ArticlePDF AvailableLiterature Review

Abstract

Introduction: Fifty-three years after the first description of an inherited prothrombotic condition (antithrombin deficiency), our knowledge on hereditary and acquired causes of hypercoagulability that can predispose carriers to venous thromboembolism (VTE) has greatly improved. Areas covered: Main causes of hereditary thrombophilia are summarized alongside new prothrombotic mutations recently discovered. The main causes of acquired thrombophilia, and namely, antiphospholipid antibody syndrome and hyperhomocysteinemia, are also discussed together with other common acquired prothrombotic states characterized by an increase of procoagulant factors and/or a decrease of natural anticoagulants. Finally, suggestions for thromboprophylaxis in carriers of hereditary thrombophilia according to current guidelines/evidence are made for the most challenging high-risk situations (i.e. surgery, pregnancy, contraception, cancer, economy class syndrome) as well as for the prevention of post-thrombotic syndrome. Expert opinion: A carrier of inherited thrombophilia should be evaluated in the framework of other (genetic and/or acquired) coexisting risk factors for first or recurrent VTE when assessing the need and duration of prevention (primary prophylaxis). Prevention strategies should be tailored to each patient and every situational risk factor. The knowledge of the carriership status of severe thrombophilia in the proband can be important to provide asymptomatic relatives with adequate counseling on thrombophilia screening or primary thromboprophylaxis.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=ierr20
Expert Review of Hematology
ISSN: 1747-4086 (Print) 1747-4094 (Online) Journal homepage: https://www.tandfonline.com/loi/ierr20
Thrombophilia, risk factors and prevention
Elena Campello, Luca Spiezia, Angelo Adamo & Paolo Simioni
To cite this article: Elena Campello, Luca Spiezia, Angelo Adamo & Paolo Simioni
(2019): Thrombophilia, risk factors and prevention, Expert Review of Hematology, DOI:
10.1080/17474086.2019.1583555
To link to this article: https://doi.org/10.1080/17474086.2019.1583555
Accepted author version posted online: 18
Feb 2019.
Submit your article to this journal
View Crossmark data
Accepted Manuscript
Publisher: Taylor & Francis
Journal: Expert Review of Hematology
DOI: 10.1080/17474086.2019.1583555
Thrombophilia, risk factors and prevention
Elena Campello, Luca Spiezia, Angelo Adamo, Paolo Simioni*
Haemorrhagic and Thrombotic Diseases Unit, Department of Medicine (DIMED), Padova
University Hospital, Italy
*Corresponding author:
Paolo Simioni,
Haemorrhagic and Thrombotic Diseases Unit, Department of Medicine, Padova University
Hospital, Via Ospedale Civile 105, 35100 Padova, Italy
Telephone (+39) 049 8212667
Fax (+39) 049 8212651
Email: paolo.simioni@unipd.it
Accepted Manuscript
Abstract
Introduction: Fifty-three years after the first description of an inherited prothrombotic
condition (antithrombin deficiency), our knowledge on hereditary and acquired causes of
hypercoagulability that can predispose carriers to venous thromboembolism (VTE) has
greatly improved.
Areas covered: Main causes of hereditary thrombophilia are summarized alongside new
prothrombotic mutations recently discovered. The main causes of acquired thrombophilia,
and namely, antiphospholipid antibody syndrome and hyperhomocysteinemia, are also
discussed together with other common acquired prothrombotic states characterized by an
increase of procoagulant factors and/or a decrease of natural anticoagulants. Finally,
suggestions for thromboprophylaxis in carriers of hereditary thrombophilia according to
current guidelines/evidence are made for the most challenging high-risk situations (i.e.
surgery, pregnancy, contraception, cancer, economy class syndrome) as well as for the
prevention of post-thrombotic syndrome.
Expert opinion: A carrier of inherited thrombophilia should be evaluated in the framework
of other (genetic and/or acquired) coexisting risk factors for first or recurrent VTE when
assessing the need and duration of prevention (primary prophylaxis). Prevention strategies
should be tailored to each patient and every situational risk factor. The knowledge of the
carriership status of severe thrombophilia in the proband can be important to provide
asymptomatic relatives with adequate counseling on thrombophilia screening or primary
thromboprophylaxis.
Keywords: deep vein thrombosis, inherited and acquired thrombophilia, pregnancy,
pulmonary embolism, thromboprophylaxis, venous thromboembolism.
Accepted Manuscript
1. Introduction
In 1856, Virchow postulated alterations in the blood hypercoagulability as one of the
pathomechanisms that can lead to thrombosis [1]. Nowadays, the term “hypercoagulable
state” is used as synonym of pre-thrombotic state even though the majority of patients with
hypercoagulability will never develop thrombosis. The genetic mutations responsible for
inherited hypercoagulability were mainly studied in families with a high incidence of
thrombotic manifestations [2]. Acquired diseases may also result in coagulation changes
that may predispose to a thrombotic tendency. Thrombophilia may therefore be defined as
either an inherited or acquired abnormality of the haemostasis predisposing the affected
subjects to venous and/or arterial thrombosis [3, 4] (Table 1). The focus of this review is to
evaluate the current knowledge on inherited and acquired thrombophilia and to summarize
of the main pathophysiologic pathways and prevention strategies for thrombosis.
Anticoagulant therapy and secondary presentation in patients with thrombophilia will not
be treated.
2. Inherited thrombophilia
Inherited thrombophilia is a genetic propensity to develop venous thromboembolism
(VTE). The most frequent causes are the factor V Leiden and the prothrombin gene
mutation G20210A, accounting for about 50% to 70% of the diagnosed genetic
thrombophilia. The less frequent but more severe defects of antithrombin (AT), protein C
(PC) and protein S (PS) account for most of the remaining cases of diagnosed genetic
thrombophilia [5, 6, 7, 8]. More recently, new genetic defects responsible for severe
thrombophilia have been identified, and namely, pseudo-homozygosity for activated
protein C (APC) resistance, the hyperfunctional factor IX Padua, and the resistance to AT
[9, 10, 11]. And last but not least, ABO blood group is the most common genetic risk factor
for VTE [12]. Figure 1 summarizes known and currently diagnosed hereditary
thrombophilias. There is a large number of families (about 30-40%) with symptomatic
thrombophilia in which none of the known inherited conditions is identified. This
Accepted Manuscript
unexplained thrombophilia is very likely to be due to genetic mutations that are still
unknown.
2.1 Most frequent inherited thrombophilias – Factor V Leiden and prothrombin G20210A
mutations
Factor V Leiden (FVL) and prothrombin G20120A are inherited mutations resulting in the
so-called “gain-of function” for the correspondent procoagulant factors. FVL is an
autosomal dominant disorder related to a missense mutation in the F5 gene at G1691A
which substitutes the amino acid glutamine for arginine at position 506 (R506Q) [13]. This
missense mutation (Arg506Gln) causes factor V resistance to inactivation by APC [14].
Several point mutations in the F5 gene causing APC resistance have been identified in
different populations, including Arg306Thr (FV Cambridge), Arg306Gly (FV Hong
Kong), Ile359Thr (FV Liverpool), Glu666Asp (mechanism unknown), and Ala512Val
(FV Bonn) [15, 16, 17]. Nevertheless, FVL is responsible for about 95% of cases of APC
resistance. FVR2 (H1299R) also exhibits reduced APC cofactor activity and can cause
mild APC resistance, showing a lower thrombotic risk as compared to FVL [15, 18]. The
prothrombin mutation — G to A nucleotide change at 20210 in the 3'-untranslated region
of the prothrombin (F2) gene (PT G20210A) — leads to an impaired F2 3’-end cleavage
signal, resulting in RNA accumulation and thus, increased prothrombin synthesis [19]. This
mutation can increase plasma levels of prothrombin by approximately 30% in
heterozygotes and 70% in homozygotes. However, in many cases the presence of this
mutation is not associated with increased levels of prothrombin and the reverse is also
true. PT G20210A predisposes to VTE either by promoting thrombin generation or by
inhibiting factor Va inactivation by APC, thus creating indirectly APC resistance [20, 21].
Other still undefined mechanisms could contribute to the risk of thrombosis in carriers of
PT G20210A including other genetic lesions which could be in linkage disequilibrium with
PT G20210A mutation. From the clinical point of view, FVL currently represents the most
common known hereditary defect predisposing to VTE: heterozygotes showed an
increased annual risk of VTE from 1 in 1,000 to 3-8 in 1,000, whereas homozygotes
exhibited an annual risk of 80 in 1,000 [22, 23]. As for PT G20210A, heterozygous carriers
present with an increased risk for VTE, with a relative risk (RR) of 2 to 3, overall slightly
lower than the risk associated with FVL [24, 25, 26]. In homozygous individuals the risk
has not clearly defined yet but in a recent study a RR of 5 (95% CI 2.1–11.92) was shown
compared to non-carriers [25]. Interestingly enough, carriers of both mutations (double
heterozygous for FVL and prothrombin mutation) have been reported to present with a six-
Accepted Manuscript
time higher VTE risk than their non-carrier relatives, even though a recent meta-analysis
found the risk of double heterozygosity to be similar to that observed in FVL alone [27, 28].
These discrepancies may be related mainly to the selection of the populations and to
different study design.
2.2 Most severe inherited thrombophilia – loss-of-function of endogenous anticoagulants
Antithrombin is one of the most important natural anticoagulants of the coagulation system
that inhibits several activated factors including thrombin, factor IXa, Xa, XIa and XIIa.
Heterozygous mutations in the AT gene can result in AT defects that are characterized by
reduced inhibition of factor Xa and thrombin and, as a consequence, by increased
thrombin generation and activity [2]. More than 250 genetic lesions have been identified in
patients with AT defects, including missense and nonsense point mutations, small or large
insertions and deletions [29]. The two more common types of defects are the so-called
“Type I”, a quantitative defect resulting in a decreased synthesis of a functional protein;
and Type II, a qualitative defect with normal levels of an abnormal protein with impaired
functional activity. Type II is further divided into three subtypes related to impair binding to
the enzyme reactive sites; subtype IIb is due to mutations in the heparin-binding sites;
subtype IIc is characterized by pleiotropic defects affecting both antigen concentration and
heparin binding site or binding to enzyme reactive sites [29, 30]. Cases of homozygous AT
defects were rarely reported so far mainly involving the heparin-binding site, indicating that
homozygosity for type I or other type II subtypes is associated with embryonic lethality [31,
32]. The absolute risk of a first VTE in asymptomatic carriers of AT deficiency belonging to
families with a symptomatic proband is approximately 2% per year [26, 30]. A large
population-based study showed that AT deficiency was associated with a five-fold (95% CI
0.7– 34) increased risk of a first episode of deep venous thrombosis [33].
Protein C and protein S are vitamin K-dependent glycoproteins synthesized in the liver.
The activated form of protein C (APC) is an important natural anticoagulant that, along with
its co-factor PS, decreases thrombin generation by inhibiting factors Va and VIIIa [34].
Therefore, inherited deficiencies of each of the two determine increased thrombin
generation and predisposition to thrombosis. Protein C deficiency is inherited as an
autosomal dominant trait, and is classified into type I (quantitative) and type II (qualitative)
defects [4, 35]. Unlike AT deficiency, homozygous or double heterozygous PC deficiency
does not determine fetal death, but newborns with these severe thrombophilic conditions
may develop purpura fulminans characterized by severe thrombosis of small dermal
Accepted Manuscript
vessels, resulting in the ischaemic necrosis of extremities [36]. More than 200 mutations,
mostly missense, in the PC gene (PROC) have been described so far responsible for the
different types of defects [37].
Protein S circulates in a free form (~40%) or bound to complement C4b-binding protein
(~60%). Bound protein S has no protein C co-factor activity [38]. Inherited PS deficiency
has been classified as type I (low plasma levels of total and free antigen as well as
reduced functional activity), type II (normal plasma levels of total and free antigen and
reduced functional activity; or dysfunctional PS), and type III (normal or near normal
plasma levels of total antigen and reduced free antigen and functional activity) [2, 4]. More
than 200 different mutations in the PROS1 gene responsible for PS defects have been
identified, mostly being missense mutations or short deletions or insertions [4]. From the
clinical point of view, heterozygous PC deficiency is associated with a 4- to 6.5-fold
increased risk of VTE, while homozygous deficiency results in severe thrombotic
complications in the foetus, neonates or children, as mentioned previously [26]. The risk of
VTE associated with PS deficiency is still a matter of debate: family studies have
suggested a risk comparable to that observed in PC deficiency, but population studies
suggest a lower risk [26]. As in PC deficiency, homozygous PS deficiency may cause
severe purpura fulminans in neonates.
2.3 ABO blood group
Non-O blood group can be considered the most frequent genetic risk factor associated
with VTE. The prothrombotic effect is probably carried by increased levels of von
Willebrand factor (VWF) and factor VIII, as mean levels of these factors are ~25% higher
in individuals with non-O blood groups than in those with O-blood group [39]. The
underlying mechanism for high VWF levels is the addition of A and B antigens to the
specific H oligosaccharides of VWF by means of glycosyltransferase enzymes, resulting in
increased plasma levels of this protein. Additionally, changes in the sugar composition of
VWF due to different ABO alleles affect the rate of cleavage by the VWF-cleaving protease
ADAMTS-13 [40]. However, other factors might also be involved, since non-O blood
groups remained significantly associated with the risk of VTE even after adjustment for
factor VIIII levels [4]. Many clinical trials have previously documented a higher risk of VTE
in non-O blood subjects. Wu and colleagues performed a meta-analysis which selected
4709 VTE cases from 21 studies, and found an OR of 1.79 (95% confidence interval [CI]
Accepted Manuscript
1.56-0.05) in non-O versus O status [12]. A further meta-analysis considering 38 studies,
hence 10,305 VTE cases, reported that non-O blood groups increase approximately
twofold the risk of VTE (OR 2.08; 95%CI 1.83–2.37) [41]. Furthermore, the hazard of VTE
in individuals with hereditary thrombophilia and with non-O blood groups is up to 23.2-fold
higher than that observed in those without thrombophilia and with O blood group [42].
2.4 Novel gain-of-function mechanisms
It can happen that heterozygous FVL carriers present with a concomitant heterozygous F5
gene mutation responsible for FV deficiency, resulting in the 50% of FV plasma levels
being all FVL. This is the case of the so-called pseudo-homozygous FVL which show a
severe APC resistance and a stronger tendency towards thrombosis as compared to
heterozygous FVL alone [9]. In pseudo-homozygotes, APC-resistance levels are
consistent with homozygosity though DNA testing shows heterozygous FVL. This
discrepancy between the genotype (heterozygous FVL) and the laboratory (severe APC
resistance) or clinical phenotypes (severe thrombotic tendency) should prompt further
analyses to exclude the presence of pseudo-homozygosity. Indeed, the thrombotic risk in
pseudo-homozygous patients is as high as that observed in homozygous individuals [43].
High plasma levels of procoagulants, such as factor VIII, IX, X, XI, and fibrinogen can
increase the risk of VTE and should therefore be included among the gain-of-function
thrombophilia abnormalities [4]. However, the genetic basis behind elevated levels of
these procoagulants is only partially known. The extent of the increased risk of VTE
ranges from 1.6-fold for plasma factor X levels >90th percentile, to fourfold for a fibrinogen
level >5.0 g/l [4]. More specifically, individuals with sustained elevations of factor VIII (i.e.
serum concentrations 150 IU/dL) have a reported RR of VTE 5 times that observed in
those with levels <100 IU/dL [44]. The same FVIII elevation found among first-degree
relatives could indicate a hereditary transmission. A genetic polymorphism in the low-
density lipoprotein receptor-related protein 1 (LRP1 663C>T) has been showed to be
associated with high levels of factor VIII. The carriers had a 3-fold enhanced VTE risk
regardless of other prothrombotic risk factors [45]. Moreover, the gene polymorphism
encoding the B-domain substitution Asp1241Glu in the F8 gene (92714C>G) has been
reported to be associated with elevated factor VIII levels [46].
Levels of FIX antigen >129 U/dl are associated with a 2.3-fold increased risk of VTE
(95%CI, 1.6-3.5) [47]. Factor IX Malmö is an A>G sequence variant (rs6048) in the F9
gene associated with VTE, although the prothrombotic mechanism remains unknown [48].
Accepted Manuscript
Factor IX Padua is a gain-of-function mutation in the F9 gene (R338L) discovered by our
group, which was detected in some components of a single family symptomatic for VTE
and carrying extremely high plasma factor IX activity (eight times the normal levels)
contrasting with normal antigen levels [10]. The mutation has not been reported in other
cohorts of patients with VTE so far [49].
Elevations of factor XI >90th percentile were found to be an independent risk factor for VTE
with a 2-fold increased risk [50]. No genetic mutations responsible for increased FXI levels
have been identified so far.
A novel gain-of-function mechanism defined as “resistance to antithrombin” has been
identified in families with thrombophilia. The molecular basis is a missense mutation at the
prothrombin Arg596 residue (exon 14) leading to a defect of thrombin–antithrombin
binding and impaired inhibition by AT of the mutated thrombin [51]. The first prothrombin
mutation causing antithrombin-resistance was reported in 2012 (Prothrombin Yukuhashi)
[51]. Subsequently, similar cases were recorded in Serbia, India and Italy [52, 53, 54]. The
five families carry three different mutations on the same Arginine 596, and namely:
Prothrombin Yukuhashi Arg596Leu [51], Prothrombin Belgrade and Amrita Arg596Gln [52,
54] and Prothrombin Padua 2, Arg596Trp [53]. Point mutations of prothrombin Arg596
have been identified in several thrombotic patients of different race and ethnicity
worldwide. However, the real incidence of these mutations and the associated thrombotic
risk have not been clearly defined yet. It is worth mentioning that these novel hereditary
mechanisms of thrombophilia are rare and currently do not affect much the clinical
practice. However, clinicians should think even to these novel mutations or to other new
potential genetic mechanisms when approaching patients or families with unexplained
history of recurrent VTE.
3. Acquired thrombophilia
The term thrombophilia also includes the hypercoagulable states caused by acquired
disorders of haemostasis. Acquired disorders can foster a prothrombotic state via elevated
procoagulant factors, reduced anticoagulants, pro-inflammtory/autoimmune mechanisms,
as well as multiple alterations in homeostatic regulation of blood coagulation. The main
acquired disorders associated to thrombophilia are antiphospholipid antibody syndrome,
hyperomocisteinaemia, transiently/permanently increased levels of procoagulant factors,
and finally, decreased levels of natural anticoagulants.
Accepted Manuscript
3.1 Antiphospholipid antibody syndrome
The antiphospholipid antibody syndrome (APS) is an immune-mediated disease
characterized by thrombotic and/or obstetrical events [55]. Thrombotic APS is
characterized by venous, arterial, or microcirculation thrombosis; whereas obstetrical APS
is determined by foetal loss after the 10th week of gestation, recurrent early miscarriages,
intrauterine growth restriction, or severe preeclampsia. This disease is caused by
antibodies directed against membrane anionic phospholipids (i.e. antiphospholipids
antibodies [aPL], mainly anticardiolipin and antiphosphatidylserine antibodies) or their
associated plasma proteins, mainly beta-2 glycoprotein I (β2GPI), or the presence of a
lupus anticoagulant (LAC). A diagnosis of APS is made if at least one of the above
mentioned clinical manifestations occurs and at least one of the laboratory antibodies is
identified in two or more occasions [56]. The main prothrombotic mechanism in APS
involves the binding of aPL to β2GPI on cellular surfaces leading to the following
consequences: i) up-regulation of the expression of potentially prothrombotic adhesion
molecules on the cell-surface (e.g. E-selectin, tissue factor); ii) down-regulation of tissue
factor pathway inhibitor activity; iii) impairment of APC activity; iv); hypofibrinolysis (via
reduction of tissue plasminogen activator-tPA activity); v) complement activation [55, 57].
The risk of a first VTE among asymptomatic subjects who are positive for LAC,
anticardiolipin, and anti–β2GPI antibodies — so-called triple positive patients — is 5.3%
per year. Importantly, 44% of triple positive APS patients will undergo recurrent thrombosis
over a 10-year follow-up period (notwithstanding anticoagulant therapy) [58].
3.2 Hyperhomocysteinaemia
The amino acid homocysteine is formed from the demethylation of methionine and its
plasma levels depend on two main mechanisms: remethylation to methionine (by
methionine synthase which requires both vitamin B12 and N5-methyltetrahydrofolate
reductase activity) or trans-sulphuration to cysteine (by cystathionine β-synthase which
requires vitamin B6) [3]. Both genetic and acquired factors concur to plasma homocysteine
levels. Genetic factors include mutations in the N5-methyltetrahydrofolate reductase
(thermolabile variant of MTHFR) and cystathionine β-synthase genes. On the other hand,
acquired conditions comprise deficiencies of folate, vitamin B12 or B6, hypothyroidism,
psoriasis, chronic renal failure, inflammatory bowel diseases, rheumatoid arthritis, organ
transplantation, antifolate drugs (e.g. anticonvulsants, l-dopa, niacin, methotrexate,
thiazides, and cyclosporine A), or B12 antagonists (e.g. nitrous oxide). Acquired moderate
Accepted Manuscript
hyperhomocysteinaemia may also stem from lifestyle risk factors, such as physical
inactivity, smoking, coffee consumption, and physiologic variables (increasing age,
postmenopausal status, and male gender). Hyperhomocysteinaemia is associated with a
mildly increased risk of atherosclerosis, arterial occlusive disorders and VTE [59]. A meta-
analysis showed that hyperhomocysteinaemia correlates with an estimated 2.9-fold
increased risk of VTE [60]. The degree of inheritance of hyperhomocysteinaemia is low,
suggesting a bigger role for environmental causes [61]. As the matter of fact, the
presence of the thermolabile variant of MTHFR in the absence of hyperhomocysteinaemia,
is not associated with an increased risk of thrombosis [61].
3.3 Acquired high levels of coagulation factors and anticoagulants deficiency
The levels of pro- and anticoagulant factors are genetically controlled though they can be
influenced by age and acquired conditions, and mostly inflammation [3]. Indeed, the
majority of the acquired risk factors for thrombosis — surgery, trauma, acute medical
conditions, inflammation, malignancy and myeloproliferative disorders, obesity — are
characterized by elevated levels of procoagulant factors, particularly factor VIII. Moreover,
acquired risk factors for thrombosis, and namely pregnancy, hormonal therapy, cancer,
nephrotic syndrome are also associated with reduced anticoagulant factors levels. Table 2
summarizes the risks of VTE in acquired thrombophilic conditions while showcasing the
trend of pro- and anticoagulant factors levels. Particularly, the hypercoagulability observed
in pregnancy is due to increased levels of VWF, fibrinogen, factors VII, VIII, X; decreased
PS and acquired APC resistance [62]. The mechanism underlying hypercoagulability in
cancer is quite complex [63, 64]. Briefly, tissue factor is highly expressed in tumor cells;
activated platelets release procoagulant microvesicles (MV) providing a procoagulant
surface; natural anticoagulants levels are also frequently reduced. VTE is a frequent
leading cause of death in cancer, and occurs in about 20% of all cases [62].
Myeloproliferative neoplasms have a high incidence of both venous (10%) and arterial
thrombotic complications (3%). The main causes of hypercoagulability are the following:
endothelial dysfunction, leukocytosis, vascular cell activation, high platelet-induced
thrombin generation, decreased levels of PS (possibly due to absorption on the expanded
blood cell surfaces) and acquired APC resistance, as well as elevated plasma viscosity
[65].
4. Prevention in hereditary thrombophilia
Accepted Manuscript
Each of the aforementioned thrombophilic traits contributes to increasing the thrombotic
risk; it is higher in patients with AT, PC, PS deficiencies, homozygous FVL or PT 20210A,
or concomitant alterations (severe thrombophilia) than in heterozygotes for FVL or PT
20210A (mild thrombophilia). Overall, hereditary thrombophilia reportedly increases the
RR of VTE by a factor of ~ 4 to 30. The highest incidence of 0.9–4.0 per 100 person-years
is observed in carriers of AT deficiency [3, 4], with the exception of type II heparin binding
site defects featured by reduced probability of developing VTE unless homozygous defects
are present. Conversely, mild thrombophilia confers a 2- to 7-fold increased risk of VTE
(0.14–0.67 per 100 person-years for FVL; 0.05–0.42 per 100 person-years for prothrombin
mutation) [3, 4]. Interestingly, low borderline levels of AT, PC, and PS also have been
associated with a twofold increased risk of VTE, comparable with that of mild
thrombophilia [66].
VTE can be considered like a double- or triple-hit process, which means a single risk factor
(e.g., heterozygous FVL) will provide an element of risk that is unlikely to singlehandedly
cause thrombosis, but an additional risk factor (e.g., surgery) may provide the trigger for
overt thrombosis to occur [67]. Indeed, carriers may remain asymptomatic for many years,
suggesting that the development of VTE may require further triggering factors. In family
studies conducted on carriers of natural anticoagulants deficiencies, about half of
thrombotic events were triggered by a concomitant acquired risk factor [4]. Thus,
interactions between genetic and acquired risk factors should be considered to determine
the overall prothrombotic profile of patients.
Indeed, the incidence of VTE in patients with AT, PC, or PS defects with exposure to
acquired risk factors is estimated to be 1.2–8.1 per 100 person-years, whereas said
incidence in mild thrombophilia is much lower during exposure to acquired risk factors,
around 0.2-2.3 per 100 person-years [2]. First of all, a positive familial history of VTE is
per se” a strong risk factor for VTE and should be always taken into consideration when
evaluating a patient’s thrombotic risk. The second element is age; in fact, in family cohort
studies, carriers of AT, PC, or PS defects, exhibited VTE at a younger age (median 29
years) than those with FVL or PT G20210A (median 40 years) [44, 68]. Recent findings
from our group showed that circulating MVs may also concur to the occurrence of VTE in
hereditary thrombophilic carriers — both in mild and severe states — as well as in APS
[69, 70, 71]. Circulating MVs, which may constitute an epiphenomenon of the
Accepted Manuscript
thrombophilic state itself, could be up-regulated in acute conditions thus enhancing the
overall hypercoagulable state up to the threshold of VTE occurrence [69, 70, 71].
Surgery, immobilization, and pregnancy are some of the additional risk factors for
thrombosis that may prompt to consider thromboprophylaxis in patients with hereditary
thrombophilia. Here, we summarize the latest evidence on prevention of VTE in
thrombophilia patients in the most common high-risk situations.
5. Prevention of surgery- related VTE
Because of the risk of bleeding complications that can occur during thromboprophylaxis,
the risk-benefit ratio of antithrombotic therapy as primary prevention for surgery-related
VTE in at-risk patients need to be carefully assessed. In 2012, the American College of
Chest Physicians (ACCP) Guidelines suggest the use of thromboprophylaxis with low-
molecular-weight heparin (LMWH) after general and abdominal-pelvic surgery in patients
at moderate risk for VTE (~3.0%; Caprini score 3-4) who did not present with a high risk for
major bleeding [72]. The Caprini Scoring system is a tool to identify patients at increased
postoperative risk for VTE [73]. Interestingly enough, 3 points were assigned to the
presence of FVL or PT G20201A, LAC, elevated anticardiolipin antibodies, elevated serum
homocysteine, or other congenital or acquired thrombophilia. Accordingly, pharmacological
thromboprophylaxis could be indicated in patients otherwise considered at low risk for VTE
[74]. Interestingly, a positive family history of VTE represents itself an additional risk factor
(3 points in Caprini score), irrespective of a confirmed thrombophilic state. Thus, the
detection of inherited thrombophilia is not likely to make a significant contribution to the
stratification of the risk if family history of VTE is already present. Likewise, there is still no
evidence that modality and duration of thromboprophylaxis should be different in subjects
with or without inherited thrombophilia. It is worth mentioning however, that some authors
empirically recommend higher doses of heparin (whichever formulation) in AT-deficient
patients, potentially at risk of heparin resistance, and a longer duration of postoperative
prophylaxis because of the presence of the severe thrombophilic defect [75]. Similarly,
data gathered from case reports suggest replacement therapy using AT concentrates [30]
to maintain AT plasma levels closer to normal. Clark et al. [76] described the successful
management of 3 patients with AT deficiency undergone coronary artery bypass surgery
using a preoperative loading dose of AT on the evening before surgery, followed by
reduced doses for 5 to 7 days postoperatively. In addition, all patients received
postoperative heparin and no thrombotic complications occurred [76]. Moreover, AT
Accepted Manuscript
replacement therapy could be given when the use of standard thromboprophylaxis is
precluded by a high risk of bleeding or in patients at highest risk of VTE (e.g., orthopedic
or abdominal/pelvic cancer surgeries) carrying hereditary AT deficiency, although either
doses or duration of replacement therapy are still undefined [75, 77]. The use of plasma-
derived protein C concentrates has also been suggested in patients with (severe) PC
deficiency undergoing surgery [78].
The ACCP guidelines suggest the use of mechanical prophylaxis (i.e. elastic stockings or
intermittent pneumatic compression) in addition to pharmacological prophylaxis after
general and abdominal-pelvic surgery in individuals at high risk for VTE (~6.0%; Caprini
score 5). [72].
6. Prevention of pregnancy-related VTE
Decisions whether using of prophylactic anticoagulation during pregnancy to prevent VTE
depend on the benefit-risk ratio. The rate of clinically relevant maternal bleeding due to the
LMWH use is about 2%, mainly associated with the delivery (1%) or wound haematoma
(0.6%) [79]. On the other hand, in a systematic review of 9 case-control studies (n=2526
patients) considering the association between thrombophilia and pregnancy-associated
VTE, the highest risk was associated to homozygous FVL or PTG20201A [80].
Deficiencies of AT, PC, and PS were associated with moderate risk increases, while
pregnant women with mild thrombophilias presented with a lower risk of VTE. Considering
a basal incidence of VTE complications during pregnancy of about 1/1000 deliveries, the
calculated absolute risks suggest a low prothrombotic profile (0.5–1.2% of affected
pregnancies) for most inherited thrombophilias, except homozygotes for FVL or PT
G20201A where the risk estimate increases to approximately 4% [81]. Needless to say,
this observation can be influenced by the prevalence of thrombophilic defects which is
higher for the less severe thrombophilic conditions and lower for the more severe defects.
Interestingly, a positive family history of VTE confers a two- to four-fold increased risk for
VTE, depending on the number of symptomatic relatives, while thrombophilic women with
asymptomatic family have a lower risk of VTE [81]. Regarding acquired thrombophilias,
although less studied, repeated aPL positivity is associated with an increased risk of VTE
[82]. The risk of pregnancy-related VTE in women with aPL and without previous VTE is
uncertain. It is worth mentioning that thrombophilia screening is not recommended as a
routine initial test in pregnancy or as prenatal/newborn screening; however women should
Accepted Manuscript
undergo an individualized VTE risk profile assessment both prior to pregnancy, as well as
once pregnancy is achieved and throughout pregnancy as soon as new clinical situations
arise [81].
In summary, pregnant women with asymptomatic inherited thrombophilia have a
significantly increased risk of VTE particularly if they have a positive family history of VTE
or severe thrombophilia (i.e. homozygous FVL or PTG20201A, double heterozigosity,
natural anticoagulant defects). Most guidelines recommend antenatal LMWH prophylaxis
in those cases [4, 81, 83, 84, 85]. Conversely, antenatal clinical surveillance is believed to
be sufficient for pregnant women with mild thrombophilia, and clinicians should consider
antenatal LMWH prophylaxis only in the presence of additional risk factors (positive family
history, hypo mobility, obesity, age >35 years, gross varicose veins) [4]. As for post-
partum, antithrombotic LMWH prophylaxis is recommended for 4-6 weeks in carriers of
severe thrombophilia. The same is true of mild thrombophilia, even though data are
inconsistent, with antithrombotic prophylaxis ranging from at least 7 days to up to 6 weeks
post-partum but only for women carrying additional risk factors [4]. ACCP guidelines are
more restrictive in thromboporophylaxis indications pointing out the positive family history
more than the carriership of thrombophilia [86]. When the decision is made to use
antepartum prophylaxis, it should be initiated early in the pregnancy. Finally, prophylaxis is
recommended after caesarean delivery in case of a known thrombophilia. As for AT
deficiency, it has been recommended that AT concentrate be administered prior to delivery
and during post-partum if the expectant mother recently experienced an acute VTE event
or has a history of VTE. Indeed, it was reported that in AT-deficient patients with recent or
recurrent thrombosis, the administration of AT during labor successfully prevented
thrombotic recurrences; this is a period during which administration of anticoagulation may
be dangerous [30].
7. Prevention of VTE induced by oestrogen–progestogen therapies
Combined oral contraceptives (COC) therapy can increase the risk of VTE by two to
ninefold. This is due both to the dose of oestrogen and the type of progestogen contained
in the preparations. The so called “third-generation pills” containing desogestrel or
gestodene are associated with a twofold increased risk of VTE compared to levonorgestrel
(present in the “second-generation” pills) [87]. Additionally, the most recent “fourth-
generation” type of COC containing progestins such as drospirenone, nomegestrol acetate
or dienogest present with 3-fold increased risk of VTE compared to the second-generation
Accepted Manuscript
pills [88]. A multiplicative effect of COC use and inherited thrombophilia on the risk of VTE
was observed, with a two to fivefold increase in women with both risk factors versus either
risk factor considered individually [89]. Interestingly, a recent study on pre-menopausal
women showed a statistically significant interaction between COC use and presence of
FVL on VTE risk (OR 1.37, 95%CI 1.06–1.77). This interaction appeared higher for
drospirenone (OR 1.99 [95%CI 1.18–3.38]) or third-generation cyproterone acetate users
(OR 1.71 [95%CI 1.20–2.45]), but not significant for 1st or 2nd COC users [90]. Recently,
WHO Medical Eligibility Criteria confirmed that COC use in women with hereditary
thrombophilias is associated with an unacceptable health (life threatening) risk [91]. A
recent meta-analysis regarding 12 case–control and three cohort studies showed that mild
thrombophilia increased the risk of VTE in COC users by 6-fold (RR 5.89; 95%CI 4.21–
8.23) (14 studies considered), while the presence of severe thrombophilia enhanced the
risk in COC users by approximately 7-fold (RR 7.15; 95%CI 2.93–17.45) (3 studies
considered) [89]. VTE incidence in COC users with mild thrombophilia was 0.49 (95%CI
0.18–1.07) to 2.0 (0.3–7.2) per 100 pill-years; it was 0.86 (95%CI, 0.10–3.11) per 100 pill-
years in COC users double heterozygous or homozygous for FVL or PT G20201A and 4.3
(95%CI 1.4–9.7) to 4.62 (95%CI 2.5–7.9) per 100 pill-years in COC users who were
carriers of AT, PC or PS defects. These results confirm that COC should not be
administered to women with severe hereditary thrombophilia, including asymptomatic
women. Possible options with similar contraceptive efficacy include progestogen-only pills,
intrauterine devices (IUDs) containing only levonorgestrel and Cu-IUDs containing at least
300 mm2 Cu [89]. Thrombophilia screening is not generally recommended before the use
of oral contraceptives/hormone replacement therapy/other selective estrogen receptor
modulators, as it is not cost-effective and may increase patient anxiety. However, a
clinician should bear in mind that screening and identification of asymptomatic women who
are family members of symptomatic carriers of thrombophilia (particularly severe
thrombophilia) before prescribing oral contraceptives can be beneficial. By contrast, the
additive VTE risk of oral contraceptives in mild thrombophilia is maintained to be modest.
In women with known mild thrombophilia, detailed counseling on all contraceptive options
is strongly recommended to supply them all the elements to decide whether the increased
risk of VTE related to oral contraceptives use, albeit modest, is balanced by the benefit
coming from the chosen strategy. In the absence of other risk factors — family history,
body mass index, others — COCs are often prescribed in asymptomatic carriers of mild
thrombophilia when reliable alternative contraceptives are refused/not tolerated, bearing
Accepted Manuscript
also in mind that in these patients the risk of pregnancy-related VTE outweighs the COC-
associated VTE risk. In such cases, COCs with the lowest prothrombotic profile are
recommended as the first choice (i.e. levonorgestrel, norgestimate- or norethisterone-
containing COCs with a low ethinylestradiol amount [35 micrograms]). As far as
thrombophilia screening is concerned, it is generally not recommended to test
asymptomatic women from families with known mild thrombophilia [92] but a clear
evidence on the benefit-risk ratio of such an approach is still lacking . As a consequence, it
is recommended to fully inform those women on the implications of screening so that they
can knowingly decide whether or not to be tested.
8. Prevention of cancer-associated thrombosis
A few studies evaluating the role of FVL and PT G20210A on the risk of cancer-associated
thrombosis have been published [93]. Overall, despite conflicting results, it would appear
that cancer patients carrying either of these mutations tend to show an increase risk of
thrombosis. In a case–control study by Blom et al. [94] on 205 adult cancer patients, the
risk of VTE was 2-fold higher in patients with than those without FVL (adjusted OR: 2.2
[95%CI 0.3-17.8]). Moreover, patients with cancer and PT G20210A had an OR for VTE of
4.1 (95%CI 0.3-60.8) compared with those without mutation. In a study by a Turkish group,
the prevalence of FVL was significantly higher (30.2%) in cancer patients with VTE
compared to those without VTE (3.7%, p <0.001) [95], while no significant difference in the
prevalence of PT G20210A was observed. Particularly, patients with cancer and central
venous catheters (CVC) who had inherited thrombophilia showed increased risk of VTE. In
a meta-analysis by Dentali et al. [96] including 10 studies (211 cases and 860 controls)
increased risk of CVC-related thrombosis in carriers of FVL [OR 4.6 (95%CI 2.6-8.1)] and
PT G20210A [OR 4.9 (95%CI 1.7-14.3)], respectively was shown. The measured
attributable risk of CVC-related thrombosis was 13.1% and 4.5% for FVL and PT
G20210A, respectively. In carriers with FVL receiving allogeneic bone marrow
transplantation, the risk of CVC-associated thrombosis was increased by 7-fold
independently of the administration of thromboprophylaxis [95]. Similar data were obtained
in FVL carriers affected by breast cancer [97]. Furthermore, in women with FVL,
thrombotic events occurred mainly during the first 6 months of adjuvant therapy with
tamoxifen [22]. In a cohort of 175 patients with gastrointestinal adenocarcinoma, 17.9% of
the patients with VTE were heterozygotes for FVL versus 4.8% of those without VTE (p =
.026, OR 4.4), whereas heterozygosity for PT G20210A did not affect the incidence of
thrombosis [22]. More recently, FVL was found to be associated with the occurrence of
Accepted Manuscript
VTE in metastatic colorectal cancer treated with first-line chemotherapy plus bevacizumab
(only at univariate analysis) [98]. No specific association between AT, PC, and PS
hereditary deficiencies and cancer-associated thrombosis has been described so far, with
the only exception of children with acute lymphoblastic leukemia (ALL). In a meta-analysis
performed by Caruso et al. [99] based on 17 prospective studies and 1,752 children with
ALL, the rate of VTE was 5.2%. Thrombophilia (FVL, PTG20210A, MTHFR genotype,
elevated lipoprotein, deficiencies of PC, PS, AT,) was evaluated in 557 children (5 studies)
out of 1,752 and the thrombotic risk in ALL patients with thrombophilia rose up
approximately 8-fold (RR 8.5, 95%CI 4.4–17.4). Even though data are still limited, it
appears that thrombophilia could increase the risk of cancer-associated thrombosis.
However, inherited thrombophilia testing is not currently recommended in the strategies of
prophylaxis and treatment of VTE in cancer patients.
9. Prevention of economy class syndrome
The ‘economy class syndrome’ is defined as the occurrence of thrombotic complications
during long-haul flights that manifest mainly in passengers travelling in economy class. In
normal healthy individuals the estimated absolute risk of VTE on flights lasting more than 4
h was about 1 in 6000 [100]. In a meta-analysis [101] of 14 studies including 4055 cases of
VTE occurred during both air and overland trips lasting up to 8 h and with a follow-up time
post-journey ranging from two to eight weeks, the overall RR of VTE was 2.8 [95%CI 2.2–
3.7] with an approximate 18% increase in the risk at each increment of 2 h in travel time.
This risk increased up to 26% if only air travels were considered. In travellers with
thrombophilia, the risk of VTE was 16.8 times higher (95%CI 3.8–74.7) than in non-
thrombophilic, non-travelers, accounting for a synergistic interaction [102]. In the case of
thrombophilia, antithrombotic measures are of paramount importance. Travellers should
be informed of the potential thrombotic risks, encouraged to exercise regularly with short
walks along the aisles, and avoid dehydration [100]. The Aerospace Medical Association
(AsMA) published guidelines on optimal antithrombotic prophylaxis in subjects who travel
by air and patients with known thrombophilia is considered at high risk for thrombosis
[103]. Recommendations for travellers with high thrombotic risk on flights lasting over 8 h
are: avoidance of constrictive clothing around the lower extremities or waist; adequate
hydration; repeated calf muscle contraction. A perceived higher risk of VTE may prompt
active thromboprophylaxis measures such as properly fitted below-knee compressive
Accepted Manuscript
stockings (15-30 mmHg of pressure at the ankle) or a single prophylactic dose of LWMH,
injected prior to departure.
10. Prevention of post-thrombotic syndrome
Post-thrombotic syndrome (PTS), the most common chronic complication of deep vein
thrombosis (DVT), causes chronic limb pain, swelling and leg ulcers, which reduces the
quality of life of affected patients [104]. Previous studies reported that 20–50% of patients
with DVTs developed PTS [104]. A study by our group considering 191 patients with
thrombophilia and 339 without, showed that the adjusted HR for the development of PTS
in thrombophilic versus non-thrombophilic patients was 1.23 (95%CI, 0.92–1.64) [42].
Specifically, the adjusted HR for the development of PTS in carriers was 0.42 (95%CI,
0.20–0.88; P = 0.02) for FVL, 0.81 (0.36–1.37) for LAC, 0.96 (0.29–3.82) for PC
deficiency, 1.08 (0.29–2.70) for PS deficiency, and 1.33 (0.68–2.58) for PT G20210A. Of
the two patients with AT deficiency, neither developed PTS. A meta-analysis by
Rabinovich et al. [105] considered 16 studies (13 assessing FVL, 10 PT G20210A, 5 PC
and PS deficiencies, 3 AT deficiency, 4 elevated FVIII, and 6 aPL). The Authors confirmed
that none of the studies acknowledged any of the thrombophilias as predictive of PTS.
Unexpectedly, FVL and PT G20210A even appeared to protect against PTS in studies
including patients with first and recurrent DVT events, and studies in which over 50% of
patients had an unprovoked event. A very recent Japanese Registry gathered data on
1218 patients with DVT who completed 3 years of follow-up [104]. The Authors showed
that chronic kidney disease (OR 2.21, 95% CI 1.45–3.39), leg swelling (OR 4.15, 95%CI
2.25–7.66), no transient risk factors for VTE (OR 2.39, 95%CI 1.55–3.67), active cancer
(OR 3.66, 95%CI 2.30–5.84), and thrombophilia (OR 2.07, 95%CI 1.06–4.04) were
independent risk factors for the development of PTS. Patients with thrombophilia were 79
(6.1%) and, interestingly, they considered only severe thrombophilia (APS; PC, PS and AT
deficiencies). This is the only study where severe thrombophilia appears to be associated
with the development of PTS. At the moment, the association between thrombophilia and
PTS is not deemed important enough to warrant a thrombophilia screening or different
secondary prophylaxis strategies in thrombophilic carriers. Needless to say, this
conclusion is based on low levels of evidence due to the lack of properly designed studies.
Finally, a recent study by our group suggested that non-O blood type might be a risk factor
for the development of PTS, as individuals with non-O blood group were associated with a
significantly higher PTS occurrence (HR 1.53, 95% CI, 1.05–2.24; p = 0.028) than O group
Accepted Manuscript
[106]. Although larger studies are needed to confirm our results, blood group might be an
additional element to take into account in the evaluation of the strategies to prevent PTS.
11. Expert Opinion
Fifty-three years following the first description of a hereditary prothrombotic condition (AT
deficiency) we have greatly increased our knowledge on both hereditary and acquired
causes of hypercoagulability which can temporary or long-life predispose carriers to VTE.
Focusing on hereditary thrombophilia, currently available data uncovered a thrombotic risk
gradient from carriers of natural anticoagulant deficiencies and in the presence of
homozygous defects or multiple abnormalities (severe thrombophilia) to heterozygotes for
FVL and prothrombin G20210A (mild thrombophilia). Additionally, novel prothrombotic
hereditary conditions have been recently described and the thrombotic risk conferred by
them is still unknown. Among the most common acquired prothromboitc conditions,
clinicians have to consider APS and hyperhomocisteinaemia, as well as all the conditions
characterized by a temporary acquired increase in procoagulant and/or decrease in
anticoagulant factors. Importantly, the management of a patient with a thrombophilic
condition should take into consideration other (genetic and/or acquired) coexisting risk
factors for first or recurrent VTE in order to determine the necessity and duration of
prevention (primary prophylaxis). The main role of the clinicians is to individualize case-by-
case the approach for the prevention of VTE and consider each patient’s risk taking into
account strong situational risk factors (e.g., surgery, pregnancy, therapies, etc.). Notably,
the susceptibility to VTE is modulated by the presence or absence of a family history of
VTE, thus family history plays an important role in the evaluation of a patient’s risk profile.
In addition, the knowledge of the carriership status of the so-called severe thrombophilia
can be important to properly counsel asymptomatic relatives — particularly women of
childbearing age — on thrombophilia screening or primary prophylaxis. Finally, there is no
consensus on some hot-topic issues such as care of fertile-age women to date. In these
situations, counseling by qualified clinicians addressing the associated risks-benefit ratio
and the available prevention strategies are strongly suggested in order to provide these
women an aware choice.
Novel severe thrombophilic conditions have been identified in the last few years, and
namely, factor IX Padua [10], prothrombin Yukuhashi [52], prothrombin Belgrade [52],
prothrombin Amrita [54], and prothrombin Padua 2 [53]. Even more new genetic defects
Accepted Manuscript
predisposing to thrombosis are likely to be discovered soon. The discovery of novel
genetic mutations undoubtedly opens new doors to understanding the mechanisms of
thrombosis. Studies on thrombophilia need to be conducted anew, taking into account a
more thorough evaluation and definition of thrombophilia. Granted, we now possess a
wealth of knowledge as it pertains to the slight impact of mild thrombophilias on the overall
thrombotic risk; nevertheless, there are still areas of doubt and uncertainty regarding
severe and novel thrombophilia. Future research projects should focus on identifying novel
thrombophilic factors in order to explain (and possibly prevent) more cases of unprovoked
thrombosis, and to clarify the molecular mechanisms underlying the complex interaction
between genetic and environmental risk factors for thrombosis. Finally, the more favorable
safety and efficacy profiles of direct oral anticoagulants recently introduced in clinical
practice should be considered in the light of future strategies for prevention and treatment
of VTE in patients with inherited or acquired thrombophilia.
Article Highlights
Thrombophilia is a clinical condition defined as either an inherited or acquired
abnormality of haemostasis predisposing to venous thromboembolism (VTE).
Hereditary and acquired thrombophilia determine a prothrombotic state. Acquired
disorders foster a prothrombotic state via elevated procoagulant factors, reduced
anticoagulants, pro-inflammatory/autoimmune mechanisms.
Hereditary thrombophilia is divided into mild or severe according to the thrombotic
risk associated with the genetic abnormalities.
New genetic gain-of-functions defects responsible for severe thrombophilia have
been identified, and namely, pseudo-homozygosity for activated protein C
resistance, factor IX Padua and resistance to antithrombin.
Individual’s ABO blood group is the most common genetic risk factor for VTE to
remember.
The main acquired thrombophilia conditions are antiphospholipid antibody
syndrome, hyperomocisteinemia, acquired increased levels of procoagulant factors
and/or decreased levels of natural anticoagulants.
Accepted Manuscript
A subject with thrombophilia should be evaluated in the framework of other (genetic
and/or acquired) coexisting risk factors for first or recurrent VTE when assessing
the need and duration of prevention (primary prophylaxis).
Funding
This paper was not funded.
Declaration of conflicts of interest
The authors have no relevant affiliations or financial involvement with any organization or
entity with a financial interest in or financial conflict with the subject matter or materials
discussed in the manuscript. This includes employment, consultancies, honoraria, stock
ownership or options, expert testimony, grants or patents received or pending, or royalties.
Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other relationships to
disclose.
References
Papers of special note have been highlighted as either of interest (•) or of considerable
interest (••) to readers.
1. Bagot CN, Arya R. Virchow and his triad: a question of attribution. Br J Haematol.
2008;143(2):180-90.
2. Middeldorp S, van Hylckama Vlieg A. Does thrombophilia testing help in the clinical
management of patients? Br J Haematol. 2008;143(3):321-35.
3. Mannucci PM, Franchini M. The real value of thrombophilia markers in identifying
patients at high risk of venous thromboembolism. Expert Rev Hematol.
2014;7(6):757-65.
• The review clearly summarizes epidemiology and management of hereditary
thrombophilia
4. Martinelli I, De Stefano V, Mannucci PM. Inherited risk factors for venous
thromboembolism. Nat Rev Cardiol. 2014;11(3):140-56.
•• This very inclusive review cover relevant issues on diagnosis and management of
hereditary thrombophilia
5. Crowther MA, Kelton JG. Congenital thrombophilic states associated with venous
thrombosis: a qualitative overview and proposed classification system. Ann Intern
Med. 2003;138(2):128-34.
6. Dahlback B. Advances in understanding pathogenic mechanisms of thrombophilic
disorders. Blood. 2008;112(1):19-27.
7. Margaglione M, Brancaccio V, Giuliani N, et al. Increased risk for venous
thrombosis in carriers of the prothrombin G-->A20210 gene variant. Ann Intern Med.
1998;129(2):89-93.
Accepted Manuscript
8. Mateo J, Oliver A, Borrell M, et al. Laboratory evaluation and clinical characteristics
of 2,132 consecutive unselected patients with venous thromboembolism--results of
the Spanish Multicentric Study on Thrombophilia (EMET-Study). Thromb Haemost.
1997;77(3):444-51.
9. Castoldi E, Kalafatis M, Lunghi B, et al. Molecular bases of pseudo-homozygous
APC resistance: the compound heterozygosity for FV R506Q and a FV null
mutation results in the exclusive presence of FV Leiden molecules in plasma.
Thromb Haemost. 1998;80(3):403-6.
10. Simioni P, Tormene D, Tognin G, et al. X-linked thrombophilia with a mutant factor
IX (factor IX Padua). N Engl J Med. 2009;361(17):1671-5.
11. Takagi Y, Murata M, Kozuka T, et al. Missense mutations in the gene encoding
prothrombin corresponding to Arg596 cause antithrombin resistance and
thrombomodulin resistance. Thromb Haemost. 2016;116(6):1022-1031.
12. Wu O, Bayoumi N, Vickers MA, et al. ABO(H) blood groups and vascular disease: a
systematic review and meta-analysis. J Thromb Haemost. 2008;6(1):62-9.
13. Dahlback B, Carlsson M, Svensson PJ. Familial thrombophilia due to a previously
unrecognized mechanism characterized by poor anticoagulant response to
activated protein C: prediction of a cofactor to activated protein C. Proc Natl Acad
Sci U S A. 1993;90(3):1004-8.
14. Svensson PJ, Dahlback B. Resistance to activated protein C as a basis for venous
thrombosis. N Engl J Med. 1994;330(8):517-22.
15. Norstrom E, Thorelli E, Dahlback B. Functional characterization of recombinant FV
Hong Kong and FV Cambridge. Blood. 2002;100(2):524-30.
16. Castoldi E, Rosing J. APC resistance: biological basis and acquired influences. J
Thromb Haemost. 2010;8(3):445-53.
17. Chan WP, Lee CK, Kwong YL, et al. A novel mutation of Arg306 of factor V gene in
Hong Kong Chinese. Blood. 1998;91(4):1135-9.
18. Bernardi F, Faioni EM, Castoldi E, et al. A factor V genetic component differing from
factor V R506Q contributes to the activated protein C resistance phenotype. Blood.
1997;90(4):1552-7.
19. Poort SR, Rosendaal FR, Reitsma PH, et al. A common genetic variation 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(10):3698-703.
20. Kyrle PA, Mannhalter C, Beguin S, et al. Clinical studies and thrombin generation in
patients homozygous or heterozygous for the G20210A mutation in the prothrombin
gene. Arterioscler Thromb Vasc Biol. 1998;18(8):1287-91.
21. Smirnov MD, Safa O, Esmon NL, et al. Inhibition of activated protein C
anticoagulant activity by prothrombin. Blood. 1999;94(11):3839-46.
22. Campello E, Spiezia L, Simioni P. Diagnosis and management of factor V Leiden.
Expert Rev Hematol. 2016;9(12):1139-1149.
23. Van Cott EM, Khor B, Zehnder JL. Factor V Leiden. Am J Hematol. 2016;91(1):46-
9.
24. Salomon O, Steinberg DM, Zivelin A, et al. Single and combined prothrombotic
factors in patients with idiopathic venous thromboembolism: prevalence and risk
assessment. Arterioscler Thromb Vasc Biol. 1999;19(3):511-8.
25. Shemesh A, Hoffman R, Nadir Y, et al. Clinical significance of prothrombin
G20210A mutation in homozygous patients. Am J Hematol. 2017;92(10):E618-
E620.
26. MacCallum P, Bowles L, Keeling D. Diagnosis and management of heritable
thrombophilias. BMJ. 2014;349:g4387.
Accepted Manuscript
27. Martinelli I, Bucciarelli P, Margaglione M, et al. The risk of venous
thromboembolism in family members with mutations in the genes of factor V or
prothrombin or both. Br J Haematol. 2000;111(4):1223-9.
28. Emmerich J, Rosendaal FR, Cattaneo M, et al. 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(3):809-
16.
29. Lane DA, Kunz G, Olds RJ, et al. Molecular genetics of antithrombin deficiency.
Blood Rev. 1996;10(2):59-74.
30. Bauer KA, Nguyen-Cao TM, Spears JB. Issues in the Diagnosis and Management
of Hereditary Antithrombin Deficiency. Ann Pharmacother. 2016;50(9):758-67.
• The review fully discuss epidemiology, diagnosis, management and challenging
situation of antithrombin hereditary deficiency.
31. Finazzi G, Caccia R, Barbui T. Different prevalence of thromboembolism in the
subtypes of congenital antithrombin III deficiency: review of 404 cases. Thromb
Haemost. 1987;58(4):1094.
32. Rossi E, Chiusolo P, Za T, et al. Report of a novel kindred with antithrombin
heparin-binding site variant (47 Arg to His): demand for an automated progressive
antithrombin assay to detect molecular variants with low thrombotic risk. Thromb
Haemost. 2007;98(3):695-7.
33. Middeldorp S. Evidence-based approach to thrombophilia testing. J Thromb
Thrombolysis. 2011;31(3):275-81.
34. Esmon CT. Regulation of blood coagulation. Biochim Biophys Acta. 2000;1477(1-
2):349-60.
35. De Stefano V, Finazzi G, Mannucci PM. Inherited thrombophilia: pathogenesis,
clinical syndromes, and management. Blood. 1996;87(9):3531-44.
36. Price VE, Ledingham DL, Krumpel A, et al. Diagnosis and management of neonatal
purpura fulminans. Semin Fetal Neonatal Med. 2011;16(6):318-22.
37. Reitsma PH, Bernardi F, Doig RG, et al. Protein C deficiency: a database of
mutations, 1995 update. On behalf of the Subcommittee on Plasma Coagulation
Inhibitors of the Scientific and Standardization Committee of the ISTH. Thromb
Haemost. 1995;73(5):876-89.
38. Dahlback B. Inhibition of protein Ca cofactor function of human and bovine protein
S by C4b-binding protein. J Biol Chem. 1986;261(26):12022-7.
39. Gill JC, Endres-Brooks J, Bauer PJ, et al. The effect of ABO blood group on the
diagnosis of von Willebrand disease. Blood. 1987;69(6):1691-5.
40. Sodetz JM, Pizzo SV, McKee PA. Relationship of sialic acid to function and in vivo
survival of human factor VIII/von Willebrand factor protein. J Biol Chem.
1977;252(15):5538-46.
41. Dentali F, Sironi AP, Ageno W, et al. Non-O blood type is the commonest genetic
risk factor for VTE: results from a meta-analysis of the literature. Semin Thromb
Hemost. 2012;38(5):535-48.
42. Spiezia L, Campello E, Bon M, et al. ABO blood groups and the risk of venous
thrombosis in patients with inherited thrombophilia. Blood Transfus. 2013;11(2):250-
3.
43. Simioni P, Castoldi E, Lunghi B, et al. An underestimated combination of opposites
resulting in enhanced thrombotic tendency. Blood. 2005;106(7):2363-5.
44. Lijfering WM, Brouwer JL, Veeger NJ, et al. Selective testing for thrombophilia in
patients with first venous thrombosis: results from a retrospective family cohort
Accepted Manuscript
study on absolute thrombotic risk for currently known thrombophilic defects in 2479
relatives. Blood. 2009;113(21):5314-22.
45. Vormittag R, Bencur P, Ay C, et al. Low-density lipoprotein receptor-related protein
1 polymorphism 663 C > T affects clotting factor VIII activity and increases the risk
of venous thromboembolism. J Thromb Haemost. 2007;5(3):497-502.
46. Viel KR, Machiah DK, Warren DM, et al. A sequence variation scan of the
coagulation factor VIII (FVIII) structural gene and associations with plasma FVIII
activity levels. Blood. 2007;109(9):3713-24.
47. van Hylckama Vlieg A, van der Linden IK, Bertina RM, et al. High levels of factor IX
increase the risk of venous thrombosis. Blood. 2000;95(12):3678-82.
48. Bezemer ID, Arellano AR, Tong CH, et al. F9 Malmo, factor IX and deep vein
thrombosis. Haematologica. 2009;94(5):693-9.
49. Koenderman JS, Bertina RM, Reitsma PH, et al. Factor IX-R338L (Factor IX Padua)
screening in a Dutch population of sibpairs with early onset venous
thromboembolism. Thromb Res. 2011;128(6):603.
50. Meijers JC, Tekelenburg WL, Bouma BN, et al. High levels of coagulation factor XI
as a risk factor for venous thrombosis. N Engl J Med. 2000;342(10):696-701.
51. Miyawaki Y, Suzuki A, Fujita J, et al. Thrombosis from a prothrombin mutation
conveying antithrombin resistance. N Engl J Med. 2012;366(25):2390-6.
52. Djordjevic V, Kovac M, Miljic P, et al. A novel prothrombin mutation in two families
with prominent thrombophilia--the first cases of antithrombin resistance in a
Caucasian population. J Thromb Haemost. 2013;11(10):1936-9.
53. Bulato C, Radu CM, Campello E, et al. New Prothrombin Mutation (Arg596Trp,
Prothrombin Padua 2) Associated With Venous Thromboembolism. Arterioscler
Thromb Vasc Biol. 2016;36(5):1022-9.
54. Sivasundar S, Oommen AT, Prakash O, et al. Molecular defect of 'Prothrombin
Amrita': substitution of arginine by glutamine (Arg553 to Gln) near the Na(+) binding
loop of prothrombin. Blood Cells Mol Dis. 2013;50(3):182-3.
55. Garcia D, Erkan D. Diagnosis and Management of the Antiphospholipid Syndrome.
N Engl J Med. 2018;378(21):2010-2021.
56. Miyakis S, Lockshin MD, Atsumi T, et al. International consensus statement on an
update of the classification criteria for definite antiphospholipid syndrome (APS). J
Thromb Haemost. 2006;4(2):295-306.
57. Giannakopoulos B, Krilis SA. The pathogenesis of the antiphospholipid syndrome.
N Engl J Med. 2013;368(11):1033-44.
58. Pengo V, Ruffatti A, Legnani C, et al. Clinical course of high-risk patients diagnosed
with antiphospholipid syndrome. J Thromb Haemost. 2010;8(2):237-42.
59. Eldibany MM, Caprini JA. Hyperhomocysteinemia and thrombosis: an overview.
Arch Pathol Lab Med. 2007;131(6):872-84.
60. Ray JG. Meta-analysis of hyperhomocysteinemia as a risk factor for venous
thromboembolic disease. Arch Intern Med. 1998;158(19):2101-6.
61. Martinelli I, Bucciarelli P, Zighetti ML, et al. Low risk of thrombosis in family
members of patients with hyperhomocysteinaemia. Br J Haematol.
2002;117(3):709-11.
62. Rybstein MD, DeSancho MT. Hypercoagulable States and Thrombophilias: Risks
Relating to Recurrent Venous Thromboembolism. Semin Intervent Radiol.
2018;35(2):99-104.
63. Montagnana M, Lippi G, Danese E. An Overview of Thrombophilia and Associated
Laboratory Testing. Methods Mol Biol. 2017;1646:113-135.
•This is a very recent review covering relevant issues laboratory diagnosis of
hereditary and acquired thrombophilia
Accepted Manuscript
64. Campello E, Henderson MW, Noubouossie DF, et al. Contact System Activation
and Cancer: New Insights in the Pathophysiology of Cancer-Associated
Thrombosis. Thromb Haemost. 2018;118(2):251-265.
65. Mihaila RG. Thrombin generation - a potentially useful biomarker of thrombotic risk
in Philadelphia-negative myeloproliferative neoplasms. Biomed Pap Med Fac Univ
Palacky Olomouc Czech Repub. 2017;161(1):50-53.
66. Bucciarelli P, Passamonti SM, Biguzzi E, et al. Low borderline plasma levels of
antithrombin, protein C and protein S are risk factors for venous thromboembolism.
J Thromb Haemost. 2012;10(9):1783-91.
67. Favaloro EJ, McDonald D, Lippi G. Laboratory investigation of thrombophilia: the
good, the bad, and the ugly. Semin Thromb Hemost. 2009;35(7):695-710.
68. Brouwer JL, Veeger NJ, Kluin-Nelemans HC, et al. The pathogenesis of venous
thromboembolism: evidence for multiple interrelated causes. Ann Intern Med.
2006;145(11):807-15.
69. Campello E, Radu CM, Spiezia L, et al. Modulating thrombotic diathesis in
hereditary thrombophilia and antiphospholipid antibody syndrome: a role for
circulating microparticles? Clin Chem Lab Med. 2017;55(7):934-943.
70. Campello E, Spiezia L, Radu CM, et al. Circulating microparticles in carriers of
factor V Leiden with and without a history of venous thrombosis. Thromb Haemost.
2012;108(4):633-9.
71. Campello E, Spiezia L, Radu CM, et al. Circulating microparticles and the risk of
thrombosis in inherited deficiencies of antithrombin, protein C and protein S.
Thromb Haemost. 2016;115(1):81-8.
72. Gould MK, Garcia DA, Wren SM, et al. Prevention of VTE in nonorthopedic surgical
patients: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American
College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest.
2012;141(2 Suppl):e227S-e277S.
73. Caprini JA. Thrombosis risk assessment as a guide to quality patient care. Dis Mon.
2005;51(2-3):70-8.
74. Coppola A, Tufano A, Cerbone AM, et al. Inherited thrombophilia: implications for
prevention and treatment of venous thromboembolism. Semin Thromb Hemost.
2009;35(7):683-94.
75. Patnaik MM, Moll S. Inherited antithrombin deficiency: a review. Haemophilia.
2008;14(6):1229-39.
76. Clark P, Walker ID, Neilson RF. Coronary artery bypass surgery in patients with
inherited antithrombin deficiency. Br J Haematol. 1995;90(2):479-82.
77. Rodgers GM. Role of antithrombin concentrate in treatment of hereditary
antithrombin deficiency. An update. Thromb Haemost. 2009;101(5):806-12.
78. Tufano A, Coppola A, Cerbone AM, et al. Preventing postsurgical venous
thromboembolism: pharmacological approaches. Semin Thromb Hemost.
2011;37(3):252-66.
• Comprehensive review on post-surgical prevention of venous thromboembolism
79. Greer IA, Nelson-Piercy C. Low-molecular-weight heparins for thromboprophylaxis
and treatment of venous thromboembolism in pregnancy: a systematic review of
safety and efficacy. Blood. 2005;106(2):401-7.
80. Robertson L, Wu O, Langhorne P, et al. Thrombophilia in pregnancy: a systematic
review. Br J Haematol. 2006;132(2):171-96.
81. Bates SM, Middeldorp S, Rodger M, et al. Guidance for the treatment and
prevention of obstetric-associated venous thromboembolism. J Thromb
Thrombolysis. 2016;41(1):92-128.
Accepted Manuscript
•• This review clearly summarizes prevention guidance of pregnancy-related venous
thromboembolism
82. Galli M, Barbui T. Antiphospholipid syndrome: clinical and diagnostic utility of
laboratory tests. Semin Thromb Hemost. 2005;31(1):17-24.
83. James A, Committee on Practice B-O. Practice bulletin no. 123: thromboembolism
in pregnancy. Obstet Gynecol. 2011;118(3):718-29.
84. Chan WS, Rey E, Kent NE, et al. Venous thromboembolism and antithrombotic
therapy in pregnancy. J Obstet Gynaecol Can. 2014;36(6):527-53.
85. Royal College of Obstetricians and Gynaecologists. Green-top Guideline No. 37a.
Reducing the risk of thrombosis and embolism during pregnancy and the
puerperium. https://wwwrcogorguk/en/guidelines-research-
services/guidelines/gtg37a. 2015.
86. Bates SM, Greer IA, Middeldorp S, et al. VTE, thrombophilia, antithrombotic
therapy, and pregnancy: Antithrombotic Therapy and Prevention of Thrombosis, 9th
ed: American College of Chest Physicians Evidence-Based Clinical Practice
Guidelines. Chest. 2012;141(2 Suppl):e691S-e736S.
87. Lidegaard O, Lokkegaard E, Svendsen AL, et al. Hormonal contraception and risk
of venous thromboembolism: national follow-up study. BMJ. 2009;339:b2890.
88. Larivée N, Suissa S, Coulombe J, et al. Drospirenone-Containing Oral
Contraceptive Pills and the Risk of Venous Thromboembolism: An Assessment of
Risk in First-Time Users and Restarters. Drug Saf. 2017;40(7):583-596.
89. van Vlijmen EF, Wiewel-Verschueren S, Monster TB, et al. Combined oral
contraceptives, thrombophilia and the risk of venous thromboembolism: a
systematic review and meta-analysis. J Thromb Haemost. 2016;14(7):1393-403.
•• This systematic review and meta-analysis summarizes the additional risk of
venous thromboembolism in combined oral contraceptives-users and
thrombophilia
90. Hugon-Rodin J, Horellou MH, Conard J, et al. Type of combined contraceptives,
factor V Leiden mutation and risk of venous thromboembolism. Thromb Haemost.
2018;118(5):922-928
91. WHO. Medical Eligibility Criteria for Contraceptive use. Reproductive Health and
Research, 5th ed Geneva, Switzerlanda: World Health Organization. 2015.
92. Middeldorp S. Is thrombophilia testing useful? Hematology Am Soc Hematol Educ
Program. 2011;2011:150-5.
93. Horowitz N, Brenner B. Thrombophilia and cancer. Pathophysiol Haemost Thromb.
2008;36(3-4):131-6.
94. Blom JW, Doggen CJ, Osanto S, et al. Malignancies, prothrombotic mutations, and
the risk of venous thrombosis. JAMA. 2005;293(6):715-22.
95. Eroglu A, Ulu A, Cam R, et al. Prevalence of Factor V 1691 G-A (Leiden) and
prothrombin G20210A polymorphisms and the risk of venous thrombosis among
cancer patients. J Thromb Thrombolysis. 2007;23(1):31-4.
96. Dentali F, Gianni M, Agnelli G, et al. Association between inherited thrombophilic
abnormalities and central venous catheter thrombosis in patients with cancer: a
meta-analysis. J Thromb Haemost. 2008;6(1):70-5.
97. Decousus H, Moulin N, Quenet S, et al. Thrombophilia and risk of venous
thrombosis in patients with cancer. Thromb Res. 2007;120 Suppl 2:S51-61.
98. Falvella FS, Cremolini C, Miceli R, et al. Variant alleles in factor V, prothrombin,
plasminogen activator inhibitor-1, methylenetetrahydrofolate reductase and risk of
thromboembolism in metastatic colorectal cancer patients treated with first-line
chemotherapy plus bevacizumab. Pharmacogenomics J. 2017;17(4):331-336.
Accepted Manuscript
99. Caruso V, Iacoviello L, Di Castelnuovo A, et al. Thrombotic complications in
childhood acute lymphoblastic leukemia: a meta-analysis of 17 prospective studies
comprising 1752 pediatric patients. Blood. 2006;108(7):2216-22.
100. Dusse LMS, Silva MVF, Freitas LG, et al. Economy class syndrome: what is it and
who are the individuals at risk? Rev Bras Hematol Hemoter. 2017;39(4):349-353.
101. Chandra D, Parisini E, Mozaffarian D. Meta-analysis: travel and risk for venous
thromboembolism. Ann Intern Med. 2009;151(3):180-90.
102. Martinelli I, Taioli E, Battaglioli T, et al. Risk of venous thromboembolism after air
travel: interaction with thrombophilia and oral contraceptives. Arch Intern Med.
2003;163(22):2771-4.
103. Thibeault C, Evans AD. AsMA Medical Guidelines for Air Travel: Airline Special
Services. Aerosp Med Hum Perform. 2015;86(7):657-8.
104. Nishimoto Y, Yamashita Y, Morimoto T, et al. Risk factors for post-thrombotic
syndrome in patients with deep vein thrombosis: from the COMMAND VTE registry.
Heart Vessels. 2018.
105. Rabinovich A, Cohen JM, Prandoni P, et al. Association between thrombophilia and
the post-thrombotic syndrome: a systematic review and meta-analysis. J Thromb
Haemost. 2014;12(1):14-23.
106. Spiezia L, Campello E, Valle FD, et al. ABO blood group and the risk of post-
thrombotic syndrome. Ann Hematol. 2018;97(6):1057-1060.
Accepted Manuscript
Accepted Manuscript
Table 1. Increased thrombotic risk in hereditary and acquired thrombophilia
Thrombophilia Relative risk for a first VTE
(compared to community controls)
Hereditary thrombophilia
Factor V Leiden
Heterozygous
Homozygous
3-7x
80x
Prothrombin G20210A
Heterozygous
Homozygous
2-3x
5x
Double heterozygosity (FVL and prothrombin G20210A)
AT deficiency
6x
5x
Protein C deficiency 4-6.5x
Protein S deficiency 1-3x
Pseudohomozygous FVL 80x
Factor IX Padua 10x
AT resistance 2-3x
Non-0 blood type
Factor VIII 150 IU/dL
Factor IX 129 IU/dL
Factor XI 121 IU/dL
2x
3-5x
2.3x
2x
Acquired thrombophilia
Antiphospholipid antibody syndrome 3-10x
Hyperhomocysteinaemia 1.5-3x
Adapted from [2-4, 62], Phillippe HM et al. Inherited Thrombophilia. J Pharm Pract. 2014; 27(3) 227-233;
Makris M. Thrombophilia: grading the risk. Blood. 2009;113(21):5038-9; Bauer KA et al. Overview of the
causes of venous thrombosis. www.uptodate.com ©2018 UpToDate.
AT: antithrombin; FVL: factor V Leiden; VTE: venous thromboembolism.
Accepted Manuscript
Table 2. Thrombotic risk in acquired thrombophilia and main hypercoagulable changes in
coagulation pathways
Acquired thrombophilia Relative risk
for a first VTE
Haemostatic changes
Surgery
Trauma
Pregnancy
Oestrogen–progestogen therapies
Cancer
Myeloproliferative neoplasms
Economy class syndrome
1.7-2.8x
3-5x
5-50x
2-9x
4-7x
3x
2-4x
release or exposure of TF
AT
hypofibrinolysis ( PAI-1)
procoagulant factors (fibrinogen, VII, VIII,
X, vW)
protein S
acquired APC resistance
protein S
TFPI
release of TF
procoagulant factors
anticoagulant factors
platelet activation
procoagulant factors
protein S
acquired APC resistance
platelet-induced thrombin generation
Hypofibrinolysis ( prothrombin fragment
F1+F2, tPA)
Haemoconcentration
procoagulant factors (fibrinogen, VII, VIII,
X, vW)
hypofibrinolysis ( PAI-1)
platelet aggregation
Accepted Manuscript
Obesity (BMI 30 Kg/m2)
2-3x
Adapted from [62-65,98], Sandor T. Travel thrombosis: pathomechanisms and clinical aspects.
Pathophysiology. 2008;15(4):243–52; Bauer KA et al. Overview of the causes of venous thrombosis.
www.uptodate.com ©2018 UpToDate.
TF: tissue factor; AT: antithrombin; PAI-1: plasminogen activator inhibitor-1; BMI: body mass index; vW: von
Willebrand; APC: activated protein C; TFPI: tissue factor pathway inhibitor, tPA: tissue plasminogen
activator.
... Besides, PS functions as a cofactor of tissue factor pathway inhibitor (TFPI), which inhibits the activity of tissue factor 24 , the main trigger of blood coagulation in vivo. Protein S deficiency is among the most frequent risk factors in thrombophilia 25 and is clinically a risk factor for thromboembolic disease 26-28 . Therefore, this case control study hypothesizes that the IF during Ramadan has the potential to alter the levels of the natural anticoagulants (AT, PC and PS) and operate as a protective antithrombotic mechanism. ...
Article
Full-text available
Data on the pathophysiological mechanisms of hemostatic alterations in the thrombotic events that occur during Ramadan intermittent fasting (RIF), particularly in the natural coagulation inhibitors, are very limited. Thus, our objective was to evaluate the effect of RIF on the natural anticoagulants level, antithrombin, protein C, and total and free protein S (PS) in healthy participants. Participants were divided into two groups. Group I consisted of 29 healthy fasting participants whose blood samples were taken after 20 days of fasting. Group II included 40 healthy non-fasting participants whose blood samples were taken 2–4 weeks before the month of Ramadan. Coagulation screening tests including prothrombin time (PT), activated partial thromboplastin time (APTT) and plasma fibrinogen level, natural anticoagulants; antithrombin, protein C, free and total PS and C4 binding protein (C4BP) levels were evaluated in the two groups. High levels of total and free PS without change in antithrombin, protein C, and C4BP levels were noted in the fasting group as compared with non-fasting ones (p < 0.05). PT and APTT showed no difference between the two groups. However, the fibrinogen level was higher in the fasting group. In conclusion, RIF was found to be associated with improved anticoagulant activity in healthy participants, which may provide temporal physiological protection against the development of thrombosis in healthy fasting people.
... The coagulation dysfunction of femoral head necrosis is affected by two molecular pathways, namely, thrombolysis and hypofibrinolysis. Hypercoagulability, also known as hypercoagulability, is an abnormality of the blood clotting mechanism that promotes the formation of blood clots in the walls of circulating blood vessels [55], characterized by an increase in procoagulant factors or a decrease in natural anticoagulants [56]. Glucocorticoid-induced fibrinolysis is the process of breaking down blood clots or clots that is tightly regulated by activators such as tissue plasminogen activators (TPA) and urokinase-type plasminogen activators (UPA), as well as by inhibitors such as tissue factor plasminogen inhibitors (TFPI) and plasminogen activator inhibitor-1(PAI-1), as well as plasminogen proteinases. ...
Article
Full-text available
Hormonal necrosis of the femoral head is caused by long-term use of glucocorticoids and other causes of abnormal bone metabolism, lipid metabolism imbalance and blood microcirculation disorders in the femoral head, resulting in bone trabecular fracture, bone tissue necrosis collapse, and hip dysfunction. It is the most common type of non-traumatic necrosis of the femoral head, and its pathogenesis is complex, while impaired blood circulation is considered to be the key to its occurrence. There are a large number of microvessels in the femoral head, among which H-type vessels play a decisive role in the “angiogenesis and osteogenesis coupling”, and thus have an important impact on the occurrence and development of femoral head necrosis. Glucocorticoids can cause blood flow injury of the femoral head mainly through coagulation dysfunction, endothelial dysfunction and impaired angiogenesis. Glucocorticoids may inhibit the formation of H-type vessels by reducing the expression of HIF-1α, PDGF-BB, VGEF and other factors, thus causing damage to the “angiogenesis-osteogenesis coupling” and reducing the ability of necrosis reconstruction and repair of the femoral head. Leads to the occurrence of hormonal femoral head necrosis. Therefore, this paper reviewed the progress in the study of the mechanism of hormone-induced femoral head necrosis based on microvascular blood flow at home and abroad, hoping to provide new ideas for the study of the mechanism of femoral head necrosis and provide references for clinical treatment of femoral head necrosis.
... Pregnancy is a condition that promotes blood hypercoagulability, becoming more apparent as the pregnancy progresses [16]. The adequate development of placental circulation is crucial for a successful pregnancy, and genetic predisposition to blood clotting disorders can pose a risk for complications related to placental function during pregnancy [17]. The concept of biological hypercoagulability refers to changes in coagulation and fibrinolysis processes that lean toward clot formation during pregnancy. ...
Article
Full-text available
Background: Despite pregnancy’s hypercoagulable state, the correlation between inher- ited thrombophilia and thrombotic adverse pregnancy outcomes remains uncertain. The objective of this study was to determine the prevalence of inherited thrombophilic polymorphisms among asymptomatic pregnant individuals and to examine their potential correlation with adverse perina- tal outcomes. Methods: in this single-center prospective study, 105 healthy pregnant women were included. Genotyping was conducted for factor V Leiden (FVL), prothrombin gene mutation, meth- ylenetetrahydrofolate reductase enzyme (MTHFR) C677T, MTHFR A1298C, and plasminogen acti- vator inhibitor-1 (PAI-1), alongside the assessment of protein C (PC), protein S (PS), and antithrom- bin (AT) levels. The study analyzed the association between inherited thrombophilic polymor- phisms and pregnancy complications linked to placental insufficiency, such as gestational hyper- tension (GH), preeclampsia (PE), intrauterine death (IUD), fetal growth restriction (FGR), and pla- cental abruption. Results: The prevalence of identifiable thrombophilic polymorphism mutations was 61.9% (95% confidence interval - CI 52.4–70.8%), with the most common single mutation being PAI-1 4G/5G (12/105, 11.4%, 95% CI 6.4–18.5). The most frequent combined mutation was heterozy- gosity for MTHFR C677T and PAI-1 (12/105, 11.4%, 95% CI 6.4–18.5). Notably, no FVL homozygous carriers or single homozygous and heterozygous carriers for prothrombin polymorphisms were found. Additionally, no deficiencies in PC and AT were detected among participants. Except for homozygosity for PAI-1, none of the studied polymorphisms demonstrated a significant association with pregnancy complications linked to placental insufficiency. Conclusions: The asymptomatic carriers of inherited thrombophilic polymorphisms do not have an increased risk of adverse peri- natal outcomes.
... To obtain data on the anthropometric and socioeconomic characteristics, as well as the genetic and risk factor profiles, of expectant women in western Romania [18]. The prevalence of inherited and acquired Thrombophilia was investigated in this study [19], which analyzed data from a massive RM patient group. Hereditary thrombophilia, a condition of blood coagulation caused by genetics, accounts for almost 60% of idiopathic thromboembolic events. ...
Article
Full-text available
Background: A category of hereditary diseases known as Thrombophilia induces irregular blood clotting. Pre-eclampsia, late miscarriages, premature delivery, fetal growth restriction, and deep venous thrombosis are all associated with Thrombophilia. A higher incidence of recurrent pregnancy loss (RPL) or recurrent early pregnancy loss (REPL) in the initial 6-10 weeks of conception may be seen in homozygous individuals for particular thrombophilic variables. A healthy uteroplacental circulation is necessary for a favorable pregnancy outcome. It has been hypothesized that maternal Thrombophilia could raise the probability of preeclampsia and intrauterine growth retardation (IUGR). ACE D/D (angiotensin-converting enzyme deletion/deletion) genotype and PAI 4G/4G (plasminogen activator inhibitor-1 4G/4G) genotype have been associated with an increased risk for these conditions and high blood pressure (hypertension). Aim of this study is to determinate the frequency of the different thrombophilic mutations in patients with recurrent pregnancy loss. Methods: A prospective cohort study included 309 pregnant patients with gestational age from 6 to 12 gestational weeks. The patients have a history of recurrent pregnancy loss in the past. Inherited Thrombophilia test was performed on each research participants as part of preconception counseling. Results: Plasminogen activator inhibitor 1(PAI-I) 4G/4G was the most prevalent form of thrombophilic mutation identified (82,83%), followed by MTHFR mutation (85.43%), mutations in the prothrombin gene (12,61%) and factor V Leiden (12.29%). Additionally, (12.62%) of pregnant women had IUGR babies, (29,44%) of patients had preeclampsia as a diagnosis and gestational diabetes (24,27%). In conclusion-The genetic landscape significantly impacts the trajectory of pregnancy-related complications. This study sheds light on the pivotal roles played by genetic mutations, elucidating potential avenues for refined clinical management and targeted interventions. The early detection of these genetic variations paves the way for enhanced personalized care strategies, ultimately benefitting the well-being of both maternal and fetal domains.
... Although the clinical manifestation of hereditary thrombophilia varies, thrombosis can occasionally occur in rare locations such the splanchnic veins, cerebral veins, and retinal vein. Some people never get thrombosis, some may not show symptoms until they are adults, and some develop recurrent thromboembolism before the age of 30 (5,16). Regarding genetic mutations, there are many cases of thrombophilia linked to inherited mutations in genes such as Factor V Leiden and Prothrombin G20210A (17). ...
Article
Full-text available
Thrombophilia, characterized by an increased tendency to form blood clots, presents a substantial risk during pregnancy, potentially impacting maternal and fetal outcomes. This review explores the intricate relationship between thrombophilia and oxidative stress, shedding light on their combined influence on blood clot formation. Specifically, endothelial dysfunction, driven by oxidative stress, emerges as a pivotal factor in thrombophilia, setting the stage for increased platelet activation and altered coagulation factors. Factors like Factor V Leiden mutation, Prothrombin G20210A mutation, and deficiencies in antithrombin, protein C, and protein S contribute to the pro-thrombotic state observed in thrombophilia. Furthermore, inflammation, closely intertwined with oxidative stress, exacerbates the risk of blood clot formation. Inflammatory responses lead to endothelial activation, altered endothelial function, and increased adhesion molecules expression, disrupting the delicate balance between pro- and anti-coagulant factors. Chronic inflammatory conditions, such as autoimmune disorders, potentiate a persistent state of heightened clotting risk. Additionally, impaired antioxidant defenses compound the prothrombotic state by allowing the accumulation of reactive oxygen species, thereby contributing to oxidative stress-induced endothelial dysfunction. Understanding the interplay between these factors is crucial for tailored thrombophilia management, particularly in pregnancy. Treatment strategies encompass a multifaceted approach, including anticoagulant medications, lifestyle modifications, and targeted interventions to improve endothelial health. The complex nature of thrombophilia underscores the need for a collaborative healthcare approach, involving hematologists and high-risk pregnancy specialists. Early detection, vigilant monitoring, and timely intervention are paramount in mitigating risks for both mother and baby.
... La presencia de factores predisponentes, como ciertos elementos genéticos específicos y una variedad de factores adquiridos, es cada vez más común en los pacientes. Entre estos factores adquiridos se encuentran estados inflamatorios como sepsis, neoplasias y enfermedades sistémicas, uso de fármacos procoagulantes, y daño directo causado por cirugías o dispositivos invasivos [2][3][4] . Estas condiciones son especialmente prevalentes en pacientes que requieren un mayor nivel de soporte médico, siendo la unidad de cuidados intensivos (UCI) el entorno donde se conjugan en mayor medida estos elementos. ...
Article
Full-text available
A 65-year-old woman, with diffuse large B-cell lymphoma and catastrophic respiratory failure due to COVID-19, requiring extracorporeal membrane oxygenation support. She developed macro agglutination and multisystem arterio-venous thrombosis. Which factor activated the coagulation cascade? Le Grand Mort: la tríada, o más bien orgía, de Virchow que acabó en muerte Macroagglutination as cause of death: when Virchow's perfect storm strikes
... In recent years, with the aging of the population and changes in human lifestyles, human diseases caused by thrombosis and fat embolism, such as acute myocardial infarction, pulmonary embolism, and fat embolism syndrome, which seriously jeopardize human life and health, have become more common (1,2). Prompt diagnosis and early treatment are key to improving prognosis. ...
Article
Full-text available
Background The early diagnosis of thrombosis and fat embolism is important for subsequent treatment regimens. Spectral computed tomography (CT) virtual non-contrast (VNC) scanning can not only accurately diagnose thrombosis and medium fat embolism but can also reduce the radiation dose and scanning time. However, there is a relative paucity of studies on what contrast concentration and exposure conditions are best for the quality of VNC images. To address this issue, this study aimed to investigate the effects of different exposure conditions and contrast concentrations on the quality of VNC images of low-density substances in spectral CT. Methods Four solution groups [i.e., groups A (15 mgI/mL), B (10 mgI/mL), C (5 mgI/mL), and D (the control group)] were matched with normal saline and contrast agent groups. Four groups of solution, duck blood clots, and fat were injected into four sections of the pig large intestine, respectively. CT scans with different exposure amounts were performed under the condition of 120 KV. Comparing the true non-contrast (TNC) image based on solution D group with the VNC images of the other three solution groups. The differences in the CT values, standard deviation (SD) values, and contrast noise ratio (CNR) values of the duck blood and fat under different iodine concentrations and exposures were compared. The image quality was evaluated using a three-point method and the Kappa consistency test was performed. The consistency of the tissue CT values in the TNC and VNC images was analyzed by drawing Bland-Altman scatter plots. Results The CT values of the duck blood in the VNC20mAs and VNCC groups were lower than those in the TNC groups (P<0.05). Under different exposures and contrast agent concentrations, the CT value of the fat in the VNC group was higher than that in the TNC group (P<0.05). The SD values of the duck blood and fat in three groups (i.e., groups A, B, and C) were lower than those in the TNC group (P<0.05). The CNR value of the duck blood in the VNC20mAs group was lower than that in the TNC group (Z=−2.10, P=0.04), and the CNR values of the duck blood and fat in the VNC group were higher than those in the TNC groups in the remaining different exposure and concentration groups (P<0.05). The CT values of the lesions in the two groups were consistent, and there were no statistically significant differences between the subjective scores of the TNC and VNC images (z=−1.34, P=0.18); the subjective evaluations of the two physicians had good consistency (K=0.80). Conclusions Under the conditions of higher contrast agent concentrations and proper exposure conditions, the VNC images were better able to restore the CT values of the blood clots, reduce the SD values of the blood clots and fat. In addition, and improve the CNR values of the blood clots and fat. In addition, the quality of the two images was similar.
Article
Introduction: Acute ischemic stroke (AIS) stands as a leading cause of death and disability globally. This study aimed to investigate the risk factors and relevance linked with AIS in patients undergoing maintenance hemodialysis (MHD) and to create and validate nomogram models. Methods: We examined the medical records of 314 patients with stage 5 chronic kidney disease (CKD 5) undergoing MHD, who sought neurology outpatient department consultation for suspected AIS symptoms between January 2018 and December 2023. These 314 patients were randomly divided into the training cohort (n = 222) and validation cohort (n = 92). The least absolute shrinkage selection operator (LASSO) regression model was employed for optimal feature selection in the AIS risk model. Subsequently, multivariable logistic regression analysis was used to construct a predictive model incorporating the features selected through LASSO. This predictive model's performance was assessed using the C-index and the area under the receiver operating characteristic curve (AUC). Additionally, calibration and clinical utility were evaluated through calibration plots and decision curve analysis (DCA). The model's internal validation was conducted using the validation cohort. Results: Predictors integrated into the prediction nomogram encompassed cardiovascular disease (CVD) (odds ratio [OR] 7.95, 95% confidence interval [CI] 2.400-29.979), smoking (OR 5.7, 95% CI: 1.661-21.955), dialysis time (OR: 5.91, 95% CI: 5.866-29.979), low-density lipoprotein (OR: 2.99, 95% CI: 0.751-13.007), and fibrin degradation products (OR: 5.47, 95% CI: 1.563-23.162). The model exhibited robust discrimination, with a C-index of 0.877 and 0.915 in the internal training and validation cohorts, respectively. The AUC for the training set was 0.857, and a similar AUC of 0.905 was achieved in the validation cohort. DCA demonstrated a positive net benefit within a threshold risk range of 2-96%. Conclusion: The proposed nomogram effectively identifies MHD patients at high risk of AIS at an early stage. This model holds the potential to aid clinicians in making preventive recommendations.
Article
Background Thrombophilia is a coagulation disorder closely associated with venous thromboembolism. Hereditary antithrombin III (AT III) deficiency is a type of genetic thrombophilia. In China, genetic thrombophilia patients mainly suffer from deficiencies in AT III, protein S, and protein C. Multiple mutations in the serpin family C member 1 ( SERPINC1 ) can affect AT III activity, resulting in thrombosis. Case presentation This case presented a 17-year-old adolescent female who developed lower extremity venous thrombosis and subsequently pulmonary embolism (PE) following a right leg injury. A missense mutation in gene SERPINC1 of c.331 T > C, p.S111P was detected on the patient, resulting in a decreased AT III activity and an elevated risk of thrombosis. The patient received anticoagulation treatment for approximately 5 months. During follow-up, the blood clot gradually dissolved, and there have been no recurrent thrombotic events reported thus far. Discussion Hereditary AT deficiency can be classified into two types based on the plasma levels of the enzymatic activity and antigen. Type I is a quantitative defect, while Type II is a qualitive defect. Until 2021, 486 SERPINC1 gene mutations have been registered, more than 18% of which are point mutations. The SERPINC1 mutation c.331 T > C in was firstly reported in 2017, which was classified into type I AT III deficiency. Conclusion Hereditary thrombophilia is a coagulation disorder with a high omission diagnostic rate. Minor mutations in the SERPINC1 gene can also lead to hereditary AT III deficiency, which in turn can cause PE. We emphasized the importance of etiological screening for hereditary thrombophilia in venous thromboembolism patients without obvious high-risk factors. Long-term anticoagulation treatment and avoidance of potential thrombosis risk factors are critical for such patients.
Article
Full-text available
Vitamin K-dependent protein S exists in two forms in plasma, as free protein and in a bimolecular, noncovalent complex with the regulatory complement protein C4b-binding protein (C4BP). The effects of C4BP on the protein Ca cofactor activity of protein S were studied in a plasma system and in a system using purified components from both human and bovine origin. Bovine protein S was found to interact with human C4BP with a 5-fold higher affinity than that observed for the interaction between human protein S and human C4BP. The binding of protein S, from either species, to human C4BP results in the loss of the protein Ca cofactor function. In bovine plasma, protein S could be totally complexed by the addition of human C4BP, with a concomitant total loss of protein Ca cofactor activity. The addition of purified human C4BP to human plasma resulted in only partial loss of protein Ca cofactor activity and the plasma protein S was not completely complexed. Human protein S functioned as a cofactor to human protein Ca, but not to bovine protein Ca, whereas bovine protein S demonstrated very little species specificity and functioned as a cofactor both with human and bovine protein Ca. The species specificity of the protein Ca-protein S interaction was useful in elucidating the effect of C4BP in the plasma system. In the system with purified bovine components, protein S was required for the degradation of factor Va by low concentrations of protein Ca, whereas in the system with human components protein Ca alone, even when added at very low concentrations, exhibited potential to degrade factor Va, and the presence of protein S only enhanced the reaction rate approximately 5-fold. In both these systems, the stimulating effect of protein S on factor Va degradation by protein Ca was completely lost when protein S bound to C4BP.
Article
Context.—Homocysteine, a sulfur-containing amino acid, absent in natural diets, is a metabolic intermediary in transmethylation and transsulfuration reactions. Such reactions are essential to normal cellular growth, differentiation, and function. Excess homocysteine is associated with vascular disease and related disorders. Objective.—To review homocysteine metabolism, the pathogenesis and classification of hyperhomocysteinemia, and the published literature investigating the association of homocysteine and methylenetetrahydrofolate reductase defects with arterial and venous thromboembolism and related disorders. The role of vitamin supplementation in patients with hyperhomocysteinemia is addressed. Data Sources.—Published medical and scientific literature. Articles addressing the objectives were selected and reviewed. Pertinent studies and conclusions were summarized, grouped, and contrasted. Conclusions.—The association of hyperhomocysteinemia and arterial and venous thrombosis is controversial. Severe hyperhomocysteinemia is associated with atherosclerosis. The effect of mild hyperhomocysteinemia is less certain. Coinheritance of methylenetetrahydrofolate reductase defects and factor V Leiden is likely to increase the risk of venous thromboembolism. The association of methylenetetrahydrofolate reductase defects combined with no additional thrombophilic risk factors with venous thrombosis is less clear. High doses of folic acid to lower homocysteine levels might not be necessary.
Article
In this study, we test the hypothesis that prothrombin levels may modulate activated protein C (APC) anticoagulant activity. Prothrombin in purified systems or plasma dramatically inhibited the ability of APC to inactivate factor Va and to anticoagulate plasma. This was not due solely to competition for binding to the membrane surface, as prothrombin also inhibited factor Va inactivation by APC in the absence of a membrane surface. Compared with normal factor Va, inactivation of factor Va Leiden by APC was much less sensitive to prothrombin inhibition. This may account for the observation that the Leiden mutation has less of an effect on plasma-based clotting assays than would be predicted from the purified system. Reduction of protein C levels to 20% of normal constitutes a significant risk of thrombosis, yet these levels are observed in neonates and patients on oral anticoagulant therapy. In both situations, the correspondingly low prothrombin levels would result in an increased effectiveness of the remaining functional APC of ≈5-fold. Thus, while the protein C activation system is impaired by the reduction in protein C levels, the APC that is formed is a more effective anticoagulant, allowing protein C levels to be reduced without significant thrombotic risk. In situations where prothrombin is high and protein C levels are low, as in early stages of oral anticoagulant therapy, the reduction in protein C would result only in impaired function of the anticoagulant system, possibly explaining the tendency for warfarin-induced skin necrosis.
Article
Factor V gene polymorphisms were investigated to detect components that may contribute to the activated protein C (APC) resistance phenotype in patients with venous thromboembolism. A specific factor V gene haplotype (HR2) was defined by six polymorphisms and its frequency was found to be similar in normal subjects coming from Italy (0.08), India (0.1), and Somalia (0.08), indicating that it was originated by ancestral mutational events. The relationship between the distribution of normalized APC ratios obtained with the functional assay and haplotype frequency was analyzed in patients heterozygous for factor V R506Q (factor V Leiden). The HR2 haplotype was significantly more frequent in patients with ratios below the 15th percentile than in those with higher ratios or in normal controls. Moreover, the study of 10 patients with APC resistance in the absence of the factor V R506Q mutation showed a 50-fold higher frequency of HR2 homozygotes. The HR2 haplotype was associated with significantly lower APC ratios both in patients with venous thromboembolism and in age- and sex-matched controls. However, the two groups showed similar HR2 haplotype frequencies. Plasma mixing experiments showed that an artificially created double heterozygote for the factor V R506Q mutation and the HR2 haplotype had an APC ratio lower than that expected for a simple R506Q heterozygote. Time-course experiments evaluating the decay of factor V in plasma showed the normal stability of the molecule encoded by the factor V gene marked by the HR2 haplotype, which ruled out the presence of a pseudo-homozygous APC resistance mechanism. Our results provide new insights into the presence of factor V genetic components other than the factor V R506Q that are able to contribute to the APC resistance phenotype in patients with venous thromboembolism.
Article
Elevated plasma levels of factor VIII (> 150 IU/dL) are an important risk factor for deep vein thrombosis (DVT). Factor VIII is the cofactor of factor IXa in the activation of factor X. The risk of thrombosis in individuals with an elevated factor IX level is unknown. This study investigated the role of elevated factor IX levels in the development of DVT. We compared 426 patients with a first objectively diagnosed episode of DVT with 473 population controls. This study was part of a large population-based case-control study on risk factors for venous thrombosis, the Leiden Thrombophilia Study (LETS). Using the 90th percentile measured in control subjects (P90 = 129 U/dL) as a cutoff point for factor IX levels, we found a 2- to 3-fold increased risk for individuals who have factor IX levels above 129 U/dL compared with individuals having factor IX levels below this cutoff point. This risk was not affected by adjustment for possible confounders (age, sex, oral contraceptive use, and high levels of factor VIII, XI, and vitamin K-dependent proteins). After exclusion of individuals with known genetic disorders, we still found an odds ratio (OR) of 2.5 (95% confidence interval [CI]: 1.6-3.9). The risk was higher in women (OR: 2.6, CI: 1.6-4.3) than in men (OR: 1.9, CI: 1.0-3.6) and appeared highest in the group of premenopausal women not using oral contraceptives (OR: 12.4, CI: 3.3-47.2). These results show that an elevated level of factor IX is a common risk factor for DVT.
Article
In order to firmly establish a normal range for von Willebrand factor antigen (vWF:Ag), we determined plasma vWF:Ag concentrations in 1,117 volunteer blood donors by quantitative immunoelectrophoresis. The presence of the ABO blood group has a significant influence on vWF:Ag values; individuals with blood group O had the lowest mean vWF:Ag level (74.8 U/dL), followed by group A (105.9 U/dL), then group B (116.9 U/dL), and finally group AB (123.3 U/dL). Multiple regression analysis revealed that age significantly correlated with vWF:Ag levels in each blood group. We then performed reverse ABO typing on stored plasma from 142 patients with the diagnosis of von Willebrand disease (vWd). Of 114 patients with type I vWd, blood group O was found in 88 (77%), group A in 21 (18%), group B in 5 (4%), and group AB in none (0%), whereas the frequency of these blood groups in the normal population is significantly different (45%, 45%, 7% and 3%, respectively) (P less than .001). Patients with type II or III vWd had ABO blood group frequencies that were not different from the expected distribution. There may be a subset of symptomatic vWd patients with decreased concentrations of structurally normal vWf (vWd, type I) on the basis of blood group O. Some individuals of blood group AB with a genetic defect of vWF may have the diagnosis overlooked because vWF levels are elevated due to blood type.
Article
Post-thrombotic syndrome (PTS) is the most common chronic complication of deep vein thrombosis (DVT). Identifying high-risk patients for the development of PTS might be useful for its prevention. The COMMAND VTE Registry is a multicenter registry that enrolled 3027 consecutive patients with acute symptomatic venous thromboembolisms (VTEs) in Japan between January 2010 and August 2014. The current study population consisted of 1298 patients with lower extremities DVTs who completed 3-year follow-up for those who developed PTS and those without PTS. We investigated risk factors for the development of PTS at the time of DVT diagnosis, using a multivariable logistic regression analysis. Of the entire 1298 study patients, 169 (13%) patients were diagnosed with PTS within 3 years. The rate for anticoagulation discontinuation during follow-up was not significantly different between those with and without PTS. Chronic kidney disease (OR 2.21, 95% CI 1.45–3.39, P < 0.001), leg swelling (OR 4.15, 95% CI 2.25–7.66, P < 0.001), absence of transient risk factors for VTEs (OR 2.39, 95% CI 1.55–3.67, P < 0.001), active cancer (OR 3.66, 95% CI 2.30–5.84, P < 0.001), and thrombophilia (OR 2.07, 95% CI 1.06–4.04, P = 0.03) were independent risk factors for the development of PTS. In this real-world Japanese DVT registry, we could identify several important risk factors for the development of PTS at the time of DVT diagnosis.
Article
Inherited and acquired thrombophilias and hypercoagulable states, such as active cancer, estrogen-induced, autoimmune disorders, major surgery, hospitalization, and trauma, are well-known risk factors for venous thromboembolism (VTE). The effect of these on recurrent VTE is different for each specific risk factor. The major risk factors affecting VTE recurrence include the presence of active cancer and an unprovoked first VTE. In addition, the use of combined female hormones in a woman with a previous history of estrogen-related VTE is a major risk factor for VTE recurrence. The extent of influence of inherited thrombophilia on the risk of recurrence is controversial. Conversely, the presence of antiphospholipid antibodies, specifically triple positive carriers, appears to increase the risk of VTE recurrence. Understanding the rates of recurrent VTE in a patient and the individual risk of bleeding is important in determining the duration of anticoagulation therapy.