university of groningen optimal dosing strategy for ...14 general introduction table 1: differences...

19
University of Groningen Optimal dosing strategy for prothrombin complex concentrate Khorsand, Nakisa 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: 2014 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): Khorsand, N. (2014). Optimal dosing strategy for prothrombin complex concentrate. [S.n.]. 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: 07-04-2021

Upload: others

Post on 24-Oct-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

  • University of Groningen

    Optimal dosing strategy for prothrombin complex concentrateKhorsand, Nakisa

    IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

    Document VersionPublisher's PDF, also known as Version of record

    Publication date:2014

    Link to publication in University of Groningen/UMCG research database

    Citation for published version (APA):Khorsand, N. (2014). Optimal dosing strategy for prothrombin complex concentrate. [S.n.].

    CopyrightOther 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 theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

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

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

    Download date: 07-04-2021

    https://research.rug.nl/en/publications/optimal-dosing-strategy-for-prothrombin-complex-concentrate(63c4a083-0828-40bc-97d9-04ad6fe6bb89).html

  • CHaPter 1Introduction

  • 10

    General introduction

    HISTOry OF VITAMIN K ANTAGONISTS

    The mysterious ability of blood to clot has intrigued people over millennia.

    The fascinating story of the discovery of vitamin K antagonists to manipulate

    this clotting begins on the Canadian prairies in 1920s. The story ran as

    follows:1 Previously healthy cattle in these areas died of internal bleeding

    with no obvious cause. Given that livestock was one of the most important

    industries in these areas combined with The Great Depression, this was a

    disaster for the farmers. As there was an apparent lack of a recognizable

    pathological disorder responsible for the haemorrhage, the diet of the

    livestock was questioned. The cattle and sheep had grazed on sweet

    clover hay (Melilotus alba and Melilotus officinalis) and the incidence of

    bleeding occurred most frequently when the climate, and therefore the

    hay, in these areas was damp. Damp hay became infected by moulds such as

    Penicillium nigricans and Penicillium jensi, which appeared to be integral

    in the disease process occurring in the cattle. As Duxbury and Poller point

    out in their review article on warfarin,2 such hay would normally have been

    discarded if it spoiled in storage, but in the financial hardship of the 1920s

    few farmers could afford to buy supplementary fodder for their cattle and

    thus the mouldy hay was used for feed. The resultant haemorrhagic disease

    was called ‘sweet clover disease’ in 1922.

    In 1929, Roderick observed that the affected cattle were deficient in a

    clotting factor, prothrombin.3 At the same time, Dam was investigating a

    severe bleeding condition with similar depletion of prothrombin in hens that

    were maintained on sterol-depleted diets.4 This work led to the discovery

    of vitamin K, for which Dam received the Nobel Prize for Medicine in 1943.

    Another story is very informative:5 Ten years after the original outbreak of

    sweet clover disease, a young Wisconsin farmer, Ed Carlson, angry about

    losing his cows from internal bleeding, drove 200 miles with a dead cow in

    the back of his truck to the local agricultural experimental station.

  • 11

    Chapter 1

    As the investigator Karl Link was the only person left working late, Carlson

    entered Link’s office handing him a milk can of unclotted blood. Link

    experimented with the blood that evening and, as Duxbury and Poller

    comment, ‘The can of uncoagulated blood lying on the floor of Link’s

    laboratory was to change the course of history, and little did Link know

    what the long-term implications would be’.2 Link and colleagues got to

    work on finding the active substance from the spoiled hay causing the

    internal bleed. In 1940, they published the purification and synthesis of

    dicumarol (3,3-methylenebis-9 [4-hydroxycoumarin]), the active component

    in the spoiled sweet clover.6 This agent, referred to as “coumarin”, was

    promptly made available for clinical studies and already one year later first

    experiences on the effectiveness in deep vein thrombosis as well as its

    hemorrhagic complications were published.7-9 At that time, the most potent

    synthesized coumarin, warfarin, was successfully used to fight rats.10

    The first clinical study with administration of coumarins in thromboembolic

    conditions is reported in 1948.11 Not much later, president Eisenhower

    was treated with warfarin following a heart attack.12 By that time, it was

    empirically known that vitamin K reversed the bleeding problem caused

    by coumarins as these agents were also called ‘vitamin K antagonists’.

    However, it took another 2 decades until in 1974 the vitamin K cycle was

    proposed.13, 14 After 30 years the complicating biochemical relation between

    vitamin K, vitamin k-epoxide, and the role of vitamin K antagonists was

    clarified by identification of VKORC1 in Nature in 2004.15, 16

    It is rare for any drug introduced more than 50 years ago to remain

    unsurpassed today, yet millions of patients are still being treated with

    vitamin K antagonists.17 In 1985, a goal was set to develop new oral

    anticoagulants to replace vitamin K antagonists.18 However, due to many

    challenges facing the scientists, novel oral anticoagulants did not enter the

    market until the 21st century.19-21

  • 12

    General introduction

    General hemostasis

    In case of vessel damage, the body reacts fast to prevent blood loss. This

    process, ‘hemostasis’ or ‘coagulation’, involves platelets, coagulation factors

    and several other proteins, all working closely together with the vessel wall

    in a complicated and precisely balanced process (figure 1).22 Briefly, when the

    vessel wall is damaged, tissue factor (TF) is released. This initiates activation

    of factor VII in blood with formation of TF-factor VIIa-complex. This complex

    enables the conversion of factor X into Xa. After its activation, factor Xa

    facilitates the formation of small amounts of thrombin, which is necessary

    to activate platelets and cofactor V and VIII. Activated factor X together with

    activated factor V configures the prothrombinase complex. This complex

    converts prothrombin (factor II) to thrombin (factor IIa). Thrombin initiates

    many pathways. For hemostasis, the most important one is the configuration

    of fibrinogen into a fibrin and subsequently into fibrinclot.

    Figure 1: Coagulation pathway adopted from Borissoff et al, NEJM 2011.22

  • 13

    Chapter 1

    Vitamin K antagonists

    Most coagulation factors are synthesized in the liver. Some of these factors

    need vitamin K as a co-enzyme for their formation.23 These factors are

    coagulation factors II (prothrombin), VII, IX, X, and coagulation inhibiting

    factors Protein C and S.

    For this process, vitamin K is oxidized into vitamin K epoxide. As vitamin K

    is very limitedly available in tissues, the epoxide must rapidly be reduced

    again to vitamin K. An enzyme called ‘vitamin K epoxide reductase complex’

    (VKOr) is needed to facilitate this step.16

    Vitamin K antagonists (earlier referred to as coumarins) block the VKOr

    enzyme and prevent the regeneration of reduced vitamin K.23 Therefore, the

    synthesis of coagulation factors cannot be completed; subsequently these

    factors cannot take part in the coagulation cascade. As this blockage only

    affects the new formation of the clotting factors, the effect of vitamin K

    antagonists is delayed for 72 to 96 hours after the start of treatment.

    To date, different vitamin K antagonist agents are known of which warfarin,

    acenocoumarol, and phenprocoumon are the most widely used. In the

    United States of America and the United Kingdom only warfarin is used,

    whereas in the Netherlands only acenocoumarol and phenprocoumon are

    available. The most important differences of these drugs are summarized

    in table 1.

    The effectiveness of vitamin K antagonists has been established by well-

    designed clinical trials.17, 29 They are applied for the primary and secondary

    prevention of venous thromboembolism, for the prevention of systemic

    embolism in patients with prosthetic heart valves or atrial fibrillation and for

    the prevention of stroke, recurrent infarction or death after acute coronary

    syndrome. For the prevention of stroke in patients with atrial fibrillation,

    vitamin K antagonists showed a 64% reduction of stroke risk compared to

    placebo and 37% risk reduction compared to antiplatelet therapy.30

  • 14

    General introduction

    table 1: Differences in VKA drugs

    Onset period Half-life time Wash out period

    Metabolism27-29

    Warfarin 36-72 hours 40 hours 4-5 days CyP2C9 mainly, also CyP1A2 and CyP3A4

    acenocoumarol 36-48 hours 11 hours 48 hours CyP2C9 mainly, also CyP2C19

    Phenprocoumon 48-72 hours 160 hours 1-2 weeks CyP2C9 (less than acenocoumarol), also

    CyP3A4 and urinary

    and gut excretion

    Vitamin K antagonists all have a narrow therapeutic range as the effective

    dose is close to the minimum and maximum safe dose. Deviating from the

    effective dose to under-treatment increases the risk of thrombosis and to

    over-treatment the risk of bleeding.24 However, finding the right dose is

    difficult because of the large inter- and intra-individual differences in dosage

    requirement for optimal efficacy. The inter- and intra-individual variability

    thereby the range in required dosages can, at least in part, be explained by

    factors such as age, gender, co-morbidity, vitamin K intake, co-medication,

    and genetic variation in its main metabolizing enzyme, cytochrome P450

    isoform CyP2C9.25, 26 This means that the anticoagulant effect in patients on

    vitamin K antagonists needs to be measured regularly and that doses need

    to be adjusted accordingly.

    The level of anticoagulation is historically monitored by a coagulation test

    that measures the prothrombin time (PT). However, because laboratories

    used different reagents for this test, PT results between different

    laboratories were not comparable. Therefore, a standardized expression of

    the level of anticoagulation, the International Normalized ratio (INr) has

    been adopted.31-33 For this, the International Sensitivity Index (ISI) is used,

    which indicates the level of tissue factor in the PT-reagent. When the ISI

    and PT are known, INr is calculated using a formula:

  • 15

    Chapter 1

    The INr, being a ratio, has no units. The normal INr is 1. When using VKA,

    elevated INrs from 2 to 3,5 are desired. Higher INr intensities are associated

    with higher incidences of bleeding complications, whereas lower intensities

    are associated with more thromboembolic events.24, 34

    Complications of vitamin K antagonists

    As its toxicity was discovered before the active therapeutic component

    itself, it is of no surprise that vitamin K antagonists are among the most

    toxic drugs in current medicine. In fact, these drugs are one of the main

    reason for drug-related hospital admissions.35, 36

    Despite many thrombosis services and anticoagulation clinics world-wide

    and rapid INr controls for individual vitamin K antagonist dosage regimens,

    major bleeding complications are still common practice. Intracranial bleeds

    are the most feared complication as these bleeds are most often fatal. Other

    frequently seen major bleeds include gastrointestinal, intra-peritoneal,

    intraocular and muscle bleeds.

    Numerous studies have shown that the incidence of vitamin K antagonist-

    associated major bleeding is 0.4-7.2% per year.37-39 This wide range is

    considered to be the result of the many patient-related factors that can

    alter the pharmacokinetics and pharmacodynamics of vitamin K antagonists.

    In addition, earlier studies often had different definitions of major

    bleed. More recent studies have been more consistent and usually define

    major bleed as proposed by the International Society of Thrombosis and

    Haemostasis.40 According to this, a major bleed is defined as a fatal bleed,

    and/or a symptomatic bleed in a critical area or organ, such as intracranial,

    intraspinal, intraocular, retroperitoneal, intra-articular or pericardial, or

    intramuscular with compartment syndrome, and/or bleeding that causes

    a fall in hemoglobin level of 20 g L-1 (1.24 mmol L-1) or more, or leading to

    transfusion of two or more units of whole blood or red blood cells.

    In recent years, several well-designed studies have been conducted due to

    the discovery of novel oral anticoagulants and their effectiveness compared

  • 16

    General introduction

    to the well-established vitamin K antagonists. Throughout these studies,

    knowledge has increased about incidences of major, intracranial and fatal

    bleeds in the studied populations. In table 2 these incidences are shown.

    table 2: incidences of different VKA-associated bleeds obtained from novel oral anticoagulant studies

    Gastrointestinal Major Intracranial Fatal

    reLY19 1.02%/year 3.36%/year 0.74%/year -

    rocket-aF20 2.2%/year 3.4%/year 0.7%/year 0.5%/year

    aristotle21 0.86%/year 3.09%/year 0.80%/year -

    re-Cover41 2.8% 1.9% 0.24% 0.08%

    einstein42 - 1.2%* 0.3%* -

    * per treatment period resulting in lower percentages than other studies

    treatment of vitamin K antagonist-associated bleeds

    Management of vitamin K antagonist-associated bleeds depends mainly on

    the severity of the bleed and is still a common clinical challenge.

    In addition to hemodynamic stabilization, and possible surgical, endoscopic

    or other interventional control, the anticoagulant effect of vitamin K

    antagonist must be reversed.17 This can be accomplished by withholding

    vitamin K antagonist therapy. In this case, it takes 3 to 5 days, depending on

    the vitamin K antagonist (table 1), before the vitamin K cycle is restored and

    the vitamin K antagonist effect is diminished. In case a more rapid reversal

    is needed, vitamin K can be replaced by its administration. This allows

    the liver to directly start producing normally carboxylized clotting factors.

    By administering vitamin K, the vitamin K antagonist effect is diminished

    within 24 hours. However, for a longer-term correction of the coagulopathy,

    daily administration of vitamin K is necessary.

    For an urgent, direct reversal of vitamin K antagonists, the depleted vitamin

    K dependent clotting factors as described above, should be administered.

    This could either be done by infusion of fresh frozen plasma in which clotting

    factors are present or by transfusion of prothrombin complex concentrates,

    which contain clotting factors II, IX and X and a variable amount of factor

  • 17

    Chapter 1

    VII (see below). Prothrombin complex concentrates usage is associated with

    several benefits when compared to fresh frozen plasma, of which the lower

    volume of prothrombin complex concentrate43 and the rapid correction

    of the INr resulting to better clinical outcome44 are the most important

    ones. Currently, the American College of Chest Physicians (ACCP) guidelines

    recommend the use of prothrombin complex concentrates rather than

    plasma for reversal of VKA-induced coagulopathy in patients with a vitamin

    K antagonist-associated major bleeding.45

    Prothrombin Complex Concentrate

    Prothrombin Complex Concentrates are human plasma-derived products

    containing clotting factors II, IX, X, and VII. In addition to these so-called

    4 factor concentrates, 3 factor concentrates are available that contain

    far less factor VII. Furthermore, all prothrombin complex concentrates

    contain variable amounts of natural anticoagulant proteins C and S, and

    anti-thrombin. Different prothrombin complex concentrates are available

    worldwide. These agents and their content, related to 100 units of factor IX

    are summarized in table 3. In the Netherlands only 4 factor PCCs are used.

    The safety of prothrombin complex concentrates has been subject to

    research for several decades as well. A recent meta-analysis reported the

    incidence of thromboembolic events to be +1,8% in patients after receiving

    four-factor prothrombin complex concentrates.47 It is also believed that

    the thrombogeneity of prothrombin complex concentrates increases with

    higher doses.48

  • 18

    General introduction

    tabl

    e 3:

    Pro

    thro

    mbi

    n co

    mpl

    ex c

    once

    ntra

    tes

    and

    thei

    r co

    nten

    t re

    late

    d to

    100

    IU o

    f fa

    ctor

    IX a

    dopt

    ed f

    rom

    Wor

    ld F

    eder

    atio

    n of

    Hem

    o-ph

    ilia

    regi

    ster

    of

    clot

    ting

    fac

    tor

    conc

    entr

    ates

    46 a

    nd t

    he a

    vaila

    ble

    Sum

    mar

    y of

    Pro

    duct

    Cha

    ract

    eris

    tics

    (SP

    C)

    Bran

    d na

    me

    Man

    ufac

    ture

    r3

    or 4

    fac

    tor

    Fact

    or II

    Fa

    ctor

    VII

    Fact

    or IX

    Fa

    ctor

    XO

    ther

    Beri

    plex

    CSL

    Behr

    ing,

    Ger

    man

    y

    4F12

    868

    100

    152

    Prot

    ein

    C, h

    epar

    in,

    anti

    thro

    mbi

    n an

    d

    albu

    min

    Cofa

    ctSa

    nqui

    n,

    Net

    herl

    ands

    4F56

    -140

    28-8

    010

    056

    -140

    Prot

    ein

    S an

    d C

    and

    anti

    thro

    mbi

    n

    Kask

    adil

    LFB,

    Fra

    nce

    4F14

    840

    100

    160

    Hep

    arin

    Oct

    aple

    xO

    ctap

    harm

    a,

    Aust

    ria

    & F

    ranc

    e

    4F44

    -152

    36-9

    610

    050

    Prot

    ein

    C &

    S an

    d

    hepa

    rin,

    low

    acti

    vate

    d fa

    ctor

    VII

    PPSB

    -Ht

    Nic

    hiya

    kuN

    ichi

    yaku

    , Ja

    pan

    4F10

    010

    010

    010

    0Pr

    otei

    n C

    Profi

    linG

    rifo

    ls,

    USA

    3F14

    8Lo

    w10

    064

    -

    PtX-

    VFCS

    L Bi

    opla

    sma,

    Aust

    ralia

    3F10

    0-

    100

    100

    -

    Um

    anKe

    drio

    n, It

    aly

    3F10

    0-

    100

    80An

    tith

    rom

    bin

    and

    hepa

    rin

  • 19

    Chapter 1

    The efficacy of prothrombin complex concentrate is well-established. Also,

    its safety has been subject to many trials. However, its safety and efficacy

    have never been investigated in relation to the optimal dose. Ideally, optimal

    dose of prothrombin complex concentrate is the minimum effective dose as

    its thrombogeneity is supposed to increase with the dosage. Furthermore,

    the optimal, minimum effective dose is cost-effective as this low dosage

    should not lead to any other additional interventions.

    In this thesis we aimed to find the optimal prothrombin complex concentrate

    dose while addressing its effectiveness, safety and costs.

    SCOPE OF THE THESIS

    In this thesis, we address the use of prothrombin complex concentrates in

    emergency reversal of vitamin K antagonists. In this, we focus on finding

    the optimal cost-effective and safe prothrombin complex concentrate dose

    strategy in vitamin K antagonist taking population who need prothrombin

    complex concentrate for acute reversal of vitamin K antagonists.

    In chapter 2, by means of a pilot, we explored the feasibility of a simple,

    low fixed dose prothrombin complex concentrate treatment strategy.

    In chapter 3, we prospectively enrolled consecutive patients with a major

    vitamin K antagonist associated bleed who were either treated by the

    earlier mentioned low fixed dose or a more widely used variable, INR and

    weight dependent, prothrombin complex concentrate dosing strategy.

    Subsequently, as we found the fixed dose to be non-inferior to the variable

    dosing strategy in terms of clinical outcome, an important question from

    both a clinical and costing point of view rose about whether additional

    interventions were needed in the fixed dose cohort to reach the non-

    inferior outcome. We studied this by performing a cost-study which is

    reported in chapter 4.

  • 20

    General introduction

    A review of the literature is performed in chapter 5 to assess the currently

    used prothrombin complex concentrate strategies for emergency vitamin

    K antagonist reversal and to present their efficacy in terms of target INR

    achievement and clinical outcome.

    Finally, in chapter 6, patients, who survived the vitamin K antagonist

    related major bleed in the study described in chapter 3, were followed

    to determine their long term outcome in terms of incidences of (fatal)

    thromboembolism, recurrent (fatal) bleed and mortality. In addition, we

    characterized patients who either continued or discontinued the vitamin K

    antagonist therapy.

  • 21

    Chapter 1

    rEFErENCES

    1. Schofield FS. A brief account of a disease in cattle simulating hemorrhagic

    septicemiadue to feeding sweet clover. Canad Vet Rec. 1922;3:74-78.

    2. Duxbury BM, Poller L. The oral anticoagulant saga: Past, present, and future. Clin

    Appl Thromb Hemost. 2001;7(4):269-275.

    3. roderick LM. The pathology of sweet clover disease in cattle. J Am Vet Med Assoc.

    1929;74:314-325.

    4. Dam H. Antihemorrhagic vitamin of the chick: occurence and chemical nature. .

    Nature. 1935;136:31.

    5. Wardrop D, Keeling D. The story of the discovery of heparin and warfarin. Br J

    Haematol. 2008;141(6):757-763. doi: 10.1111/j.1365-2141.2008.07119.x; 10.1111/

    j.1365-2141.2008.07119.x.

    6. Campbell HA, roberts WL, Smith WK, Link KP. Studies on the hemorrhagic sweet

    clover disease I. the preparation of hemorrhagic concentrates. J Biol Chem.

    1940;136:47-55.

    7. Lehmann J. Hypoprothrombinaemia produced by methylene-bis-(hydroxycoumarin),

    its use in thrombosis. Lancet. 1942;1:318.

    8. Butsch WC, Stewart JD. Clinical experiences with dicoumarin, 3,3’-methylene-bis-

    (4-hydroxycoumarin). JAMA. 1942;120:1025-1026.

    9. Allen EV, Barker NW, Waugh JM. A preparation from spoiled sweet clover

    (3,3’-methylene-bis-[4-hydroxycoumarin]) which prolongs coagulation and pro-

    thrombin time of the blood: A clinical study. JAMA. 1942;120:1009-1015.

    10. Link KP. The discovery of dicumarol and its sequels. Circulation. 1959;19:97-107.

    11. Wright IS. Experience with dicoumarol in the treatment of coronary thrombosis with

    myocardial infarction. Am Heart J. 1948;32:20-31.

    12. Scully M. Warfarin therapy: rat poison and prevention of thrombosis. The biochemist.

    2002:15-17.

    13. Willingham AK, Matschiner JT. Changes in phylloquinone epoxidase activity related

    to prothrombin synthesis and microsomal clotting activity in the rat. Biochem J.

    1974;140(3):435-441.

    14. Sadowski JA, Suttie JW. Mechanism of action of coumarins. significance of vitamin K

    epoxide. Biochemistry. 1974;13(18):3696-3699.

  • 22

    General introduction

    15. rost S, Fregin A, Ivaskevicius V, et al. Mutations in VKOrC1 cause warfarin resistance

    and multiple coagulation factor deficiency type 2. Nature. 2004;427(6974):537-541.

    doi: 10.1038/nature02214.

    16. Li T, Chang CY, Jin DY, Lin PJ, Khvorova A, Stafford DW. Identification of the gene

    for vitamin K epoxide reductase. Nature. 2004;427(6974):541-544. doi: 10.1038/

    nature02254.

    17. Ageno W, Gallus AS, Wittkowsky A, et al. Oral anticoagulant therapy: Antithrombotic

    therapy and prevention of thrombosis, 9th ed: American college of chest physicians

    evidence-based clinical practice guidelines. Chest. 2012;141(2 Suppl):e44S-88S.

    doi: 10.1378/chest.11-2292; 10.1378/chest.11-2292.

    18. Gustafsson D, Bylund r, Antonsson T, et al. A new oral anticoagulant: The 50-year

    challenge. Nat Rev Drug Discov. 2004;3(8):649-659. doi: 10.1038/nrd1466.

    19. Wallentin L, Yusuf S, Ezekowitz MD, et al. Efficacy and safety of dabigatran

    compared with warfarin at different levels of international normalised ratio control

    for stroke prevention in atrial fibrillation: An analysis of the RE-LY trial. Lancet.

    2010;376(9745):975-983. doi: 10.1016/S0140-6736(10)61194-4; 10.1016/S0140-

    6736(10)61194-4.

    20. Patel Mr, Mahaffey KW, Garg J, et al. rivaroxaban versus warfarin in nonvalvular

    atrial fibrillation. N Engl J Med. 2011;365(10):883-891. doi: 10.1056/NEJMoa1009638;

    10.1056/NEJMoa1009638.

    21. Agnelli G, Buller Hr, Cohen A, et al. Oral apixaban for the treatment of acute

    venous thromboembolism. N Engl J Med. 2013;369(9):799-808. doi: 10.1056/

    NEJMoa1302507; 10.1056/NEJMoa1302507.

    22. Borissoff JI, Spronk HM, ten Cate H. The hemostatic system as a modulator

    of atherosclerosis. N Engl J Med. 2011;364(18):1746-1760. doi: 10.1056/

    NEJMra1011670; 10.1056/NEJMra1011670.

    23. Stafford DW. The vitamin K cycle. J Thromb Haemost. 2005;3(8):1873-1878. doi:

    10.1111/j.1538-7836.2005.01419.x.

    24. Cannegieter SC, rosendaal Fr, Wintzen Ar, van der Meer FJ, Vandenbroucke JP,

    Briet E. Optimal oral anticoagulant therapy in patients with mechanical heart

    valves. N Engl J Med. 1995;333(1):11-17. doi: 10.1056/NEJM199507063330103.

    25. Teichert M, van Schaik rH, Hofman A, et al. Genotypes associated with reduced activity

    of VKORC1 and CYP2C9 and their modification of acenocoumarol anticoagulation

    during the initial treatment period. Clin Pharmacol Ther. 2009;85(4):379-386. doi:

    10.1038/clpt.2008.294; 10.1038/clpt.2008.294.

  • 23

    Chapter 1

    26. Federatie van Nederlandse Thrombosediensten. De Kunst Van Het Doseren. 2nd

    version ed. Voorschoten: ; 2011.

    27. Beinema M, Brouwers Jr, Schalekamp T, Wilffert B. Pharmacogenetic differences

    between warfarin, acenocoumarol and phenprocoumon. Thromb Haemost.

    2008;100(6):1052-1057.

    28. Ufer M. Comparative pharmacokinetics of vitamin K antagonists: Warfarin,

    phenprocoumon and acenocoumarol. Clin Pharmacokinet. 2005;44(12):1227-1246.

    doi: 10.2165/00003088-200544120-00003.

    29. Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G. Pharmacology and

    management of the vitamin K antagonists: American college of chest physicians

    evidence-based clinical practice guidelines (8th edition). Chest. 2008;133(6 SUPPL.

    6):160S; 198S.

    30. Hart rG, Pearce LA, Aguilar MI. Meta-analysis: Antithrombotic therapy to prevent

    stroke in patients who have nonvalvular atrial fibrillation. Ann Intern Med.

    2007;146(12):857-867.

    31. WHO expert committee on biological standardization. thirty-third report. World

    Health Organ Tech Rep Ser. 1983;687:1-184.

    32. Hirsh J, Deykin D, Poller L. “Therapeutic range” for oral anticoagulant therapy.

    Chest. 1986;89(2 Suppl):11S-15S.

    33. Palareti G, Coccheri S, Poggi M, et al. Oral anticoagulant therapy control: Evidence

    that INr expression improves the inter-laboratory comparability of results--the

    bologna oral anticoagulant control exercise. Thromb Haemost. 1987;58(3):905-910.

    34. rosendaal Fr, Cannegieter SC, van der Meer FJ, Briet E. A method to determine

    the optimal intensity of oral anticoagulant therapy. Thromb Haemost. 1993;69(3):

    236-239.

    35. Leendertse AJ, Egberts AC, Stoker LJ, van den Bemt PM, HArM Study Group.

    Frequency of and risk factors for preventable medication-related hospital admissions

    in the netherlands. Arch Intern Med. 2008;168(17):1890-1896. doi: 10.1001/

    archinternmed.2008.3; 10.1001/archinternmed.2008.3.

    36. van der Hooft CS, Dieleman JP, Siemes C, et al. Adverse drug reaction-related

    hospitalisations: A population-based cohort study. Pharmacoepidemiol Drug Saf.

    2008;17(4):365-371. doi: 10.1002/pds.1565; 10.1002/pds.1565.

  • 24

    General introduction

    37. Palareti G, Leali N, Coccheri S, et al. Bleeding complications of oral anticoagulant

    treatment: An inception-cohort, prospective collaborative study (ISCOAT). italian

    study on complications of oral anticoagulant therapy. Lancet. 1996;348(9025):

    423-428.

    38. Spencer FA, Gore JM, Lessard D, Douketis JD, Emery C, Goldberg rJ. Patient

    outcomes after deep vein thrombosis and pulmonary embolism: The worcester

    venous thromboembolism study. Arch Intern Med. 2008;168(4):425-430. doi:

    10.1001/archinternmed.2007.69; 10.1001/archinternmed.2007.69.

    39. van der Meer FJ, rosendaal Fr, Vandenbroucke JP, Briet E. Bleeding complications

    in oral anticoagulant therapy. an analysis of risk factors. Arch Intern Med.

    1993;153(13):1557-1562.

    40. Schulman S, Kearon C, Subcommittee on Control of Anticoagulation of the Scientific

    and Standardization Committee of the International Society on Thrombosis and

    Haemostasis. Definition of major bleeding in clinical investigations of antihemostatic

    medicinal products in non-surgical patients. J Thromb Haemost. 2005;3(4):692-694.

    doi: 10.1111/j.1538-7836.2005.01204.x.

    41. Schulman S, Kearon C, Kakkar AK, et al. Dabigatran versus warfarin in the treatment

    of acute venous thromboembolism. N Engl J Med. 2009;361(24):2342-2352. doi:

    10.1056/NEJMoa0906598; 10.1056/NEJMoa0906598.

    42. EINSTEIN-PE Investigators, Buller Hr, Prins MH, et al. Oral rivaroxaban for the

    treatment of symptomatic pulmonary embolism. N Engl J Med. 2012;366(14):1287-

    1297. doi: 10.1056/NEJMoa1113572; 10.1056/NEJMoa1113572.

    43. Key NS, Negrier C. Coagulation factor concentrates: Past, present, and future.

    Lancet. 2007;370(9585):439-448. doi: 10.1016/S0140-6736(07)61199-4.

    44. Sarode R, Milling TJ,Jr, Refaai MA, et al. Efficacy and safety of a 4-factor

    prothrombin complex concentrate in patients on vitamin K antagonists presenting

    with major bleeding: A randomized, plasma-controlled, phase IIIb study. Circulation.

    2013;128(11):1234-1243. doi: 10.1161/CIrCULATIONAHA.113.002283; 10.1161/

    CIrCULATIONAHA.113.002283.

    45. Holbrook A, Schulman S, Witt DM, et al. Evidence-based management of anticoagulant

    therapy: Antithrombotic therapy and prevention of thrombosis, 9th ed: American

    college of chest physicians evidence-based clinical practice guidelines. Chest.

    2012;141(2 Suppl):e152S-84S. doi: 10.1378/chest.11-2295; 10.1378/chest.11-2295.

    46. Brooker M. registry of clotting factor concentrates, world federation of hemophilia,

    ninth edition http://www1.wfh.org/publications/files/pdf-1227.pdf.

  • 25

    Chapter 1

    47. Dentali F, Marchesi C, Pierfranceschi MG, et al. Safety of prothrombin complex

    concentrates for rapid anticoagulation reversal of vitamin K antagonists. A meta-

    analysis. Thromb Haemost. 2011;106(3):429-438. doi: 10.1160/TH11-01-0052;

    10.1160/TH11-01-0052.

    48. Dager WE. Using prothrombin complex concentrates to rapidly reverse oral

    anticoagulant effects. Ann Pharmacother. 2011;45(7-8):1016-1020. doi: 10.1345/

    aph.1Q288; 10.1345/aph.1Q288.