalized Activated Protein C Ratio itself not associated with increased risk of venous thromboembolism. Contraception 1998;58:321-322.

45. Tans G, van Hylckama Vlieg A, Christella M, Thomassen MLGD, Curvers J, Bertina RM, Rosing J, Rosendaal FR. Activated protein C resistance deter- mined with a thrombin generation-based test predicts for venous thrombo- sis in men and women. Br J Hematol 2003;122:465-470.

46. Tchaikovski S, Rosing J. Mechanisms of Estrogen-Induced Venous Throm- boembolism. Thromb Res 2010;126:5-11.

32 Introduction

47. Middeldorp S, Meijers JC, van den Ende AE, van Enk A, Bouma BN, Tans G, Rosing J, Prins MH, Büller HR. Effects on coagulation of levonorgestrel- and desogestrel-containing low dose oral contraceptives: a cross-over study. Thromb Haemost 2000;84:4-8.

48. Tans G, Curvers J, Middeldorp S, Thomassen MC, Meijers JC, Prins MH, Bouma BN, Büller HR, Rosing J. A randomized cross-over study on the effects of levonorgestrel- and desogestrel-containing oral contraceptives on the anticoagulant pathways. Thromb Haemost 2000;84:15-21.

49. Meijers JC, Middeldorp S, Tekelenburg W, van den Ende AE, Tans G, Prins MH, Rosing J, Büller HR, Bouma BN. Increased fibrinolytic activity during use of oral contraceptives is counteracted by an enhanced factor XI indepen- dent down regulation of fibrinolysis: a randomized cross-over study of two low-dose oral contraceptives. Thromb Haemost 2000;84:9-14.

50. Heit JA, Kobbervig CE, James AH, Petterson TM, MS; Bailey KR, Melton III LJ. Trends in the incidence of venous thromboembolism during pregnancy or postpartum: a 30-year population-based study. Ann Intern Med. 2005;143:697-706.

51. Hellgren M. Haemostasis during normal pregnancy and puerperium. Semi- nars in thrombosis and haemostasis 2003;26:126-130.

52. James AH. Venous Thromboembolism in Pregnancy. Arterioscler Thromb Vasc Biol 2009;29:326-331.

53. Egeberg O. Inherited antithrombin deficiency causing thrombophilia. Thromb Diath Haemorrh 1965;13:516.

54. Griffin JH, Evatt B, Zimmerman TS, Kleiss AJ, Wideman C. Deficiency of protein C in congenital thrombotic disease. J Clin Invest 1981;68:1370-1373.

55. Schwarz HP, Fischer M, Hopmeier P, Batard MA, Griffin JH. Plasma protein S deficiency in familial thrombotic disease. Blood 1984;64:1297-1300.

56. Tait RC, Walker ID, Perry DJ, Islam SI, Daly ME, McCall F, Conkie JA, Carrell RW. Prevalence of antithrombin deficiency in the healthy population. Br J Haematol 1994;87:106-112.

33 Introduction

57. Tait RC, Walker ID, Reitsma PH, Islam SI, McCall F, Poort SR, Conkie JA, Bertina RM. Prevalence of protein C deficiency in the healthy population. Thromb Haemost 1995;73:87-93.

58. Dykes AC, Walker ID, McMahon AD, Islam SI, Tait RC. A study of Protein S antigen levels in 3788 healthy volunteers: influence of age, sex and hormone use, and estimate for prevalence of deficiency state. Br J Haematol 2001;113:636.

59. Martinelli I, Mannucci PM, De Stefano V, Taioli E, Rossi V, Crosti F, Pacia- roni K, Leone G, Faioni EM,. Different risks of thrombosis in four coagula- tion defects associated with inherited thrombophilia: a study of 150 families. Blood 1998;92:2353-2358.

60. Simioni P, Sanson BJ, Prandoni P, Tormene D, Friederich PW, Girolami B, Prins MH. Incidence of venous thromboembolism in families with inherited thrombophilia. Thromb Haemost 1999;81:198-202.

61. Vossen CY, Conard J, Fontcuberta J, Makris M, van der Meer FJM, Pabinger I, Palareti G, Preston FE, Scharrer I, Souto JC, Svensson P, Walker ID, Rosen- daal FR. Familial thrombophilia and lifetime risk of venous thrombosis. J Thromb Haemost 2004; 2:1526-1532.

62. Brouwer JLP, Veeger NJGM, Kluin-Nelemans HC, van der Meer J. Multicau- sal pathogenesis of venous thromboembolism: evidence from a study in families with hereditary deficiencies of protein C, protein S, or antithrombin. Ann Intern Med. 2006;145:807-815.

63. Koster T, Rosendaal FR, Briet E, van der Meer FJM, Colly LP, Trienekens PH, Poort SR. Pieter H. Reitsma PH, Vandenbroucke JP. Protein C Deficiency in a Controlled Series of Unselected Outpatients: An Infrequent But Clear Risk Factor for Venous Thrombosis (Leiden Thrombophilia Study). Blood 1995;85:2756-2761.

64. Pintao MC, Ribeiro DD, Bezemer ID, Garcia AA, de Visser MCH, Doggen CJM, Lijfering WM, Reitsma PH, Rosendaal FR. Protein S levels and the risk of venous thrombosis: results from the MEGA case-control study. Blood 2013;122:3210-3219.

34 Introduction

65. Bertina RM, Koeleman BP, Koster T, Rosendaal FR, Dirven RJ, de Ronde H, van der Velden PA, Reitsma PH. Mutation in blood coagulation factor V associated with resistance to activated protein C. Nature 1994;369:64-67.

66. Poort SR, Rosendaal FR, Reitsma PH, Bertina RM. A common genetic varia- tion in the 3’-untranslated region of the prothrombin gene is associated with elevated plasma prothrombin levels and an increase in venous thrombosis. Blood 1996;88:3698-3703.

67. Rees DC, Cox M, Clegg JB. World distribution of factor V Leiden. Lancet 1995;346:1133-1134.

68. Rosendaal FR, Doggen CJM, Zivelin A, Arruda VR, Aiach M, Siskovick DS, Hillarp H, Watzke HH, Bernardi F, Cumming AM, Reitsma PH. Geographic distribution of the 20210 G to A prothrombin variant. Thromb Haemost 1998;79:706-708.

69. Koster T, Rosendaal FR, De Ronde H, Briët E, Vandenbroucke JP, Bertina RM. Venous thrombosis due to a poor anticoagulant response to activated protein C: Leiden Thrombophilia Study. Lancet 1993;342:1503-1506.

70. Middeldorp S, CM, Koopman MM, van Pampus EC, Hamulyák K, van der Meer, Büller HR. The incidence of venous thromboembolism in family mem- bers of patients with factor V Leiden mutation and venous thrombosis. Ann Intern Med 1998;128:15-20.

71. Rosendaal FR, Koster T, Vandenbroucke JP, Reitsma PH. High risk of throm- bosis in patients homozygous for factor V Leiden (activated protein C resistance). Blood 1995;85:1504-1508.

72. Emmerich J1, Rosendaal FR, Cattaneo M, Margaglione M, De Stefano V, Cumming T, Arruda V, Hillarp A, Reny JL. Combined effect of factor V Leiden and prothrombin 20210A on the risk of venous thromboembolism--pooled analysis of 8 case-control studies including 2310 cases and 3204 controls. Study Group for Pooled-Analysis in Venous Thromboembolism. Thromb Haemost 2001;86:809-816.

73. Pabinger I, Schneider B, and the study GTH group on natural inhibitors. Thrombotic risk of women with hereditary antithrombin III, protein C- and protein S deficiency taking oral contraceptive medication. Thromb Haemost 1994;71:548-552.

35 Introduction

74. de Bruijn SFTM, Stam J, Koopman MMW, Vandenbroucke JP for the Cerebral Venous Sinus Thrombosis Study Group. Case-control study of cerebral sinus thrombosis in oral contraceptive users who are carriers of hereditary prothrombotic conditions. BMJ 1998;316:589-592.

75. Andersen BS, Olsen J, Nielsen GL, Steffensen FH, Sorensen HT, Baech J, Gregersen H. Third generation oral contraceptives and heritable throm- bophilia as risk factors of non-fatal venous thromboembolism. Thromb Haemost 1998;79:28-31.

76. Bennet L, Odeberg H. Resistance to activated protein C, highly prevalent amongst users of oral contraceptives with venous thromboembolism. J Intern Med 1998;244:27-32.

77. Martinelli I, Sacchi E, landi G, Taioli E, Duca F, Manucci PM. High risk of cerebral vein thrombosis in carriers of a prothrombin-gene mutation and in users of oral contraceptives. N Engl J Med 1998;338:1793-1797.

78. Martinelli I, Taioli E, Bucciarelli P, Akhavan S, Manucci PM. Interaction between the G20210A mutation of the prothrombin gene and oral contraceptives use in deep vein thrombosis. Arterioscler Thromb Vasc Biol 1999;19:700-703.

79. Spannagl M, Heinemann LA, Schramm W. Are factor V Leiden carriers who use oral contraceptives at extreme risk for venous thromboembo- lism? Eur J contracept reprod health care. 2000; 5:105-112.

80. Aznar J, Vaya A, Estelles A, MiraY, Segui R, Villa P, Ferrando F, Falco C, Corella D, Espana F. Risk of venous thrombosis in carriers of the prothrombin G20210A variant and factor V Leiden and their interaction with oral contraceptives. Haematologica 2000;85:1271-1276.

81. Bloemenkamp, K.W., Rosendaal, F.R., Helmerhorst, F.M., Vanden broucke, J.P. Higher risk of venous thrombosis during early use of oral contraceptives in women with inherited clotting defects. Arch Intern Med 2000;160:49-52.

82. Legnani C, Palareti G, Guazzaloca G, Cosmi B, Lunghi B, Bernardi F, Coccheri S. Venous thromboembolism in young women: role of throm bophilic mutations and oral contraceptive use. Eur Heart J 2002;23;984-990.

36 Introduction

83. Vaya A, Mira Y, Mateo J, Falco C, Villa P, Estelles A, Aznar J. Prothrombin G20210A mutation and oral contraceptive use increase upper extremity deep vein thrombotic risk. Thromb Haemost. 2003;89:452-457.

84. Martinelli I, Battaglioli T, Buccarelli P, Passamonti SM, Manucci PM. Risk factors and recurrence rate of primary deep vein thrombosis of the upper extremities. Circulation 2004;110:566-570.

85. Ehrenforth S, Nemes L, Mannhalter C, Rosendaal FR, Koder S, Zoghlami-Rintelen C, Pabinger I. Impact of environmental and hereditary risk factors on the clinical manifestation of thrombophilia in homozygous carriers of factor V:1691A. J Thromb Haemost 2004;2:430-436.

86. Sidney S, Petitti DB, Soff GA, Cundiff DL, Tolan KK, Quesenberry CP. Venous thromboembolic disease in users of low-estrogen combined estrogen-progestin oral contraceptives. Contraception 2004;70;3-10.

87. Gadelha T, André C, Jucá AAV, Nucci M. Prothrombin 20210a and oral contraceptive use as risk factors for cerebral venous thrombosis. Cere brovasc Dis 2005;19:49-52.

88. Roach REJ, Lijfering WM, Helmerhorst FM, Cannegieter SC, RosendaalFR, van Hylckama Vlieg. The risk of venous thrombosis in women over 50 years old using oral contraception or postmenopausal hormone therapy. J Thromb Haemost 2013;11:124-131.

89. Tufano A, Guida A, Coppola A, Nardo A, Capua MD, Quintavalle G, Di Minno MND, Cerbone, AM, Di Minno G. Risk factors and recurrent thrombotic episodes in patients with cerebral venous thrombosis. Blood Transfus 2014;12:s337-342.

90. Bergendal A, Persson I, Odeberg J, Sundström A, Holmström M, Schulman S, Björgell O, Kieler H. Association of venous thromboembolism with hormonal contraception and thrombophilic genotypes. Obstet Gynecol 2014;124:600-609.

91. Van Boven HH, Vandenbroucke JP, Briët E, Rosendaal FR. Gene-gene and gene-environment interactions determine the risk of thrombosis in families with inherited antithrombin deficiency. Blood 1999;94:2590-2594.

37 Introduction

92. Santamaria A, Mateo J, Oliver A, Menendez B, Souto J, Borrel M, Fontcu berta J. Risk of thrombosis associated with oral contraceptives of women from 97 families with inherited thrombophilia: high risk of thrombosis in carriers of the G20210A mutation of the thrombin gene. Haematologica 2001;86:965-971.

93. Bezemer ID, van der Meer FJM, Eikenboom JCJ, Rosendaal FR, Doggen CJM. The value of family history as a risk indicator for venous thrombosis. Arch Intern Med 2009;169:610-615.

94. Noboa S, Le Gal, Lacut K, Mercier B, Leroyer C, Nowak E, Mottier D, Oger E: for the EDITH Collaborative Study Group. Family history as a risk factor for venous thromboembolism. Thromb Res 2008;122:624-629.

95. Dowling NF, Austin H, Diley A, Whitsett C, Evatt BL, Hooper WC. The epidemiology of venous thromboembolism in Caucasians and African- Americans: the GATE Study. J Thromb Haemost 2003;1:80-87.

96. Silverstein MD, Heit JA, Mohr DN, Petterson TM, O'Fallon WM, Melton III LJ. Trends in incidence of deep vein thrombosis and pulmonary embolism. Arch Intern Med 1998;158:585-593.

97. Zöller B, Li X, Sundquist J, Sundquist K. Age- and Gender-Specific Familial Risks for Venous Thromboembolism. A Nationwide Epidemiological Study Based on Hospitalizations in Sweden. Circulation 2011;124:1012-1020.

98. Couturaud, F, Leroyer C, Tromeur C, Julian JA, Kahn SR, Ginsberg JS, Wells PS, Douketis JD, Mottier D, Kearon C. Factors that predict throm- bosis in relatives of patients with venous thrombosis. Blood 2014;124:2124-2130.

99. Cushman M, Glynn RJ, Goldhaber SZ, Moll S, Bauer KA, Deitcher S, Shrivastava S, Ridker PM. Hormonal factors and risk of recurrent venous thrombosis: the Prevention of Recurrent Venous Thromboembolism trial. J Thromb Haemost 2006;4:2199-2203.

100. Le Gal G, Kovacs MJ, Carrier M, Do K, Kahn SR, Wells PS, Anderson DA, Chagnon I, Solymoss S, Crowther M, Righini M, Lacut K, White RH, Vickars L, Rodger M. Risk of recurrent venous thromboembolism after a first oestrogen-associated episode. Data from the REVERSE cohort study. Thromb Haemost 2010;104:498-503.

38 Introduction

101. Vaillant-Roussel H, Ouchchane L, Dauphin C, Philippe P, Ruivard M. Risk factors for recurrence of venous thromboembolism associated with the use of oral contraceptives. Contraception 2011;84:e23-30. 102. Le Moigne E, Delluc A, Tromeur C, Nowak E, Mottier D, Lacut K, Le Gal G. Risk of recurrent venous thromboembolism among young women after a first event while exposed to combined oral contraception versus not exposed to: A cohort study. Thromb Res 2013;132:51-55. 103. Eischer L, Eichinger S, Kyrle PA. The risk of recurrence in women with venous thromboembolism while using : a prospective cohort study. J Thromb Haemost 2014;12:635-640. 104. Cosmi B, Legnani C, Bernardi F, Coccheri S, Palareti G. Value of family history in identifying women at risk of venous thromboembolism during oral contraception: observational study. BMJ 2001;322:1024–1025. 105. Vandenbroucke JP, van der Meer FJM, Helmerhorst FM, Rosendaal FR. Family history and risk of venous thromboembolism with oral contracep- tion. BMJ 2001;323:752.

39 40