ArticlePDF AvailableLiterature Review

Portal Vein Thrombosis in the Setting of Cirrhosis: A Comprehensive Review

MDPI
Journal of Clinical Medicine
Authors:

Abstract and Figures

Portal vein thrombosis constitutes the most common thrombotic event in patients with cirrhosis, with increased rates in the setting of advanced liver disease. Despite being a well-known complication of cirrhosis, the contribution of portal vein thrombosis to hepatic decompensation and overall mortality is still a matter of debate. The incorporation of direct oral anticoagulants and new radiological techniques for portal vein recanalization have expanded our therapeutic arsenal. However, the lack of large prospective observational studies and randomized trials explain the heterogenous diagnostic and therapeutic recommendations of current guidelines. This article seeks to make a comprehensive review of the pathophysiology, clinical features, diagnosis, and treatment of portal vein thrombosis in patients with cirrhosis.
Content may be subject to copyright.
Citation: Odriozola, A.; Puente, Á.;
Cuadrado, A.; Rivas, C.; Anton, Á.;
González, F.J.; Pellón, R.; Fábrega, E.;
Crespo, J.; Fortea, J.I. Portal Vein
Thrombosis in the Setting of
Cirrhosis: A Comprehensive Review.
J. Clin. Med. 2022,11, 6435. https://
doi.org/10.3390/jcm11216435
Academic Editor: Antonio
M Caballero-Mateos
Received: 8 October 2022
Accepted: 28 October 2022
Published: 30 October 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
Journal of
Clinical Medicine
Review
Portal Vein Thrombosis in the Setting of Cirrhosis:
A Comprehensive Review
Aitor Odriozola 1,Ángela Puente 1, Antonio Cuadrado 1, Coral Rivas 1,Ángela Anton 1,
Francisco JoséGonzález 2, Raúl Pellón2, Emilio Fábrega 1, Javier Crespo 1and JoséIgnacio Fortea 1, *
1Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases,
Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39008 Santander, Spain
2Radiology Department, Marqués de Valdecilla University Hospital, 39008 Santander, Spain
*Correspondence: joseignacio.fortea@scsalud.es; Tel.: +34-942202520 (ext. 72929)
Abstract:
Portal vein thrombosis constitutes the most common thrombotic event in patients with
cirrhosis, with increased rates in the setting of advanced liver disease. Despite being a well-known
complication of cirrhosis, the contribution of portal vein thrombosis to hepatic decompensation
and overall mortality is still a matter of debate. The incorporation of direct oral anticoagulants and
new radiological techniques for portal vein recanalization have expanded our therapeutic arsenal.
However, the lack of large prospective observational studies and randomized trials explain the
heterogenous diagnostic and therapeutic recommendations of current guidelines. This article seeks
to make a comprehensive review of the pathophysiology, clinical features, diagnosis, and treatment
of portal vein thrombosis in patients with cirrhosis.
Keywords:
portal hypertension; cirrhosis; portal vein thrombosis; anticoagulation; transjugular
intrahepatic portosystemic shunt
1. Introduction
Portal vein obstruction can occur due to a malignant tumor (frequently but improperly
referred to as malignant thrombosis), in which the obstruction is secondary to portal vein
narrowing, and/or direct invasion of the portal vein by the neoplasm, or due to non-
malignant portal vein thrombosis (PVT). The latter is defined as a thrombus that develops
within the portal vein trunk and intrahepatic portal branches, which may also involve
the splenic (SV) or superior mesenteric veins (SMV). In the absence of recanalization,
the portal venous lumen is obliterated, and porto-portal collaterals develop, resulting
in portal cavernoma. Non-malignant PVT can arise in two settings depending on the
presence/absence of cirrhosis. This differentiation is critical since etiology, manifestations,
natural history, and therapeutic options differ [
1
4
]. Previous early literature combined
patients in these two settings and this should be accounted for to better interpret the
results [58].
This article seeks to make a comprehensive review of the pathophysiology, clinical
features, diagnosis, and treatment of PVT in patients with cirrhosis. It constitutes the
most common thrombotic event in this population, with increased rates in the setting
of advanced liver disease. Despite being a well-known complication of cirrhosis, the
contribution of PVT to hepatic decompensation and overall mortality is still a matter of
debate. There is consequently no consensus on its optimal management, and no definitive
recommendations were reported in clinical guidelines or consensus conferences [
1
4
,
9
12
].
2. Anatomy of the Venous Portal System
The liver is a highly vascular organ that receives up to 25% of the total cardiac output
from a dual blood supply. The hepatic artery delivers well-oxygenated blood and comprises
J. Clin. Med. 2022,11, 6435. https://doi.org/10.3390/jcm11216435 https://www.mdpi.com/journal/jcm
J. Clin. Med. 2022,11, 6435 2 of 26
approximately 25% of total hepatic blood flow, whereas the remaining 75% is deoxygenated
blood supplied by the portal vein [
13
]. These two afferent vascular systems drain into
the modified capillary network of the liver, composed of fenestrated sinusoids. From the
sinusoids, blood flows into the central veins that drain in the hepatic veins and then in the
inferior vena cava [2].
The hepatic portal system is the most known portal venous system in the body, hence
its name. The latter is defined as a circulatory system in which veins connect two capillary
beds without first carrying blood to the heart. In the case of the hepatic portal venous
system, capillary blood from the entire gastrointestinal tract (except for the upper esophagus
and distal rectum), pancreas, gallbladder, and spleen is carried to the hepatic sinusoids.
The portal vein is an 8-cm, valveless conduit originating from the confluence of the SMV
and SV posterior to the neck of the pancreas [
2
]. The inferior mesenteric vein (IMV) is a
tributary vessel that usually drains in the SV (up to 40% of the cases). However, there are
many variants in the drainage of this vein [
14
]. In the other two most frequent variants, the
IMV enters into the angle of the confluence of the SV and the SMV (30%) or the SMV (20%).
Other lesser frequent variants (10%) consist of accessory mesenteric veins entering the SV
or SMV.
The portal vein develops during the second and third month of gestation from two
vitelline veins, which drain the yolk sac. These veins form few anastomoses between
each other which result in the formation of the portal vein. Deviations from the normal
process of these anastomosis result in variations in the branching pattern of the portal
vein. Its distribution within the liver is segmental and closely follows the hepatic artery.
Cheng Y et al. classified these intrahepatic portal vein variations into five different types
(Figure 1) [15].
J. Clin. Med. 2022, 11, x FOR PEER REVIEW 2 of 26
2. Anatomy of the Venous Portal System
The liver is a highly vascular organ that receives up to 25% of the total cardiac output
from a dual blood supply. The hepatic artery delivers well-oxygenated blood and com-
prises approximately 25% of total hepatic blood flow, whereas the remaining 75% is de-
oxygenated blood supplied by the portal vein [13]. These two afferent vascular systems
drain into the modified capillary network of the liver, composed of fenestrated sinusoids.
From the sinusoids, blood flows into the central veins that drain in the hepatic veins and
then in the inferior vena cava [2].
The hepatic portal system is the most known portal venous system in the body, hence
its name. The latter is defined as a circulatory system in which veins connect two capillary
beds without first carrying blood to the heart. In the case of the hepatic portal venous
system, capillary blood from the entire gastrointestinal tract (except for the upper esoph-
agus and distal rectum), pancreas, gallbladder, and spleen is carried to the hepatic sinus-
oids. The portal vein is an 8-cm, valveless conduit originating from the confluence of the
SMV and SV posterior to the neck of the pancreas [2]. The inferior mesenteric vein (IMV)
is a tributary vessel that usually drains in the SV (up to 40% of the cases). However, there
are many variants in the drainage of this vein [14]. In the other two most frequent variants,
the IMV enters into the angle of the confluence of the SV and the SMV (30%) or the SMV
(20%). Other lesser frequent variants (10%) consist of accessory mesenteric veins entering
the SV or SMV.
The portal vein develops during the second and third month of gestation from two
vitelline veins, which drain the yolk sac. These veins form few anastomoses between each
other which result in the formation of the portal vein. Deviations from the normal process
of these anastomosis result in variations in the branching pattern of the portal vein. Its
distribution within the liver is segmental and closely follows the hepatic artery. Cheng Y
et al. classified these intrahepatic portal vein variations into five different types (Figure 1)
[15].
Figure 1. The intrahepatic portal vein has a segmental distribution and it closely follows the hepatic
artery. Cheng Y [15] et al. classified intrahepatic portal vein variations into five different types. (A).
Type 1 (6580%): after its entry through the hilium, the main portal vein (MPV) divides into a larger
right portal vein (RPV) and a smaller left portal vein (LPV). RPV is divided into an anterior branch
(supplying segments V and VIII) and a posterior branch (supplying segments VI and VII). LVP runs
horizontally to the left and then turns medially (supplying segments I, II, III, and IV). (B). Type II
(1015%): trifurcation of MVP, dividing into the right anterior and posterior branches and the LPV.
(C). Type III (0.37%): the right posterior portal branch arises directly from the MPV as its first
Figure 1.
The intrahepatic portal vein has a segmental distribution and it closely follows the hepatic
artery. Cheng Y [
15
] et al. classified intrahepatic portal vein variations into five different types.
(A) Type
1 (65–80%): after its entry through the hilium, the main portal vein (MPV) divides into a
larger right portal vein (RPV) and a smaller left portal vein (LPV). RPV is divided into an anterior
branch (supplying segments V and VIII) and a posterior branch (supplying segments VI and VII).
LVP runs horizontally to the left and then turns medially (supplying segments I, II, III, and IV).
(B) Type
II (10–15%): trifurcation of MVP, dividing into the right anterior and posterior branches and
the LPV. (
C
) Type III (0.3–7%): the right posterior portal branch arises directly from the MPV as its
first branch and the LPV is the terminal branch, arising after the origin of the right anterior portal
vein. (
D
) Type IV (0.6–2.7%): trifurcation of the RPV, in which the branch of segment VII is the first
branch of the RPV. (
E
) Type V (1.3–2.4%): trifurcation of the RPV, in which the branch of segment
VI arises early as a separate branch of the RPV. (
F
) Different variants in the drainage of the inferior
mesenteric vein (IMV). I: drainage into the splenic vein (SV). II: drainage into the confluence of the
superior mesenteric vein (SMV) and SV. III: drainage into the SMV.
J. Clin. Med. 2022,11, 6435 3 of 26
3. Classification
Terminology and classification systems of PVT vary extensively in the literature, with
most developed exclusively in the liver transplant (LT) population [
3
]. Four of the major
published PVT classification systems are presented in Table 1[
16
19
]. More recently, the
latest guidelines of the American Association for the Study of the Liver (AASLD) on
vascular disorders and the Baveno VII workshop have promoted the establishment of a
standardized terminology in describing PVT to allow comparison and external validation
of future studies. They proposed a systematic documentation of initial site, extent/degree
of luminal obstruction, and chronicity of PVT to enable subsequent evaluation of the
spontaneous course and/or response to treatment (Figure 2) [3,4].
Table 1. Classification of portal vein thrombosis according several authors.
Classification Description Categories Strengths vs. Weaknesses
Yerdel et al.,
2000 [16]
Post-transplant survival
(i) Involvement of MPV, SMV
and SV
(ii) Degree of occlusion
Grade 1: <50% occlusion
Grade 2: >50% occlusion
Grade 3: Complete PV or SV
occlusionGrade 4: Complete PV
occlusion with SMV extension
- Well-defined long-term
post-transplant survival
- Only for LT candidates
- Most widely used classification
to date
Bauer et al.,
2006 [17]
PVT response to TIPS
placement in patients listed
for LT
(i) Involvement of MPV, SMV
and SV
(ii) Degree of occlusion
(iii) Stratification by location
of clot and cavernous
transformation
Grade 1: <25% occlusion of PV
Grade 2: 26–50% occlusion
Grade 3: 51–75% occlusion
Grade 4: 76–100% occlusion
- Available data not only in LT
recipients but also in non-LT
patients and patients with
cavernous transformation
- Better characterization of the
location and degree of occlusion
than other classifications
- Only for patients who
underwent TIPS placement.
Validated in 9 patients. No
long-term outcome data.
Sarin et al.,
2016 [18]
Based on revision of previous
PVT classifications
(i) Involvement of MPV, SMV,
and SV
(ii) Degree of occlusion
(iii) Underlying liver disease
Site of PVT (Type 1, 2a, 2b, 3)
* Type 1: only trunk
* Type II: only branches (a: one, b:
both branches)
* Type 3: trunk and branches
Degree of occlusion
* Occlusive
* Non-occlusive
Duration and presentation
* Recent or chronic
* Symptomatic or asymptomatic
Extension
* SV, SMV, or both
Underlying liver disease
- “Anatomical-functional”
classification
- Complex
- No correlation with outcomes
Bhangui et al.,
2019 [19]
(i) Involvement of MPV, SMV
and SV
(ii) LT surgery choices based
on collaterals
Complex
- SYerdel grade 4 and Jamieson
and Charco grades 3/4
- Non-complex
- Yerdel grade 1–3
- Simplifies anatomical
considerations for PV
management.
- Mainly focused on LT surgical
techniques not in outcomes.
Abbreviations: LT: liver transplant; MPV: main portal vein; PH: portal hypertension; PV: portal vein;
PVT: portal
vein thrombosis; SMV: superior mesenteric vein; SV: splenic vein; TIPS: transjugular intrahepatic
portosystemic shunt.
J. Clin. Med. 2022,11, 6435 4 of 26
J. Clin. Med. 2022, 11, x FOR PEER REVIEW 3 of 26
branch and the LPV is the terminal branch, arising after the origin of the right anterior portal vein.
(D). Type IV (0.62.7%): trifurcation of the RPV, in which the branch of segment VII is the first
branch of the RPV. (E). Type V (1.32.4%): trifurcation of the RPV, in which the branch of segment
VI arises early as a separate branch of the RPV. (F). Different variants in the drainage of the inferior
mesenteric vein (IMV). I: drainage into the splenic vein (SV). II: drainage into the confluence of the
superior mesenteric vein (SMV) and SV. III: drainage into the SMV.
3. Classification
Terminology and classification systems of PVT vary extensively in the literature, with
most developed exclusively in the liver transplant (LT) population [3]. Four of the major
published PVT classification systems are presented in Table 1 [1619]. More recently, the
latest guidelines of the American Association for the Study of the Liver (AASLD) on vas-
cular disorders and the Baveno VII workshop have promoted the establishment of a stand-
ardized terminology in describing PVT to allow comparison and external validation of
future studies. They proposed a systematic documentation of initial site, extent/degree of
luminal obstruction, and chronicity of PVT to enable subsequent evaluation of the spon-
taneous course and/or response to treatment (Figure 2) [3,4].
Figure 2. Classification of portal vein thrombosis. (A). It is crucial to characterize the extension of
the portal vein thrombosis and its relationship with the main portal vein (MPV). The involvement
of the superior mesenteric vein (SMV) and splenic vein (SV) should be properly characterized due
to its implication in prognosis and treatment. (B). Cavernous transformation of the portal vein. At-
rophy of the MPV (marked in red) secondary to chronic PVT causes hypertrophy in the vasa vaso-
rum of the portal vein with the apparition of porto-portal shunts.
Table 1. Classification of portal vein thrombosis according several authors.
Classification
Description
Categories
Strengths vs. Weaknesses
Yerdel et al.,
2000 [16]
Post-transplant survival
(i) Involvement of MPV,
SMV and SV
(ii) Degree of occlusion
Grade 1: <50% occlusion
Grade 2: >50% occlusion
Grade 3: Complete PV or SV
occlusion
Grade 4: Complete PV occlusion
with SMV extension
- Well-defined long-term post-
transplant survival
- Only for LT candidates
- Most widely used classification to
date
Figure 2.
Classification of portal vein thrombosis. (
A
) It is crucial to characterize the extension of the
portal vein thrombosis and its relationship with the main portal vein (MPV). The involvement of the
superior mesenteric vein (SMV) and splenic vein (SV) should be properly characterized due to its
implication in prognosis and treatment. (
B
) Cavernous transformation of the portal vein. Atrophy of
the MPV (marked in red) secondary to chronic PVT causes hypertrophy in the vasa vasorum of the
portal vein with the apparition of porto-portal shunts.
In line with the above, the initial description of PVT must detail the location and
extension of the thrombosis, specifying whether it involves the intrahepatic branches, main
portal vein, SV, and/or SMV. The percent of lumen occluded in each of these locations is
graded as completely occlusive (no persistent lumen), partially occlusive (clot obstructing
>50% of original vessel lumen), or minimally occlusive (clot
obstructing <50%
of original
vessel lumen). If present, cavernous transformation (i.e., gross porto-portal collaterals
without original portal vein seen) must be reported. This documentation is not only
important for therapeutic decisions and evaluating response to treatment but also for
establishing a correlation between the site of thrombosis and the clinical presentation of
PVT. Indeed, the involvement of SMV may cause intestinal ischemia and development of
ectopic varices, whereas SV thrombosis could lead to the appearance of fundic varices [
3
,
4
].
As far as the time course of thrombosis is concerned, PVT is classified as recent if PVT
is presumed to be present for less than 6 months and as chronic if PVT persists longer than
this time frame. The term recent is preferred over acute because the latter implies clinical
symptoms and PVT is often asymptomatic and incidentally diagnosed. That is why precise
dating is impossible in a significant proportion of patients. The 6-month threshold was
chosen based on data from a prospective study of 102 patients with recent non-cirrhotic PVT.
In this study, failure to recanalize within 6 months of PVT diagnosis led to development of
cavernous transformation in most patients despite continued anticoagulation [
20
]. Similar
results were observed in the setting of liver cirrhosis [
21
,
22
]. When chronic obstruction
persists and cavernous transformation occurs, the latter term is preferred. However,
cavernous transformation is not synonymous with chronic PVT since this finding can be
demonstrated within 1–3 weeks after PVT onset [23].
Finally, depending on the spontaneous course and/or response to treatment, PVT
is classified as progressive (thrombus increases in size or becomes completely occlusive),
stable (no changes in size nor occlusion) or regressive (thrombus decreases in size or degree
of occlusion) [3,4].
J. Clin. Med. 2022,11, 6435 5 of 26
4. Pathophysiology and Risk Factors
As with any other thrombus, the pathogenesis of PVT is generally multifactorial
and it is believed to be primarily determined by the interplay of the three physiological
factors of Virchow’s triad: slow blood flow, hypercoagulability, and endothelial damage.
Nevertheless, the exact contribution of each of these factors to PVT development was not
fully elucidated [24].
4.1. Reduced Blood Flow
A reduced portal flow velocity secondary to a “steal effect” produced by porto-
collateral circulation, combined with the increase in portal vein diameter commonly seen in
patients with clinically significant portal hypertension (CSPH) seems to be the most impor-
tant factor for PVT development in patients with cirrhosis. Specifically, a 15 cm/s threshold
was proposed to identify patients at higher risk of developing PVT. This was first reported
by Zocco et al. in a prospective study of 100 cirrhotic patients followed up for one year [
25
].
Since then, other retrospective and prospective studies have confirmed this finding [
26
28
].
However, a large satellite study of a prospective trial of ultrasound screening for hepa-
tocellular carcinoma (HCC) did not and raised concerns regarding the reproducibility of
portal vein blood flow velocity measurements, thus, making uncertain the generalization of
specific thresholds [
29
]. Further indirect data in this regard comes from studies identifying a
higher portal vein diameter and the presence of large porto-collateral vessels as risk factors
for developing PVT [
26
,
30
,
31
]. The efficacy of a transjugular intrahepatic portosystemic
shunt (TIPS) in restoring PVT patency by presumably increasing portal flow also supports
this major role of altered portal hemodynamics in PVT development [32].
As nonselective
β
-blockers (NSBB) may reduce portal venous flow by decreasing
cardiac output and by inducing splanchnic arterial vasoconstriction, it was suggested that
they might increase the risk of PVT [
33
,
34
]. This is of great importance since indications
of NSBB have recently widened, and they are now recommended not only for primary or
secondary prevention of variceal bleeding but also to prevent decompensation in patients
with compensated advanced chronic liver disease and CSPH [
4
]. A recent systemic review
and meta-analysis of mostly retrospective studies showed an increased risk of PVT in
subjects on NSSBs treatment (OR 4.62, 95% CI 2.50–8.53; p< 0.00001) [35]. However, there
was high heterogeneity in relation to sample size, definition of PVT and stage of liver
disease and most of the studies did not evaluate the potential role of confounding factors.
Indeed, in two large prospective studies, NSBB were not associated with an increased risk
of PVT after adjustment for variables related to the severity of portal hypertension [
28
,
29
].
Therefore, the use of NSBB should not be limited based on this concern, even more so
considering their larger benefits in patients with cirrhosis [24].
4.2. Alterations in Coagulation
Cirrhosis has long been perceived as an acquired bleeding disorder as a consequence
of thrombocytopenia and abnormal routine coagulation tests. However, unlike hereditary
coagulopathies, cirrhosis affects the whole spectrum of the coagulation cascade (i.e., both
procoagulant and anticoagulant factors) and is associated with both platelet hyperactiv-
ity and increased levels of von Willebrand factor, all of which results in a “rebalanced
hemostasis”. This new equilibrium is fragile and can easily be tipped towards either a
prohemorrhagic or a prothrombotic phenotype. Furthermore, it was suggested that this
potential prothrombotic state is intimately involved in the progression of liver disease and
could also be involved in PVT development [3,24,36,37].
Several studies have evaluated whether the hemostatic alterations associated with
cirrhosis increase the risk of PVT and were extensively reviewed elsewhere [
24
]. Traditional
coagulation tests (e.g., prothrombin time or partial thromboplastin time) do not adequately
reflect this new hemostatic balance since they do not take into account the inhibition of
thrombin by anticoagulant factors [
3
,
36
]. The ratio between FVIII and protein C has long
been suggested to reflect this increase in coagulation potential [
38
], although recent data
J. Clin. Med. 2022,11, 6435 6 of 26
have challenged this assumption showing that despite predicting the development of com-
plications of cirrhosis, it is unrelated to the coagulation status of patients with cirrhosis [
39
].
Regarding its role in PVT, contradictory results were published [
28
,
40
,
41
]. Similar findings
were observed with other cirrhosis-related hemostatic alterations, such as the ratio of fac-
tor II to protein C, levels of coagulation factors, thrombomodulin resistance, fibrinolysis
markers, plasminogen activator inhibitor-1 levels [
42
], or viscoelastic parameters [
24
]. Of
note, the aforementioned studies evaluated coagulation factors in systemic blood with
few studies evaluating whether the portal vein may represent a hypercoagulable vascular
bed. Although initial studies described the existence of a relative hypercoagulability in
this territory [
43
,
44
], a more recent study failed to replicate this finding in cirrhotic patients
who underwent TIPS placement [
45
]. Its hypothetical contribution in PVT development
was not tested as none of these studies evaluated patients with PVT.
There are fewer data on the role of platelet aggregation in PVT development. In
opposition to previous studies suggesting platelet dysfunction in patients with cirrhosis,
recent studies show that platelets are hyperfunctional in these patients, particularly in the
decompensated stage and in the portal vein [
46
48
]. Moreover, this increased platelet aggre-
gatory potential was associated with a higher risk of further decompensation, death, and
PVT [
47
,
49
]. In line with these findings, a recent study showed that the ADAMTS-13/von
Willebrand factor ratio was predictive of PVT [
50
]. These results establish a rational basis
for evaluating the use of antiplatelet agents to prevent PVT and halt disease progression.
Other factors that may induce blood hypercoagulability in patients with cirrhosis are
systemic inflammation, a well-recognized feature of decompensated cirrhosis [
51
], and
HCC [
52
]. The former is scarcely studied with contradictory results [
24
,
28
]. A recent
paper observed that serum albumin was inversely associated with PVT and suggested
albumin as a modulator of the hemostatic system by reducing platelet activation through its
inhibitory effects on oxidative stress. According to the authors, these findings established
a rationale for randomized interventional studies to investigate the beneficial effects of
albumin to prevent PVT in cirrhosis [
53
]. No information in this matter was provided in
the long-term albumin administration trials [
54
,
55
]. As far as HCC is concerned, there
is growing evidence suggesting that it is associated with pro-thrombotic alterations (i.e.,
increased platelet activation and function, enhanced thrombin generation, hypo-fibrinolysis,
and elevated levels of prothrombotic microvesicles) that may synergistically contribute to
hypercoagulability and thrombosis [52].
Regarding the role of inherited and other acquired prothrombotic disorders in PVT
development, current data are conflicting. The limited number of studies available are
mostly case-control studies with small sample sizes. Their study design, target popu-
lation (diverse ethnicities and geographical locations), diagnostic criteria for PVT, and
assessment of thrombophilic conditions vary widely and contribute to the inconsistent
results [21,29,5677].
Moreover, none of these studies have properly evaluated whether
the presence of thrombophilia impacts the progression rate or response to treatment [
77
].
Among the different thrombophilic genetic defects, Factor V Leiden and prothrombin
G20210A mutations are the most frequently studied. Three meta-analyses concluded that
they increased the risk of PVT in patients with cirrhosis [
78
80
], although in one of them,
this association was not shown for the prothrombin mutation [
79
], and all of them were
biased by the quality of the studies included. Inherited protein C, protein S, or antithrombin
III deficiencies are difficult to detect due to co-existent liver synthetic dysfunction [
36
].
Their levels, however, do not seem to be associated with PVT development [
81
]. The
methylene tetrahydrofolate reductase C677T and plasminogen activator inhibitor—type 1
4G–4G mutations were also described as independent predictors of PVT [
63
,
82
], although
these polymorphisms are not conclusively associated with increased thrombotic risk [
82
].
The role of other acquired prothrombotic disorders was less evaluated in patients with
liver cirrhosis and PVT. In contrast to non-cirrhotic PVT, the relevance of myeloprolifer-
ative disorders and antiphospholipid syndrome is, so far, inconclusive [
83
]. Due to the
conflicting data, current guidelines make no strong recommendations regarding testing for
J. Clin. Med. 2022,11, 6435 7 of 26
these conditions in either a screening capacity before PVT diagnosis or confirmatory once
thrombosis has developed [14,10].
In summary, there is no solid evidence that hypercoagulability due to cirrhosis-related
hemostatic alterations or to inherited and other acquired prothrombotic disorders plays a
major role in the pathophysiology of PVT.
4.3. Endothelial Damage
Of the three components of Virchow’s triad, the hypothetical role of endothelial
dysfunction in PVT generation is the least studied, partially due to the inaccessibility of the
splanchnic territory (for review see [
24
]). Therefore, more studies are needed that compare
endothelial-specific markers in blood from the portal area between cirrhotic patients with
or without PVT.
Endothelial injury due to sclerotherapy, previous abdominal surgery, splenectomy,
and portosystemic shunt surgery were also identified as risk factors for PVT, although
altered portal venous blood flow due to some of these procedures also promotes thrombus
formation [84].
5. Epidemiology
PVT constitutes the most common thrombotic event in the setting of cirrhosis. Defining
its incidence and prevalence is difficult due to the heterogeneity of studies regarding the
population included (LT candidates are the most studied), the definition of PVT, and the
tests used for its diagnosis [
3
]. The few prospective studies have reported incidence rates in
the range from 1.6% to 4.6% at 1 year [
28
,
29
,
85
,
86
]. A recent meta-analysis showed 1-year
and 3-year cumulative incidences of 4.8% and 9.3%, respectively [
87
]. This incidence varies
among compensated and decompensated patients. In the latter meta-analysis, the pool
incidence of PVT was 9.9% in Child–Pugh class A and 18.3% in Child–Pugh class B–C.
Higher rates were described in the presence of HCC (up to 40%) [
84
,
88
]. Incidence rates
also vary by disease etiology with evidence that PVT is more frequently associated with
nonalcoholic fatty liver disease [89].
6. Clinical Manifestations and Prognostic Impact
Diagnosis of PVT is most often asymptomatic and commonly discovered by routine
imaging tests [
1
]. Symptoms ascribed to PVT are non-specific and include nausea, vomiting,
mild abdominal pain, diarrhea, and loss of appetite. Rarely, mesenteric ischemia due
to the extension of PVT to the SMV can occur [
2
]. Patients with more advanced liver
cirrhosis are more protected than non-cirrhotic patients from this complication due to the
decompression achieved through the frequent presence of porto-systemic collaterals [
3
]. It
is, thus, important to establish a correlation between PVT features (time course, degree of
occlusion, and stage of liver disease) and the clinical presentation [
84
]. For example, in the
setting of acute abdominal pain, the finding of a partially occlusive PVT on ultrasound (US)
should not defer the performance of a contrast-enhanced computed tomography (CT) or
magnetic resonance (MR) scan to rule out other causes of abdominal pain (e.g., pileflebitis
or malignant infiltration into the portal vein) and to better characterize the true extension
of PVT.
In other instances, PVT is diagnosed in coincidence with a liver decompensation,
and again, the temporal relationship should not be directly interpreted as evidence of
causality. Indeed, the impact of PVT on the natural history and prognosis of cirrhosis is
controversial, and whether PVT is merely a manifestation of progressive disease, or an
actual cause of disease progression remains to be elucidated [
3
]. Discrepancies among
studies regarding patient selection criteria (compensated vs. decompensated), degree and
extent of thrombosis (occlusive vs. nonocclusive), treatment strategies (anticoagulation vs.
no anticoagulation), sample size, and time of follow-up have led to conflicting data [
90
].
Hence, several prospective [
29
,
85
,
91
,
92
] and retrospective studies [
30
,
93
] have shown that
PVT is not responsible for disease progression or increased mortality, whereas a randomized
J. Clin. Med. 2022,11, 6435 8 of 26
study by Villa et al. indirectly suggested the opposite. In this small, controlled trial, a
12-month
course of 4000 IU/day enoxaparin in patients with Child B–C (7–10 points)
cirrhosis not only prevented PVT but also improved survival and decompensation [
94
].
Moreover, PVT was shown to be independently associated with a higher risk of variceal
bleeding and failure of endoscopic control of bleeding and rebleeding [9597].
In LT recipients, the impact of PVT on survival after LT seems to depend on the size
and extent of PVT at the time of surgery [
98
]. Two large transplant database analyses
showed PVT as a strong independent predictive variable of posttransplant survival but
did not stratify this risk according to the grade of PVT [
99
,
100
]. Other studies [
16
,
101
,
102
]
and a meta-analysis [
103
] have shown that only a completely occlusive PVT increases
post-transplant mortality. The threshold of PVT extension at which outcomes are worse
is unknown but is probably related to the need for non-anatomical PV reconstructions
(renoportal anastomosis, cavoportal hemitransposition, or portal vein arterialization). These
cases have worse outcomes by adding technical difficulties and increasing graft ischemic
times [
19
]. Unfortunately, there are no randomized controlled trials showing that PVT
therapy improves post-transplant survival. Due to these discrepant results and lack of
randomized controlled trials, guidelines differ on their recommendations. Hence, the
AASLD guideline on vascular disorders states that there are insufficient data to recommend
pretransplant treatment of PVT with the goal of improving posttransplant outcomes [
3
],
while the Baveno VII consensus recommends anticoagulation in potential LT candidates
independently of the degree of occlusion and extension with the goal of preventing re-
thrombosis or progression of thrombosis to facilitate adequate portal anastomosis in LT
and reduce post-transplant morbidity and mortality [
4
]. Similarly, some guidelines only
recommend screening for PVT in potential LT candidates at the time of screening for HCC
(1, 4, 10), while others do not make any specific recommendations [3].
7. Natural History
PVT is a heterogeneous condition also with respect to its natural history, and this
heterogeneity makes PVT a unique entity among venous thromboses [
24
]. On the one hand,
the spontaneous recanalization of PVT is extensively described [
21
,
29
,
30
,
92
,
93
,
102
,
104
].
A recent meta-analysis comparing anticoagulated vs. non-anticoagulated patients with
cirrhotic PVT showed a rate of spontaneous portal vein recanalization of 42% [
105
]. The
probability of this event is higher in compensated cirrhosis or with partial PVT (up to
70%) [
29
] and much lower in patients with decompensated cirrhosis and those listed
for LT [
21
,
98
,
102
,
106
]. The different imaging techniques used to stage the extent of the
thrombus also contribute to the heterogeneous rates of spontaneous recanalization or
of progression reported. In addition to these factors, no other predictors of spontaneous
improvement or progression are known, which makes it difficult to evaluate the real efficacy
of the different treatments used for PVT. It must be noted that although spontaneous
resolution of PVT may occur, especially if non-occlusive and in compensated cirrhosis, PVT
progression occurs in 33% of untreated patients [105].
On the other hand, the rates of portal vein recanalization after anticoagulation are
substantially lower than in other thromboses, especially in an aged thrombus [
24
]. Driever
et al. recently proposed an explanation for this finding [
107
]. In their study, they described
the composition and structure of nonmalignant cirrhotic PVT that were collected during
LT in 79 patients. They observed that all PVT consisted of tunica intima thickening of the
portal vein vessel wall in an appearance resembling intimal fibrosis, with only one-third of
the thrombi containing an additional fibrin-rich thrombus. Based on these findings, they
suggested changing the name PVT to portal vein stenosis and that the absence of fibrin in
most patients may explain the low rates of recanalization with anticoagulant therapy.
8. Diagnosis
The initial diagnosis of PVT is often made with Doppler US and this diagnostic
technique is the screening method of choice for PVT [
2
,
4
]. Doppler US may demonstrate
J. Clin. Med. 2022,11, 6435 9 of 26
hyperechoic material within the vessel lumen, dilatation of the portal vein, and diminished
portal venous flow [
108
110
]. US has a sensitivity ranging from 73–93%, specificity of 99%,
and positive predictive value of 87–96% compared with angiogram [
16
] and CT scan [
108
].
Advantages of US Doppler over other diagnostic tests include lower cost, wider availability,
and lack of radiation exposure. Despite being an excellent initial screening test, US Doppler
is an operator-dependent exploration and has lower reliability in the presence of bowel
gas, obesity, partially occlusive PVT, and in delimiting the extension of the thrombi to SV
and SMV. Furthermore, it may be difficult to differentiate bland thrombi from malignant
portal vein invasion. For all these reasons, current guidelines recommend performing a
contrast-enhanced imaging study following PVT diagnosis with US [
1
4
,
10
]. Both CT and
MR scans are excellent techniques for diagnosing PVT and portal cavernoma [
108
]. In
comparison to CT, MR scan has the advantages of less radiation and a better safety profile,
but it is limited by motion and flow artifacts, lower availability, higher cost, and technical
difficulties in patients with implanted metallic devices or surgical clips [
2
]. Another less
studied technique in this setting is endoscopic US. In a small cohort of patients with
and without cirrhosis it had a sensitivity of 81% and a specificity of 93% for diagnosing
PVT [
111
]. However, given its moderately invasive nature and inability to definitively
assess for HCC or mesenteric infarction, it is not routinely recommended in the diagnostic
algorithm of PVT.
The exclusion of tumoral invasion of the portal vein is essential to both decisions
regarding further management of HCC and determination of LT candidacy [
84
]. It may
occur in up to 12% to 20% of the patients with HCC [
112
,
113
]. Findings that support this
malignant invasion include enlarged portal vein diameter, enhancement of the throm-
bus in the arterial phase of contrast injection, neovascularity, distance from tumor to
thrombus lower than 2 cm, and tumor size > 5 cm [
114
,
115
]. The recently proposed A-
VENA criteria incorporates all these findings, except for tumor size, and also includes
alfa-fetoprotein > 1000 ng/dL.
In patients with
3 of these criteria, the diagnosis of ma-
lignant invasion could be accurately made (100% sensibility, 94% specificity, 80% positive
predictive value and 100% negative predictive value) [116].
In regard to the “age” of PVT, features of recent PVT include hypoechogenic and
hypodense thrombus, increased attenuation in the portal vein on an unenhanced CT scan,
and increased attenuation in the portal vein and a central lucency on a contrast-enhanced
CT scan. Associated hepatic perfusion changes can also be seen in the form of increased
hepatic parenchymal enhancement in the arterial phase and reduced enhancement during
the portal phase. In contrast, calcification within the wall of a thrombus and presence of
cavernoma suggest chronicity [
2
,
84
] (Figure 3). It must be pointed out that the former can
only be detected using US or CT but not MR and that cavernous transformation can occur
within 1–3 weeks after PVT onset [23] (Figure 4).
J. Clin. Med. 2022,11, 6435 10 of 26
J. Clin. Med. 2022, 11, x FOR PEER REVIEW 10 of 26
Figure 3. Different presentations of portal vein thrombosis through contrast-enhanced CT scan. (A)
Axial plane showing a partially occlusive portal vein thrombosis of the main portal trunk without
collaterals or other indirect signs of portal hypertension. (B) Apparition of collateral circulation with
umbilical vein repermeabilization and enlarged spleen in the context of portal vein thrombosis. (C)
Oblique plane showing a partially occlusive portal vein thrombosis of the main portal trunk with
hepatic perfusion defects on the lower segments of the right lobe. (D) Chronic portal vein throm-
bosis in patient with cirrhosis. Note the calcification of the portal vein as an indirect sign of chronic-
ity. Atrophic liver with splenomegaly, collaterals, and ascites are found in the context of cirrhosis.
Figure 4. Cavernous transformation of the portal vein. (A) Axial plane showing the substitution of
the main portal vein trunk by porto-portal collaterals originated from the vasa vasorum of the portal
vein. (B) Coronal plane of the portal vein and its cavernous transformation with the entanglement
of the new vessels composing the porto-portal collaterals.
Figure 3.
Different presentations of portal vein thrombosis through contrast-enhanced CT scan.
(A) Axial
plane showing a partially occlusive portal vein thrombosis of the main portal trunk without
collaterals or other indirect signs of portal hypertension. (
B
) Apparition of collateral circulation
with umbilical vein repermeabilization and enlarged spleen in the context of portal vein thrombosis.
(C) Oblique
plane showing a partially occlusive portal vein thrombosis of the main portal trunk with
hepatic perfusion defects on the lower segments of the right lobe. (
D
) Chronic portal vein thrombosis
in patient with cirrhosis. Note the calcification of the portal vein as an indirect sign of chronicity.
Atrophic liver with splenomegaly, collaterals, and ascites are found in the context of cirrhosis.
Figure 4.
Cavernous transformation of the portal vein. (
A
) Axial plane showing the substitution of
the main portal vein trunk by porto-portal collaterals originated from the vasa vasorum of the portal
vein. (
B
) Coronal plane of the portal vein and its cavernous transformation with the entanglement of
the new vessels composing the porto-portal collaterals.
J. Clin. Med. 2022,11, 6435 11 of 26
9. Prophylaxis
There is only one trial assessing the use of anticoagulation for preventing PVT. In
this study by Villa et al., 70 patients with cirrhosis (Child B7-C10) were randomized to
receive enoxaparin at prophylactic doses (4000 IU/day for 48 weeks) vs. no treatment.
Patients receiving enoxaparin had lower incidence of PVT and a sustained decrease in
decompensation events that exceeded the expected from the reduction in PVT. The au-
thors hinted at an enoxaparin-induced improvement of intestinal barrier function and
reduced bacterial translocation as a potential mechanism [
94
]. It was also postulated that
these beneficial effects could also be related to the antifibrotic effects of anticoagulation.
Indeed, increasing evidence suggests that the potential prothrombotic state associated
with cirrhosis leads to liver fibrosis development and progression in liver disease pre-
sumably by generating thrombi in the hepatic microcirculation that cause parenchymal
extinction and by activating hepatic stellate cells through thrombin and Factor Xa via
protease-activated receptors [
37
,
117
]. Results from a confirmatory study using ribaroxaban
instead of enoxaparin are eagerly awaited (ClinicalTrials.gov Identifier: NCT02643212).
10. Treatment
The decision to treat a PVT in the setting of cirrhosis is determined by the age and
extent of thrombus, the presence of symptoms, and the patient’s transplant status [
84
].
In patients with concern for intestinal ischemia, early initiation of anticoagulation and
immediate consultation with surgery, critical care, interventional radiology, and hematology
is advised [
3
]. In the patient without ischemic symptoms, the weak existing evidence on
the beneficial effects of therapy and the uncertain impact of PVT on the natural history of
cirrhosis explain the heterogenous recommendations of current guidelines. In this scenario,
the aim of treatment is to prevent clot extension that could potentially lead to progression
of portal hypertension, hinder a future LT, or preclude a conventional end-to-end portal
vein anastomosis, thus, reducing post-transplant morbidity and mortality [
3
,
4
]. Overall,
and especially in non-LT candidates, treatment should be considered on a case-by-case
basis. Table 2shows the recommendations of the different guidelines regarding treatment
indications and other issues.
10.1. No Treatment
A conservative approach is generally considered in asymptomatic non-LT candidates
in whom the uncertainties of the survival benefit of anticoagulation are greater. In this
scenario, recent guidelines suggest a conservative approach in thrombosis of small in-
trahepatic sub-branches of the portal vein or minimally occlusive (<50% obstruction of
the lumen) PVT. In case of progression at serial imaging, anticoagulation should then be
considered [3,4].
The latter strategy can be also applied to patients with chronic complete occlusion
of PVT or cavernous transformation of the portal vein with established collaterals in
whom there is no established benefit for anticoagulant therapy [
3
]. The main objective of
anticoagulation in these patients is to prevent recurrent thrombosis and, to a lesser extent,
recanalization. The decision-making process in this setting should be based on bleeding
risk, SMV involvement, presence of a thrombophilic condition, LT candidacy, and patient
preferences. Thus, the guideline on disorders of the hepatic and mesenteric circulation of
the American College of Gastroenterology suggests anticoagulation in patients with chronic
PVT only if there is (i) evidence of inherited thrombophilia, (ii) progression of thrombus,
(iii) history of bowel ischemia due to thrombus extension into the mesenteric veins, or (iv)
PVT in a patient awaiting LT [2].
J. Clin. Med. 2022,11, 6435 12 of 26
Table 2. Current guidelines recommendations for management of portal vein thrombosis in cirrhosis.
Baveno VII 2022 AASLD 2021 ACG 2020 ICLDC 2018 EASL 2015
Screening
for PVT
Listed or potential candidates for
LT at the time of HCC screening -
(i) new diagnosis of cirrhosis
(ii) onset of PH
(iii) decompensation.
US every 6 months in
(i) patients with cirrhosis and PH, or
(ii) Listed or potential candidates for LT
Listed or potential
candidates for LT
Imaging
test
- Screening: US
- Diagnosis: CT scan or MRI - Diagnosis: CT scan or MRI
- Screening: US
- Diagnosis: CT scan or
MRI
- Screening: US
- Diagnosis: CT scan or MRI
- Diagnosis: CT scan or
MRI
Screening
for throm-
bophilia
-(i) family history of MPN
(ii) suggestive laboratory findings
(i) previous thrombosis
(ii) thrombosis at unusual sites
(iii) family history
“Consider on an individual basis” “Consider screening”
Indications
for
treatment
- Recommended:
(i) recent (<6 m) PVT completely
or partially occlusive of the PV
trunk
(ii) symptomatic PVT
(iii) potential candidates for LT
- Considered:
(iv) minimally occlusive that
progresses (v) compromise of
SMV
(i) Recent (<6 m) completely or
partially occlusive of the main PV
or SMV
(ii) Ischemic symptoms
(i) evidence of thrombophilia,
(ii) progression into the
mesenteric veins, or
(iii) current or previous
evidence of bowel ischemia
(i) LT candidates with occlusive main
PVT with or without extension to SMV
(ii) Yerdel Grade 2 PVT considered on
an individual basis
(i) SMV thrombosis, with a
past history suggestive of
intestinal ischemia or
(ii) liver transplant
candidates
Medical
therapies
Initial agent: preferably LWMH
Maintenance:
- LWMH, VKAs, DOACs LWMH, VKAs or DOACs
Initial agent: UH or LWMH
Maintenance:
- LWMH, VKAs - -
TIPS
(i) PVT of the main PV without
recanalization on AC, especially
in patients listed for LT
(i) PVT that hinders a
physiological anastomosis
between the graft and recipient
(ii) refractary PH complications
-
(i) acute and chronic PVT in patients
with cirrhosis requiring treatment for
significant PH
(i) LT candidates not
responding to AC
Abbreviations: AASLD: American Association for the Study of Liver Diseases; AC: anticoagulant; ACG: American College of Gastroenterology; DOACs: direct oral anticoagulants;
EASL: European Association for the Study of the Liver; HCC: hepatocellular carcinoma; ICLDC: International Coagulation in Liver Diseases Conference; LT: liver transplant; LWMH: low
weight molecular heparin; MPN: myeloproliferative neoplasm; MRI: magnetic resonance imaging; SMV: superior mesenteric vein; SV: splenic vein; TIPS: transjugular intrahepatic
portosystemic shunt; PH: portal hypertension; PV: portal vein; PVT: portal vein thrombosis; UH: unfractionated heparin; US: ultrasound; VKAs: vitamin K antagonists.
J. Clin. Med. 2022,11, 6435 13 of 26
10.2. Anticoagulation
Anticoagulation is the mainstay of PVT treatment. The majority of studies assessing the
safety and effectiveness of anticoagulants in patients with cirrhosis are small retrospective
studies that differ in inclusion criteria, magnitude of PVT (predominancy of partially occlusive
PVT), timing and type of anticoagulation, and in the assessment of outcomes. Moreover, anti-
coagulation decisions were based on provider’s judgment and were not based on predefined
protocols [
2
]. With these limitations, data from these
studies [21,22,68,73,77,102,104,118126]
and from aggregate-data meta-analyses [
105
,
127
129
] have shown that anticoagulation is
safe and effective in achieving portal vein recanalization, but its effect on survival is
still uncertain.
In regard to effectiveness, the latter meta-analyses have shown rates of recanaliza-
tion ranging between 66.6% and 71.5% for any degree of recanalization and between
40.8% and 53% for complete portal vein recanalization. In contrast, rates of thrombus
progression despite anticoagulation range between 5.7 and 9%. Early administration of
anticoagulation (<14 days in one study [
119
] or <6 months in others [
21
,
120
,
130
]) is of
utmost importance for treatment success with the mean time of recanalization ranging
from 5.5 to 8 months. However, delayed responses even after 1 year of treatment were
reported [
68
]. Other factors that are associated with a good response are less advanced
disease, less extensive thrombosis [
119
,
126
], degree of SMV occlusion less than 50%, lower
platelet count, absence of previous PHT-related bleeding, and lower spleen thickness at
baseline [
129
,
131
]. Importantly, when anticoagulation is stopped, PVT relapses in 30–40%
of the patients [
118
,
119
,
132
]. The time elapsed between discontinuation of anticoagulation
therapy and recurrence of thrombosis ranges between 2 and 5 months [119,132]. All these
findings explain the recommendations from current guidelines to anticoagulate for at
least 6 months and to maintain anticoagulation until LT (Table 2). The lack of evidence
to recommend a specific timing or type of imaging study regardless of the decision to
initiate anticoagulation or not is also of note. The AASLD suggests serial imaging every
2 to 3 months
to assess treatment response and every 3 months if a conservative approach
is chosen, but anticoagulation is considered in case of PVT progression.
The former recanalization rates in patients treated with anticoagulants are significantly
higher than those seen in untreated patients (25–42% for overall recanalization with odds
ratio ranging between 2.61 and 4.8 and 33% for complete portal vein recanalization with
odds ratio ranging between 2.14 and 3.4). Similarly, PVT progression is more likely in
untreated patients (33% with odds ratio ranging between 0.06 and 0.26) [
105
,
127
129
].
The impact of these higher rates of recanalization under anticoagulation therapy is still
uncertain, with two meta-analyses showing a decreased risk of variceal bleeding [
105
,
128
]
and another an improvement in overall survival [
129
]. An unpublished individual patient
data meta-analysis also showed an increased overall survival and found that the beneficial
effect of anticoagulation largely depended on portal vein recanalization [133].
Whatever the impact, anticoagulant treatment was shown to be safe, with similar
major and minor bleedings rates when compared to untreated patients (10.3–11%) [
105
,
129
].
Moreover, anticoagulation was not associated with an increase in 5-day treatment failure
or 6-week mortality in patients with cirrhosis having an episode of upper gastrointestinal
bleeding [
134
]. A platelet count below 50
×
109/L was identified as a risk factor for
bleeding from any site in patients with cirrhosis and PVT receiving anticoagulation [
119
].
These patients are also at higher risk of PVT, and consequently, management should be
assessed on a case-by-case basis [4].
Current options for anticoagulation include vitamin K antagonists (VKAs), unfrac-
tionated heparin, low-molecular-weight heparin (LMWH), fondaparinux, and direct oral
anticoagulants (DOACs). Table 3shows the main characteristics of each therapeutic option.
Most of the published studies have used VKAs and LMWH, and both seem to have similar
effectiveness [105].
J. Clin. Med. 2022,11, 6435 14 of 26
Table 3. Characteristics of the different anticoagulation therapies for the treatment of portal vein thrombosis.
Unfractionated Heparin Low-Weight Molecular Heparin Vitamin-K Antagonists Direct Oral Anticoagulants
Administration Endovenous Subcutaneous Oral Oral
Posology Daily infusion qd/bid Qd Apixaban/dabigatran: bid
Edoxaban: qd
Rivaroxaban: bd for 3 weeks, qd thereafter
Half-life Minutes to 1–2 h 4–12 h 10–24 h 6–18 h
Absorption and
bioavailability Caution if hypoalbuminemia BA 85–95%, caution if hypoalbuminemia Affected from bowel edema in PH
and diet Affected from bowel edema in PH
Monitoring antiXa factor or aPTT Not needed, but
caution if GFR < 15 mL/min/m2, obesity, and
female sex PT and INR (2–3) Not needed, especially if GFR > 15 mL/min/m2,
non-obese, and male sex
Renal function Dose change not necessary but monitor
with antiXa and aPTT
Contraindicated if severe renal failure or
dialysis, caution in mild/moderate renal
failure
Contraindicated if severe renal
failure or dialysis, caution in
mild/moderate renal failure
- No dose change needed
- Not indicated if GFR < 15 mL/min/m2
Side Effects
- Hemorrhage/hematoma
- Heparin-induced thrombopenia
(+++)
- Hyperkalemia
- Hemorrhage/hematoma
- Heparin-induced thrombopenia (++)
- Altered LFT Hemorrhage/hematoma - Hemorrhage/hematoma
- Altered LFT
Bleeding risk ++ ++ ++ Apixaban: +
Dabigatran/Edoxaban: ++
Rivaroxaban: +++
Antidote Protamine sulfate Protamine sulfate * Vitamin-K AntiX: andexanet alpha **
Dabigatran: idarucizumab **
Pros in cirrhosis - Allowed in renal failure
- Short half-life
- Subcutaneous administration
- Easy interruption before invasive
procedures Oral administration
- Oral administration
- Predictable effect
- Fewer interactions than VKAs
Cons in cirrhosis Not suitable for maintainance therapy
Fluctuating levels of ATIII
- Renal function lability
- Not well monitored through antiXa - INR not reliable in cirrhosis Not recommended in Child C
Caution in Child B
Higher costs
Abbreviations; AC: anticoagulation; antiX: apixaban, edoxaban, and rivaroxaban; aPPT: activated-partial thromboplastin clotting time; ATIII: antithrombin III; BA: bioavailability;
DOACs: direct oral anticoagulants; GFR: glomerular filtration rate; LFT: liver function test; LWMH: low-weight molecular heparin; PH: portal hypertension; PT: prothrombin time; INR:
international normalized ratio; UH: unfractionated heparin; VKAs: vitamin-K antagonists. * Less effective in LWMH than unfractionated heparin. ** Available only in selected centers.
J. Clin. Med. 2022,11, 6435 15 of 26
10.2.1. Unfractionated Heparin
Treatment of PVT with unfractionated heparin may be used as the initial agent in
the presence of renal insufficiency and/or in patients with concern for intestinal ischemia
due to the possibility of rapid reversal of anticoagulation [
2
]. However, its intravenous
administration precludes its use as a long-term treatment.
10.2.2. Low Molecular Weight Heparin (LMWH)
LMWH is traditionally considered the initial treatment of choice for PVT. Due to its
parenteral administration, patient compliance and quality of life can be compromised.
Therefore, it is generally used as a “bridge” therapy in patients that will ultimately receive
VKAs or DOACs. The duration of this “bridge” therapy varies greatly among centers, with
some initiating LMWH and VKAs simultaneously (with discontinuation of LMWH once
the INR reaches therapeutic range of 2–3) and others (similar to our center) postponing
oral anticoagulation for a month. However, in some specific patients, LMWH may be
more suitable than VKAs, such as in patients with refractory ascites requiring periodic
paracentesis or patients with prolonged INR.
In terms of dose, a small study showed that enoxaparin 1 mg/kg twice daily had
similar efficacy with fewer complications than 1.5 mg/kg daily [
121
]. Whether a reduction
in dose is necessary in patients with high bleeding risk (e.g., severe thrombocytopenia) is
unknown, with one small study showing that a reduced dose was not associated with a
decrease in efficacy [
130
]. Measuring anti–Factor Xa activity to monitor the anticoagulant
effect of LMWH may lead to therapeutic overdose due to the low levels of antithrombin III
(the substrate to which LMWH binds) in patients with advanced disease. Thus, anti-Xa
levels were found to be significantly lower in patients with cirrhosis despite an adequate
anticoagulation effect [
135
,
136
]. Despite this limitation, we still use anti-Xa levels to help
us adjust the dose in patients with bleeding complications on LMWH therapy, more so in
the presence of renal dysfunction and morbid obesity.
10.2.3. Fondaparinux
Fondaparinux constitutes a molecule that inhibits activated factor X through selective
high-affinity binding to antithrombin III [
137
]. Unlike heparin, fondaparinux does not
inhibit thrombin directly or platelet factor IV. For this reason, risk of heparin-induced throm-
bocytopenia is much lower [
138
]. Its once-daily administration makes it more convenient
than most LMWH.
Regarding its efficacy and safety, fondaparinux was compared to LMWH in one
retrospective study including 124 patients with cirrhosis and PVT. Fondaparinux showed
higher probability of resolution of PVT at 36 months (77% vs. 51%; p= 0.001) but also
higher bleeding rates (27% vs. 13%; p= 0.06) [
139
]. Further prospective studies are required
before making a formal recommendation.
10.2.4. Vitamin K Antagonists
The use of VKAs with the aforementioned “bridge” strategy was shown to be safe
and effective in both waitlist and non-waitlist cohorts [
102
,
126
]. Recanalization rates
and adverse effects seem to be similar when compared to LMWH [
104
,
120
]. The main
limitations of VKA are their narrow therapeutic window and the risk of underdosing due
to the baseline elevation of INR observed in patients with advanced cirrhosis.
10.2.5. Direct Oral Anticoagulants
DOACs, including direct thrombin inhibitor (dabigatran) and factor Xa inhibitors
(rivaroxaban, apixaban, and edoxaban) were recently added to the PVT therapeutic ar-
mamentarium. Their main advantages are their oral administration in fixed doses and
poor interaction with other drugs. Current available data suggest that there are no major
safety concerns regarding the use of DOACs in patients with Child–Pugh A and that
they should be used with caution in patients with Child–Pugh B (some authors do not
J. Clin. Med. 2022,11, 6435 16 of 26
recommend rivaroxaban in these patients because of increased plasma concentrations
and pharmacodynamic effects [
140
]). In patients with Child–Pugh C, DOACs are not
recommended [4]
In regard to their pharmacokinetics in cirrhosis, very little is known.
In vitro
studies
using plasma of decompensated cirrhosis have shown differences in anticoagulation po-
tency when measured by thrombin generation assays [
141
,
142
]. Defects in various steps
of drug metabolism such as plasma protein binding, cytochrome p450 function, biliary
excretion, and renal clearance due to the underlying cirrhosis may partially explain these
results, although larger in vivo studies are needed [3,84].
Several retrospective studies [
143
147
] and meta-analyses [
148
,
149
] have observed a
similar or better safety profile in terms of bleeding complications of DOACs compared to
VKAs in cirrhotic patients affected by atrial fibrillation, venous thromboembolism, or PVT.
Bleeding definitions differed among these studies, and to control for this bias, Nisly et al.
conducted a systematic review and meta-analysis considering only studies in which the
primary safety outcome was major bleeding according to the definition of the International
Society on Thrombosis and Haemostasis. They found no major differences in this regard
between DOACS and traditional anticoagulants [
148
]. It must be noted, however, that
the bleeding risk increased in patients with advanced cirrhosis. In a retrospective study,
Semmler et al. observed a significant association of spontaneous bleedings with liver
disease severity [147].
There is even less evidence in relation to the effectiveness of DOACs in patients with
cirrhosis and PVT. In the only randomized, controlled trial, Hanafy et al. showed that
rivaroxaban was more effective than warfarin in terms of recanalization rates, recurrence
of PVT, and safety in patients with HCV-related cirrhosis and PVT [
150
]. Similar findings
were found in a retrospective study comparing edoxaban with warfarin [
151
] and in a
prospective study comparing ribaroxaban and dabigatran with no treatment [152].
Due to this increasing evidence, their much easier administration, and the existence
of reversal agents (idarucizumab for dabigatran and andexanet alfa for rivaroxaban and
apixaban), these agents are increasingly utilized. Current barriers, beyond safety issues in
patients with advanced liver disease, are their high cost and limited availability.
10.3. Transjugular Intrahepatic Porto-Systemic Shunt
PVT was previously considered a contraindication to TIPS creation. However, the
introduction of new interventional radiological techniques has improved the rates of
technical success up to 86.7–95%. It must be highlighted that most of the available data
come from small, non-controlled, and retrospective cohorts that differ in inclusion criteria
and treatment applied. Moreover, complications of portal hypertension refractory to
conventional therapy, and not PVT itself, were the most common indications for TIPS
placement in patients with cirrhosis and PVT [153155].
Two meta-analyses have shown recanalization rates after TIPS of 81–84.4% and of 73%
for complete recanalization. The rates of major complications were 10% [
156
,
157
]. When
compared to anticoagulation, TIPS has a higher effectiveness in terms of portal vein recanal-
ization [
158
,
159
]. Regarding portal-hypertension-related complications, two randomized
controlled trials have evaluated the effectiveness of TIPS in comparison to standard therapy
(endoscopic band ligation + propranolol + anticoagulation) for the prevention of variceal
rebleeding in cirrhotic patients with PVT. Both studies showed that TIPS was more effective
in preventing rebleeding and in recanalizing PVT without increasing the risk of hepatic
encephalopathy and adverse effects. These beneficial effects, however, did not translate into
improved survival [
160
,
161
]. Both of these trials used anticoagulation after TIPS placement,
although indications of anticoagulation after TIPS placement are not clear. In fact, current
evidence does not support its use for further improving recanalization rates, although it
might be considered in cases of PVT extension to the SMV [32,156,158,162].
Patients with portal cavernoma or with no identifiable intrahepatic portal trunk or
branches are the most challenging. For these patients, a modified transplenic or transhepatic
J. Clin. Med. 2022,11, 6435 17 of 26
approach, known as portal vein recanalization-TIPS, was shown to improve technical
success to over 90% in the most recent series (range 75–100%) [
163
166
]. In these procedures,
portal vein recanalization is performed by angioplasty/stenting with subsequent TIPS
insertion to ensure the outflow of the system. TIPS failure may be due to lack of a landing
zone at the distal end of the portal vein or at the spleno-mesenteric confluence [167].
In line with the above, current guidelines recommend considering TIPS for the follow-
ing indications in patients with cirrhosis and PVT: (1) inadequate response to or contraindi-
cation of anticoagulation; (2) chronic PVT/portal cavernoma with portal-hypertension-
related complications refractory to medical treatment; and (3) chronic PVT that hinders a
physiological anastomosis between the graft and recipient portal vein [
1
4
,
168
]. Figure 5
shows the therapeutic algorithm according to the latest Baveno VII consensus [4].
J. Clin. Med. 2022, 11, x FOR PEER REVIEW 17 of 26
Figure 5. The algorithm for the management of portal vein thrombosis in patients with cirrhosis
according to the Baveno VII consensus recommendations.
10.4. Thrombolysis
Local or systemic thrombolysis was proposed as an adjunct to anticoagulation for the
treatment of recent PVT. Small case series have reported similar efficacy rates to those
achieved with anticoagulation alone but with a high risk of procedure-related morbidity
and mortality [169171]. Based on these preliminary observations, current guidelines only
recommend thrombolitic therapy in specialized centers for very selected patients in whom
intestinal ischemia persists despite anticoagulation [3,12].
10.5. Other Considerations
The management of complications of portal hypertension should not differ from
those of other patients with cirrhosis. Anticoagulation must be started always after imple-
menting an adequate prophylaxis for gastrointestinal bleeding [1]. Therefore, decompen-
sated patients not receiving NSBB should undergo screening endoscopy while patients
with compensated advanced chronic liver disease with unequivocal signs of clinically sig-
nificant portal hypertension may be started directly on NSBB. Otherwise, an upper endos-
copy should be performed in patients not fulfilling the Baveno VI criteria [4]. If these cri-
teria are met, this decision should be individualized depending on the age and extent of
PVT and on whether anticoagulant therapy is to be initiated. Importantly, recent data sug-
gest that anticoagulation should not be delayed until variceal eradication or complete β-
blockade is achieved [3]. This recommendation also applies to the setting of secondary
prophylaxis. Indeed, the continuation of LMWH through prophylactic endoscopic vari-
ceal ligation did not increase the risk of bleeding in patients with cirrhosis [172], with
similar findings in patients with non-cirrhotic PVT [173]. Further data in this regard are
needed.
Figure 5.
The algorithm for the management of portal vein thrombosis in patients with cirrhosis
according to the Baveno VII consensus recommendations.
10.4. Thrombolysis
Local or systemic thrombolysis was proposed as an adjunct to anticoagulation for
the treatment of recent PVT. Small case series have reported similar efficacy rates to those
achieved with anticoagulation alone but with a high risk of procedure-related morbidity
and mortality [
169
171
]. Based on these preliminary observations, current guidelines only
recommend thrombolitic therapy in specialized centers for very selected patients in whom
intestinal ischemia persists despite anticoagulation [3,12].
10.5. Other Considerations
The management of complications of portal hypertension should not differ from those
of other patients with cirrhosis. Anticoagulation must be started always after implementing
an adequate prophylaxis for gastrointestinal bleeding [
1
]. Therefore, decompensated
patients not receiving NSBB should undergo screening endoscopy while patients with
compensated advanced chronic liver disease with unequivocal signs of clinically significant
portal hypertension may be started directly on NSBB. Otherwise, an upper endoscopy
J. Clin. Med. 2022,11, 6435 18 of 26
should be performed in patients not fulfilling the Baveno VI criteria [
4
]. If these criteria are
met, this decision should be individualized depending on the age and extent of PVT and
on whether anticoagulant therapy is to be initiated. Importantly, recent data suggest that
anticoagulation should not be delayed until variceal eradication or complete
β
-blockade is
achieved [
3
]. This recommendation also applies to the setting of secondary prophylaxis.
Indeed, the continuation of LMWH through prophylactic endoscopic variceal ligation did
not increase the risk of bleeding in patients with cirrhosis [
172
], with similar findings in
patients with non-cirrhotic PVT [173]. Further data in this regard are needed.
Patients with HCC invasion of the portal vein do not benefit from anticoagulation [
2
].
However, in some rare cases, the initial “malignant” thrombus induces a stagnant flow that
can lead to clot formation and symptomatic PVT (e.g., intestinal ischemia). In this infrequent
scenario, anticoagulation may be considered after weighing the risk and benefits. In regard
to cancer-associated symptomatic recent (non-cirrhotic) splanchnic vein thrombosis, the
International Society on Thrombosis and Haemostasis recommends LMWH or DOACs.
They suggest LMWH in patients with luminal gastrointestinal cancer, active gastrointestinal
mucosal abnormalities, genitourinary cancer at high risk of bleeding, or receiving current
systemic therapy with potentially relevant drug–drug interactions with DOACs [12].
Finally, we have a few words regarding the prevention of PVT after LT. Pre-LT PVT
is a risk factor for PVT recurrence, with a greater risk if a non-anatomical anastomosis is
performed or if pre-LT PVT was of a great degree and extension [
98
]. In the recent consensus
of the Spanish Society of Liver Transplantation and the Spanish Society of Thrombosis and
Hemostasis, therapeutic LMWH (i.e., 1 mg/kg) started within the first 24 h after surgery is
recommended in patients with risk factors of PVT in the absence of coagulopathy, liver graft
dysfunction, or low platelet count (<30,000–50,000/
µ
L). Risk factors include pre-LT PVT,
slow portal flow (after reperfusion) defined as <1300 mL/min or <65 mL/min/100 g, partial
thrombectomy or vein intimal layer lesion during thrombectomy, nonphysiologically portal
vein inflow reconstruction, and thrombophilic disorders in the recipient. In the absence of
complications, therapy should be prolonged at least 2 months after LT and individualized
thereafter [174].
11. Conclusions
Portal vein thrombosis constitutes the most common thrombotic event in patients with
cirrhosis. The recent incorporation of DOACs and of new radiological techniques for portal
vein recanalization are major breakthroughs in the treatment of this complication. However,
the lack of large prospective observational studies and randomized trials explain the
uncertainties regarding its impact on the natural history of cirrhosis and the heterogenous
diagnostic and therapeutic recommendations of current guidelines. Thus, future research
to fill current gaps of knowledge is needed and will likely require multicenter collaboration.
Author Contributions:
A.O.: investigation, resources, data curation, writing—original draft prepara-
tion, writing—review and editing. J.I.F.: investigation, resources, data curation, writing—original
draft preparation, writing—review and editing, supervision, project administration. Á.P., A.C., C.R.,
Á.A., R.P., F.J.G., E.F. and J.C.: visualization and supervision. All authors have read and agreed to the
published version of the manuscript.
Funding:
JoséIgnacio Fortea was supported by grants from the Carlos III Health Institute (PI20/01258)
and the Spanish Association for Study of the Liver (AEEH, Juan Cordoba grant).
Data Availability Statement: Not applicable.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Vascular diseases of the liver. J. Hepatol.
2016
,
64, 179–202. [CrossRef] [PubMed]
2.
Simonetto, D.A.; Singal, A.K.; Garcia-Tsao, G.; Caldwell, S.H.; Ahn, J.; Kamath, P.S. ACG Clinical Guideline: Disorders of the
Hepatic and Mesenteric Circulation. Am. J. Gastroenterol. 2020,115, 18–40. [CrossRef] [PubMed]
J. Clin. Med. 2022,11, 6435 19 of 26
3.
Northup, P.G.; Garcia-Pagan, J.C.; Garcia-Tsao, G.; Intagliata, N.M.; Superina, R.A.; Roberts, L.N.; Lisman, T.; Valla, D.C. Vascular
Liver Disorders, Portal Vein Thrombosis, and Procedural Bleeding in Patients With Liver Disease: 2020 Practice Guidance by the
American Association for the Study of Liver Diseases. Hepatology 2021,73, 366–413. [CrossRef]
4.
de Franchis, R.; Bosch, J.; Garcia-Tsao, G.; Reiberger, T.; Ripoll, C. Baveno VII - Renewing consensus in portal hypertension. J.
Hepatol. 2022,76, 959–974. [CrossRef] [PubMed]
5.
Janssen, H.L.; Meinardi, J.R.; Vleggaar, F.P.; van Uum, S.H.; Haagsma, E.B.; van Der Meer, F.J.; van Hattum, J.; Chamuleau, R.A.;
Adang, R.P.; Vandenbroucke, J.P.; et al. Factor V Leiden mutation, prothrombin gene mutation, and deficiencies in coagulation
inhibitors associated with Budd-Chiari syndrome and portal vein thrombosis: Results of a case-control study. Blood
2000
,96,
2364–2368. [PubMed]
6.
Janssen, H.L.; Wijnhoud, A.; Haagsma, E.B.; Van Uum, S.H.M.; Van Nieuwkerk, C.M.J.; Adang, R.P.; Chamuleau, R.A.; Van
Hattum, J.; Vleggaar, F.P.; Hansen, B.; et al. Extrahepatic portal vein thrombosis: Aetiology and determinants of survival. Gut
2001,49, 720–724. [CrossRef] [PubMed]
7.
Sogaard, K.K.; Astrup, L.B.; Vilstrup, H.; Gronbaek, H. Portal vein thrombosis; risk factors, clinical presentation and treatment.
BMC Gastroenterol. 2007,7, 34. [CrossRef]
8.
Valla, D.; Casadevall, N.; Huisse, M.G.; Tulliez, M.; Grange, J.D.; Muller, O.; Binda, T.; Varet, B.; Rueff, B.; Benhamou, J.P. Etiology
of portal vein thrombosis in adults. A prospective evaluation of primary myeloproliferative disorders. Gastroenterol.
1988
,94,
1063–1069. [CrossRef]
9. Martin-Llahi, M.; Albillos, A.; Banares, R.; Berzigotti, A.; Garcia-Criado, M.A.; Genesca, J.; Hernandez-Gea, V.; Llop-Herrera, E.;
Masnou-Ridaura, H.; Mateoet, J.; et al. Vascular diseases of the liver. Clinical Guidelines from the Catalan Society of Digestology
and the Spanish Association for the Study of the Liver. Gastroenterol. Hepatol. 2017,40, 538–580. [CrossRef]
10.
Intagliata, N.M.; Argo, C.K.; Stine, J.G.; Lisman, T.; Caldwell, S.H.; Violi, F. Concepts and controversies in haemostasis and
thrombosis associated with liver disease: Proceedings of the 7th International Coagulation in Liver Disease Conference. Thromb.
Haemost. 2018,118, 1491–1506.
11.
O’Leary, J.G.; Greenberg, C.S.; Patton, H.M.; Caldwell, S.H. AGA Clinical Practice Update: Coagulation in Cirrhosis.
Gastroenterology 2019,157, 34–43.e1. [CrossRef] [PubMed]
12.
Di Nisio, M.; Valeriani, E.; Riva, N.; Schulman, S.; Beyer-Westendorf, J.; Ageno, W. Anticoagulant therapy for splanchnic vein
thrombosis: ISTH SSC Subcommittee Control of Anticoagulation. J. Thromb. Haemost.
2020
,18, 1562–1568. [CrossRef] [PubMed]
13.
Fortea, J.I.; Puente, Á.; Cuadrado, A.; Huelin, P.; Pellón, R.; González Sánchez, F.J.; Mayorga, M.; Cagigal, M.L.; Carrera, I.G.;
Cobreros, M.; et al. Congestive Hepatopathy. Int. J. Mol. Sci. 2020,21, 9420. [CrossRef] [PubMed]
14.
Krumm, P.; Schraml, C.; Bretschneider, C.; Seeger, A.; Klumpp, B.; Kramer, U.; Claussen, C.D.; Miller, S. Depiction of variants of
the portal confluence venous system using multidetector row CT: Analysis of 916 cases. Rofo 2011,183, 1123–1129. [CrossRef]
15.
Cheng, Y.F.; Huang, T.L.; Lee, T.Y.; Chen, T.Y.; Chen, C.L. Variation of the intrahepatic portal vein; angiographic demonstration
and application in living-related hepatic transplantation. Transplant. Proc. 1996,28, 1667–1668. [PubMed]
16.
Yerdel, M.A.; Gunson, B.; Mirza, D.; Karayalçin, K.; Olliff, S.; Buckels, J.; Mayer, D.; McMaster, P.; Pirenne, J. Portal vein thrombosis
in adults undergoing liver transplantation: Risk factors, screening, management, and outcome. Transplantation
2000
,69, 1873–1881.
[CrossRef]
17.
Bauer, J.; Johnson, S.; Durham, J.; Ludkowski, M.; Trotter, J.; Bak, T.; Wachs, M. The role of TIPS for portal vein patency in liver
transplant patients with portal vein thrombosis. Liver Transplant. 2006,12, 1544–1551. [CrossRef]
18.
Sarin, S.K.; Philips, C.A.; Kamath, P.S.; Choudhury, A.; Maruyama, H.; Nery, F.G.; Valla, D.C. Toward a Comprehensive New
Classification of Portal Vein Thrombosis in Patients with Cirrhosis. Gastroenterology 2016,151, 574–577.e573. [CrossRef]
19.
Bhangui, P.; Lim, C.; Levesque, E.; Salloum, C.; Lahat, E.; Feray, C.; Azoulay, D. Novel classification of non-malignant portal vein
thrombosis: A guide to surgical decision-making during liver transplantation. J. Hepatol. 2019,71, 1038–1050. [CrossRef]
20.
Plessier, A.; Darwish-Murad, S.; Hernandez-Guerra, M.; Consigny, Y.; Fabris, F.; Trebicka, J.; Heller, J.; Morard, I.; Lasser, L.;
Langlet, P.; et al. Acute portal vein thrombosis unrelated to cirrhosis: A prospective multicenter follow-up study. Hepatology
2010
,
51, 210–218. [CrossRef]
21.
Senzolo, M.; Sartori, T.M.; Rossetto, V.; Burra, P.; Cillo, U.; Boccagni, P.; Gasparini, D.; Miotto, D.; Simioni, P.; Tsochatzis, E.; et al.
Prospective evaluation of anticoagulation and transjugular intrahepatic portosystemic shunt for the management of portal vein
thrombosis in cirrhosis. Liver Int. 2012,32, 919–927. [CrossRef] [PubMed]
22.
Maruyama, H.; Takahashi, M.; Shimada, T.; Yokosuka, O. Emergency anticoagulation treatment for cirrhosis patients with portal
vein thrombosis and acute variceal bleeding. Scand. J. Gastroenterol. 2012,47, 686–691. [CrossRef] [PubMed]
23.
De Gaetano, A.M.; Lafortune, M.; Patriquin, H.; De Franco, A.; Aubin, B.; Paradis, K. Cavernous transformation of the portal vein:
Patterns of intrahepatic and splanchnic collateral circulation detected with Doppler sonography. AJR Am. J. Roentgenol.
1995
,165,
1151–1155. [CrossRef] [PubMed]
24.
Anton, A.; Campreciós, G.; Pérez-Campuzano, V.; Orts, L.; García-Pagán, J.C.; Hernández-Gea, V. The Pathophysiology of Portal
Vein Thrombosis in Cirrhosis: Getting Deeper into Virchow’s Triad. J. Clin. Med. 2022,11, 800. [CrossRef] [PubMed]
25.
Zocco, M.A.; Di Stasio, E.; De Cristofaro, R.; Novi, M.; Ainora, M.E.; Ponziani, F.; Riccardi, L.; Lancellotti, S.; Santoliquido, A.;
Flore, R.; et al. Thrombotic risk factors in patients with liver cirrhosis: Correlation with MELD scoring system and portal vein
thrombosis development. J. Hepatol. 2009,51, 682–689. [CrossRef]
J. Clin. Med. 2022,11, 6435 20 of 26
26.
Abdel-Razik, A.; Mousa, N.; Elhelaly, R.; Tawfik, A. De-novo portal vein thrombosis in liver cirrhosis: Risk factors and correlation
with the Model for End-stage Liver Disease scoring system. Eur. J. Gastroenterol. Hepatol. 2015,27, 585–592. [CrossRef]
27.
Stine, J.G.; Wang, J.; Shah, P.M.; Argo, C.K.; Intagliata, N.; Uflacker, A.; Caldwell, S.H.; Northup, P.G. Decreased portal vein
velocity is predictive of the development of portal vein thrombosis: A matched case-control study. Liver Int.
2018
,38, 94–101.
[CrossRef]
28.
Turon, F.; Driever, E.G.; Baiges, A.; Cerda, E.; García-Criado, Á.; Gilabert, R.; Bru, C.; Berzigotti, A.; Nuñez, I.; Orts, L.; et al.
Predicting portal thrombosis in cirrhosis: A prospective study of clinical, ultrasonographic and hemostatic factors. J. Hepatol.
2021,75, 1367–1376. [CrossRef]
29.
Nery, F.; Chevret, S.; Condat, B.; de Raucourt, E.; Boudaoud, L.; Rautou, P.E.; Plessier, A.; Roulot, D.; Chaffaut, C.; Bourcier,
V.; et al. Causes and consequences of portal vein thrombosis in 1,243 patients with cirrhosis: Results of a longitudinal study.
Hepatology 2015,61, 660–667. [CrossRef]
30.
Maruyama, H.; Okugawa, H.; Takahashi, M.; Yokosuka, O. De novo portal vein thrombosis in virus-related cirrhosis: Predictive
factors and long-term outcomes. Am. J. Gastroenterol. 2013,108, 568–574. [CrossRef]
31.
Dong, G.; Huang, X.Q.; Zhu, Y.L.; Ding, H.; Li, F.; Chen, S.Y. Increased portal vein diameter is predictive of portal vein thrombosis
development in patients with liver cirrhosis. Ann. Transl. Med. 2021,9, 289. [CrossRef] [PubMed]
32.
Luca, A.; Miraglia, R.; Caruso, S.; Milazzo, M.; Sapere, C.; Maruzzelli, L.; Vizzini, G.; Tuzzolino, F.; Gridelli, B.; Bosch, J. Short-
and long-term effects of the transjugular intrahepatic portosystemic shunt on portal vein thrombosis in patients with cirrhosis.
Gut 2011,60, 846–852. [CrossRef] [PubMed]
33.
Nery, F.; Correia, S.; Macedo, C.; Gandara, J.; Lopes, V.; Valadares, D.; Ferreira, S.; Oliveira, J.; Gomes, M.T.; Lucas, R.; et al.
Nonselective beta-blockers and the risk of portal vein thrombosis in patients with cirrhosis: Results of a prospective longitudinal
study. Aliment. Pharmacol. Ther. 2019,49, 582–588. [CrossRef] [PubMed]
34.
Xu, X.; Xu, S.; Primignani, M.; De Stefano, V.; He, Y.; Yi, F.; Guo, X.; Valla, D.; Qi, X. Nonselective
β
-Blockers May Progress the
Thrombosis of Portal Venous System in Cirrhotic Patients: A Retrospective Observational Study. Adv. Ther.
2020
,37, 1452–1463.
[CrossRef] [PubMed]
35.
Xu, X.; Guo, X.; De Stefano, V.; Silva-Junior, G.; Goyal, H.; Bai, Z.; Zhao, Q.; Qi, X. Nonselective beta-blockers and development of
portal vein thrombosis in liver cirrhosis: A systematic review and meta-analysis. Hepatol. Int. 2019,13, 468–481. [CrossRef]
36.
EASL. Clinical Practice Guidelines on prevention and management of bleeding and thrombosis in patients with cirrhosis. J.
Hepatol. 2022,76, 1151–1184. [CrossRef]
37. Turco, L.; de Raucourt, E.; Valla, D.C.; Villa, E. Anticoagulation in the cirrhotic patient. JHEP Rep. 2019,1, 227–239. [CrossRef]
38.
Tripodi, A.; Primignani, M.; Chantarangkul, V.; Dell’Era, A.; Clerici, M.; de Franchis, R.; Colombo, M.; Mannucci, P.M. An
imbalance of pro- vs anti-coagulation factors in plasma from patients with cirrhosis. Gastroenterology
2009
,137, 2105–2111.
[CrossRef]
39.
Scheiner, B.; Balcar, L.; Nussbaumer, R.J.; Weinzierl, J.; Paternostro, R.; Simbrunner, B.; Hartl, L.; Jachs, M.; Bauer, D.; Stättermayer,
A.F.; et al. Factor VIII/protein C ratio independently predicts liver-related events but does not indicate a hypercoagulable state in
ACLD. J. Hepatol. 2022,76, 1090–1099. [CrossRef]
40.
Kalambokis, G.N.; Oikonomou, A.; Christou, L.; Baltayiannis, G. High von Willebrand factor antigen levels and procoagulant
imbalance may be involved in both increasing severity of cirrhosis and portal vein thrombosis. Hepatology
2016
,64, 1383–1385.
[CrossRef]
41.
Ren, W.; Zhang, J.; Chen, Y.; Wen, M.; Su, Y.; Zhao, Y.; Lu, S.; Wu, J. Evaluation of Coagulation, Fibrinolysis and Endothe-
lial Biomarkers in Cirrhotic Patients With or Without Portal Venous Thrombosis. Clin. Appl Thromb. Hemost.
2020
,26,
1076029620982666. [CrossRef] [PubMed]
42.
Zanetto, A.; Pelizzaro, F.; Campello, E.; Bulato, C.; Balcar, L.; Gu, W.; Gavasso, S.; Saggiorato, G.; Zeuzem, S.; Russo, F.P.; et al.
Severity of systemic inflammation is the main predictor of ACLF and bleeding in patients with acutely decompensated cirrhosis.
J. Hepatol. 2022, S0168-8278(22)03110-5. [CrossRef]
43.
Delahousse, B.; Labat-Debelleix, V.; Decalonne, L.; d’Alteroche, L.; Perarnau, J.M.; Gruel, Y. Comparative study of coagulation and
thrombin generation in the portal and jugular plasma of patients with cirrhosis. Thromb. Haemost.
2010
,104, 741–749. [PubMed]
44.
Praktiknjo, M.; Trebicka, J.; Carnevale, R.; Pastori, D.; Queck, A.; Ettorre, E.; Violi, F. Von Willebrand and Factor VIII Portosystemic
Circulation Gradient in Cirrhosis: Implications for Portal Vein Thrombosis. Clin. Transl. Gastroenterol.
2020
,11, e00123. [CrossRef]
45.
Driever, E.G.; Magaz, M.; Adelmeijer, J.; Turon, F.; Baiges, A.; Olivas, P.; Pérez-Campuzano, V.; Hernandez-Gea, V.; Blasi, A.;
Garcia-Pagan, J.; et al. The portal vein in patients with cirrhosis is not an excessively inflammatory or hypercoagulable vascular
bed, a prospective cohort study. J. Thromb. Haemost. 2022,20, 2075–2082. [CrossRef]
46.
Shalaby, S.; Zanetto, A.; Campello, E.; Gavasso, S.; Barbiero, G.; Battistel, M.; Feltracco, P.; Bizzaro, D.; Burra, P.; Simioni, P.; et al.
Reply to “Peripheral versus central venous blood sampling does not influence the assessment of platelet activation in cirrhosis”.
Platelets 2022,33, 1104–1106. [CrossRef]
47.
Zanetto, A.; Campello, E.; Bulato, C.; Gavasso, S.; Farinati, F.; Russo, F.P.; Tormene, D.; Burra, P.; Senzolo, M.; Simioni, P. Increased
platelet aggregation in patients with decompensated cirrhosis indicates higher risk of further decompensation and death. J.
Hepatol. 2022,77, 660–669. [CrossRef]
J. Clin. Med. 2022,11, 6435 21 of 26
48.
Quan, X.; Ye, X.; Qian, S.; Wei, B.; Tong, H.; Wang, Z.; Tai, Y.; Guo, X.; Gao, J.; Wu, H. Portal vein thrombosis associates with
high platelet-fibrin clot strength and platelet activation in decompensated cirrhosis: A retrospective study. Dig. Liver Dis.
2022
.
[CrossRef]
49.
Zanetto, A.; Campello, E.; Burra, P.; Senzolo, M.; Simioni, P. Increased platelet ratio in patients with decompensated cirrhosis
indicates higher risk of portal vein thrombosis. Liver Int. 2022. [CrossRef]
50.
Sacco, M.; Tardugno, M.; Lancellotti, S.; Ferretti, A.; Ponziani, F.R.; Riccardi, L.; Zocco, M.A.; De Magistris, A.; Santopaolo, F.;
Pompili, M.; et al. ADAMTS-13/von Willebrand factor ratio: A prognostic biomarker for portavein thrombosis in compensated
cirrhosis. A prospective observational study. Dig. Liver Dis. 2022, S1590-8658(22)00543-6. [CrossRef]
51.
Arroyo, V.; Angeli, P.; Moreau, R.; Jalan, R.; Clària, J.; Trebicka, J.; Fernández, J.; Gustot, T.; Caraceni, P.; Bernardi, M. The systemic
inflammation hypothesis: Towards a new paradigm of acute decompensation and multiorgan failure in cirrhosis. J. Hepatol.
2021
,
74, 670–685. [CrossRef] [PubMed]
52.
Zanetto, A.; Campello, E.; Pelizzaro, F.; Farinati, F.; Burra, P.; Simioni, P.; Senzolo, M. Haemostatic alterations in patients with
cirrhosis and hepatocellular carcinoma: Laboratory evidence and clinical implications. Liver Int.
2022
,42, 1229–1240. [CrossRef]
[PubMed]
53.
Basili, S.; Carnevale, R.; Nocella, C.; Bartimoccia, S.; Raparelli, V.; Talerico, G.; Stefanini, L.; Romiti, G.F.; Perticone, F.; Corazza,
G.R.; et al. Serum Albumin Is Inversely Associated With Portal Vein Thrombosis in Cirrhosis. Hepatol. Commun.
2019
,3, 504–512.
[CrossRef]
54.
Caraceni, P.; Riggio, O.; Angeli, P.; Alessandria, C.; Neri, S.; Foschi, F.G.; Levantesi, F.; Airoldi, A.; Boccia, S.; Svegliati-Baroni, G.;
et al. Long-term albumin administration in decompensated cirrhosis (ANSWER): An open-label randomised trial. Lancet
2018
,
391, 2417–2429. [CrossRef]
55.
Solà, E.; Solé, C.; Simón-Talero, M.; Martín-Llahí, M.; Castellote, J.; Garcia-Martínez, R.; Moreira, R.; Torrens, M.; Márquez, F.;
Fabrellas, N.; et al. Midodrine and albumin for prevention of complications in patients with cirrhosis awaiting liver transplantation.
A randomized placebo-controlled trial. J. Hepatol. 2018,69, 1250–1259. [CrossRef]
56.
Mahmoud, A.E.; Elias, E.; Beauchamp, N.; Wilde, J.T. Prevalence of the factor V Leiden mutation in hepatic and portal vein
thrombosis. Gut 1997,40, 798–800. [CrossRef]
57.
Amitrano, L.; Guardascione, M.A.; Brancaccio, V.; Margaglione, M.; Manguso, F.; Iannaccone, L.; Grandone, E.; Balzano, A. Risk
factors and clinical presentation of portal vein thrombosis in patients with liver cirrhosis. J. Hepatol.
2004
,40, 736–741. [CrossRef]
58.
Amitrano, L.; Brancaccio, V.; Guardascione, M.A.; Margaglione, M.; Iannaccone, L.; D’Andrea, G.; Marmo, R.; Ames, P.R.; Balzano,
A. Inherited coagulation disorders in cirrhotic patients with portal vein thrombosis. Hepatology 2000,31, 345–348. [CrossRef]
59.
Amitrano, L.; Guardascione, M.A.; Ames, P.R.; Margaglione, M.; Iannaccone, L.; Brancaccio, V.; Balzano, A. Increased plasma
prothrombin concentration in cirrhotic patients with portal vein thrombosis and prothrombin G20210A mutation. Thromb.
Haemost. 2006,95, 221–223.
60.
Amitrano, L.; Ames, P.R.; Guardascione, M.A.; Lopez, L.R.; Menchise, A.; Brancaccio, V.; Iannaccone, L.; Balzano, A. Antiphos-
pholipid antibodies and antiphospholipid syndrome: Role in portal vein thrombosis in patients with and without liver cirrhosis.
Clin. Appl. Thromb. Hemost. 2011,17, 367–370. [CrossRef]
61.
Mangia, A.; Villani, M.R.; Cappucci, G.; Santoro, R.; Ricciardi, R.; Facciorusso, D.; Leandro, G.; Caruso, N.; Andriulli, A. Causes of
portal venous thrombosis in cirrhotic patients: The role of genetic and acquired factors. Eur. J. Gastroenterol. Hepatol.
2005
,17,
745–751. [CrossRef] [PubMed]
62.
Pasta, L.; Marrone, C.; D’Amico, M.; Virdone, R.; D
'
Amico, G.; Sammarco, P.; Fabiano, C.; Pagliaro, L. MTHFR C677T mutations
in liver cirrhosis with and without portal vein thrombosis. Liver Int. 2006,26, 269–270. [CrossRef] [PubMed]
63.
Pasta, L.; Pasta, F.; D’Amico, M. PAI-1 4G-4G, MTHFR 677TT, V Leiden 506Q, and Prothrombin 20210A in Splanchnic Vein
Thrombosis: Analysis of Individual Patient Data from Three Prospective Studies. J. Clin. Exp. Hepatol.
2016
,6, 10–14. [CrossRef]
[PubMed]
64.
Colaizzo, D.; Amitrano, L.; Guardascione, M.A.; Balzano, A.; Margaglione, M. Janus kinase-2 mutation, cirrhosis and splanchnic
vein thrombosis. Eur. J. Gastroenterol. Hepatol. 2008,20, 245–246. [CrossRef] [PubMed]
65.
Gabr, M.A.; Bessa, S.S.; El-Zamarani, E.A. Portal vein thrombosis in Egyptian patients with liver cirrhosis: Role of methylenete-
trahydrofolate reductase C677T gene mutation. Hepatol. Res. 2010,40, 486–493. [CrossRef]
66.
Ayala, R.; Grande, S.; Bustelos, R.; Ribera, C.; García-Sesma, A.; Jimenez, C.A.; Moreno, E.; Martínez-López, J. Obesity is an
independent risk factor for pre-transplant portal vein thrombosis in liver recipients. BMC Gastroenterol.
2012
,12, 114. [CrossRef]
67.
Qi, X.; Zhang, C.; Han, G.; Zhang, W.; He, C.; Yin, Z.; Liu, Z.; Bai, W.; Li, R.; Bai, M.; et al. Prevalence of the JAK2V617F mutation
in Chinese patients with Budd-Chiari syndrome and portal vein thrombosis: A prospective study. J. Gastroenterol. Hepatol.
2012
,
27, 1036–1043. [CrossRef]
68.
Werner, K.T.; Sando, S.; Carey, E.J.; Vargas, H.E.; Byrne, T.J.; Douglas, D.D.; Harrison, M.E.; Rakela, J.; Aqel, B.A. Portal vein
thrombosis in patients with end stage liver disease awaiting liver transplantation: Outcome of anticoagulation. Dig. Dis. Sci.
2013,58, 1776–1780. [CrossRef]
69.
Karakose, S.; Oruc, N.; Zengin, M.; Akarca, U.S.; Ersoz, G. Diagnostic value of the JAK2 V617F mutation for latent chronic
myeloproliferative disorders in patients with Budd-Chiari syndrome and/or portal vein thrombosis. Turk. J. Gastroenterol.
2015
,
26, 42–48. [CrossRef]
J. Clin. Med. 2022,11, 6435 22 of 26
70.
Saugel, B.; Lee, M.; Feichtinger, S.; Hapfelmeier, A.; Schmid, R.M.; Siveke, J.T. Thrombophilic factor analysis in cirrhotic patients
with portal vein thrombosis. J. Thromb. Thrombolysis 2015,40, 54–60. [CrossRef]
71.
Lancellotti, S.; Basso, M.; Veca, V.; Sacco, M.; Riccardi, L.; Pompili, M.; De Cristofaro, R. Presence of portal vein thrombosis in liver
cirrhosis is strongly associated with low levels of ADAMTS-13: A pilot study. Intern. Emerg. Med.
2016
,11, 959–967. [CrossRef]
[PubMed]
72.
Ventura, P.; Venturelli, G.; Marcacci, M.; Fiorini, M.; Marchini, S.; Cuoghi, C.; Pietrangelo, A. Hyperhomocysteinemia and MTHFR
C677T polymorphism in patients with portal vein thrombosis complicating liver cirrhosis. Thromb. Res.
2016
,141, 189–195.
[CrossRef] [PubMed]
73.
Artaza, T.; Lopes, M.; Romero, M.; Gomez, A.Z.; de la Cruz, G.; Sanchez, J.J.; Gonzalez, C.; Gómez, R. Efficacy and safety of
anticoagulation in non-malignant portal vein thrombosis in patients with liver cirrhosis. Gastroenterol. Hepatol.
2018
,41, 611–617.
[CrossRef] [PubMed]
74.
Senzolo, M.; Riva, N.; Dentali, F.; Rodriguez-Castro, K.; Sartori, M.T.; Bang, S.-M.; Martinelli, I.; Schulman, S.; Alatri, A.;
Beyer-Westendorf, J.; et al. Long-Term Outcome of Splanchnic Vein Thrombosis in Cirrhosis. Clin. Transl. Gastroenterol.
2018
,9,
176. [CrossRef]
75.
Cagin, Y.F.; Bilgic, Y.; Berber, I.; Yildirim, O.; Erdogan, M.A.; Firat, F.; Arslan, A.K.; Colak, C.; Seckin, Y.; Harputluoglu, M. The
risk factors of portal vein thrombosis in patients with liver cirrhosis. Exp. Ther. Med. 2019,17, 3189–3194. [CrossRef]
76.
Tremblay, D.; Naymagon, L.; Troy, K.; Cromwell, C.; Edwards, C.; Schiano, T.; Kremyanskaya, M.; Mascarenhas, J. The utility
of thrombophilia testing in patients with newly diagnosed portal vein thrombosis. Blood Coagul. Fibrinolysis
2020
,31, 213–218.
[CrossRef]
77.
Fortea, J.I.; Carrera, I.G.; Puente, Á.; Cuadrado, A.; Huelin, P.; Tato, C.; Fernández, P.; Montes, M.D.R.P.; Céspedes, J.N.; López,
A.B.; et al. Portal Thrombosis in Cirrhosis: Role of Thrombophilic Disorders. J. Clin. Med. 2020,9, 2822. [CrossRef]
78.
Dentali, F.; Galli, M.; Gianni, M.; Ageno, W. Inherited thrombophilic abnormalities and risk of portal vein thrombosis. a
meta-analysis. Thromb. Haemost. 2008,99, 675–682.
79.
Qi, X.; Ren, W.; De Stefano, V.; Fan, D. Associations of coagulation factor V Leiden and prothrombin G20210A mutations with
Budd-Chiari syndrome and portal vein thrombosis: A systematic review and meta-analysis. Clin. Gastroenterol. Hepatol.
2014
,12,
1801–1812.e1807. [CrossRef]
80.
Ma, S.D.; Wang, J.; Bezinover, D.; Kadry, Z.; Northup, P.G.; Stine, J.G. Inherited thrombophilia and portal vein thrombosis in
cirrhosis: A systematic review and meta-analysis. Res. Pract. Thromb. Haemost. 2019,3, 658–667. [CrossRef]
81.
Qi, X.; Chen, H.; Han, G. Effect of antithrombin, protein C and protein S on portal vein thrombosis in liver cirrhosis: A
meta-analysis. Am. J. Med. Sci. 2013,346, 38–44. [CrossRef] [PubMed]
82.
Colucci, G.; Tsakiris, D.A. Thrombophilia screening revisited: An issue of personalized medicine. J. Thromb. Thrombolysis
2020
,49,
618–629. [CrossRef] [PubMed]
83.
Intagliata, N.M.; Caldwell, S.H.; Tripodi, A. Diagnosis, Development, and Treatment of Portal Vein Thrombosis in Patients with
and without Cirrhosis. Gastroenterology 2019,156, 1582–1599.e1581. [CrossRef] [PubMed]
84. Shukla, A.; Giri, S. Portal Vein Thrombosis in Cirrhosis. J. Clin. Exp. Hepatol. 2022,12, 965–979. [CrossRef] [PubMed]
85. Noronha Ferreira, C.; Marinho, R.T.; Cortez-Pinto, H.; Ferreira, P.; Dias, M.S.; Vasconcelos, M.; Alexandrino, P.; Serejo, F.; Pedro,
A.J.; Gonçalves, A.; et al. Incidence, predictive factors and clinical significance of development of portal vein thrombosis in
cirrhosis: A prospective study. Liver Int. 2019,39, 1459–1467. [CrossRef]
86.
Violi, F.; Corazza, G.R.; Caldwell, S.H.; Talerico, G.; Romiti, G.F.; Napoleone, L.; Perticone, F.; Bolondi, L.; Pietrangelo, A.; Vestri,
A.R.; et al. Incidence and Recurrence of Portal Vein Thrombosis in Cirrhotic Patients. Thromb. Haemost.
2019
,119, 496–499.
[CrossRef]
87.
Pan, J.; Wang, L.; Gao, F.; An, Y.; Yin, Y.; Guo, X.; Nery, F.G.; Yoshida, E.M.; Qi, X. Epidemiology of portal vein thrombosis in liver
cirrhosis: A systematic review and meta-analysis. Eur. J. Intern. Med. 2022,104, 21–32. [CrossRef]
88.
Ogren, M.; Bergqvist, D.; Björck, M.; Acosta, S.; Eriksson, H.; Sternby, N.H. Portal vein thrombosis: Prevalence, patient
characteristics and lifetime risk: A population study based on 23,796 consecutive autopsies. World J. Gastroenterol.
2006
,12,
2115–2119. [CrossRef]
89.
Stine, J.G.; Shah, N.L.; Argo, C.K.; Pelletier, S.J.; Caldwell, S.H.; Northup, P.G. Increased risk of portal vein thrombosis in patients
with cirrhosis due to nonalcoholic steatohepatitis. Liver Transplant. 2015,21, 1016–1021. [CrossRef]
90.
Zanetto, A.; Garcia-Tsao, G. Some Answers and More Questions About Portal Vein Thrombosis in Patients with Decompensated
Cirrhosis. Clin. Gastroenterol. Hepatol. 2020,18, 2432–2434. [CrossRef]
91.
Berry, K.; Taylor, J.; Liou, I.W.; Ioannou, G.N. Portal vein thrombosis is not associated with increased mortality among patients
with cirrhosis. Clin. Gastroenterol. Hepatol. 2015,13, 585–593. [CrossRef] [PubMed]
92.
John, B.V.; Konjeti, R.; Aggarwal, A.; Lopez, R.; Atreja, A.; Miller, C.; Zein, N.N.; Carey, W. Impact of untreated portal vein
thrombosis on pre and post liver transplant outcomes in cirrhosis. Ann. Hepatol. 2013,12, 952–958. [CrossRef]
93.
Luca, A.; Caruso, S.; Milazzo, M.; Marrone, G.; Mamone, G.; Crinò, F.; Maruzzelli, L.; Miraglia, R.; Floridia, G.; Vizzini, G. Natural
course of extrahepatic nonmalignant partial portal vein thrombosis in patients with cirrhosis. Radiology
2012
,265, 124–132.
[CrossRef]
J. Clin. Med. 2022,11, 6435 23 of 26
94.
Villa, E.; Cammà, C.; Marietta, M.; Luongo, M.; Critelli, R.; Colopi, S.; Tata, C.; Zecchini, R.; Gitto, S.; Petta, S.; et al. Enoxaparin
prevents portal vein thrombosis and liver decompensation in patients with advanced cirrhosis. Gastroenterology
2012
,143,
1253–1260.e4. [CrossRef]
95.
D’Amico, G.; De Franchis, R. Upper digestive bleeding in cirrhosis. Post-therapeutic outcome and prognostic indicators.
Hepatology 2003,38, 599–612. [CrossRef] [PubMed]
96.
Amitrano, L.; Guardascione, M.A.; Scaglione, M.; Menchise, A.; Martino, R.; Manguso, F.; Lanza, A.G.; Lampasi, F. Splanchnic
vein thrombosis and variceal rebleeding in patients with cirrhosis. Eur. J. Gastroenterol. Hepatol.
2012
,24, 1381–1385. [CrossRef]
[PubMed]
97.
Zhang, Y.; Xu, B.-Y.; Wang, X.-B.; Zheng, X.; Huang, Y.; Chen, J.; Meng, Z.-J.; Gao, Y.-H.; Qian, Z.-P.; Liu, F.; et al. Prevalence and
Clinical Significance of Portal Vein Thrombosis in Patients with Cirrhosis and Acute Decompensation. Clin. Gastroenterol. Hepatol.
2020,18, 2564–2572.e1. [CrossRef]
98.
Chen, H.; Turon, F.; Hernandez-Gea, V.; Fuster, J.; Garcia-Criado, A.; Barrufet, M.; Darnell, A.; Fondevila, C.; Garcia-Valdecasas,
J.C.; Garcia-Pagán, J.C. Nontumoral portal vein thrombosis in patients awaiting liver transplantation. Liver Transpl.
2016
,22,
352–365. [CrossRef]
99.
Englesbe, M.J.; Schaubel, D.E.; Cai, S.; Guidinger, M.K.; Merion, R.M. Portal vein thrombosis and liver transplant survival benefit.
Liver Transplant. 2010,16, 999–1005. [CrossRef]
100.
Ghabril, M.; Agarwal, S.; Lacerda, M.; Chalasani, N.; Kwo, P.; Tector, A.J. Portal Vein Thrombosis Is a Risk Factor for Poor Early
Outcomes After Liver Transplantation: Analysis of Risk Factors and Outcomes for Portal Vein Thrombosis in Waitlisted Patients.
Transplantation 2016,100, 126–133. [CrossRef]
101.
Englesbe, M.J.; Kubus, J.; Muhammad, W.; Sonnenday, C.J.; Welling, T.; Punch, J.D.; Lynch, R.J.; Marrero, J.A.; Pelletier, S.J. Portal
vein thrombosis and survival in patients with cirrhosis. Liver Transplant. 2010,16, 83–90. [CrossRef] [PubMed]
102.
Francoz, C.; Belghiti, J.; Vilgrain, V.; Sommacale, D.; Paradis, V.; Condat, B.; Denninger, M.H.; Sauvanet, A.; Valla, D.; Durand, F.
Splanchnic vein thrombosis in candidates for liver transplantation: Usefulness of screening and anticoagulation. Gut
2005
,54,
691–697. [CrossRef] [PubMed]
103.
Rodríguez-Castro, K.I.; Porte, R.J.; Nadal, E.; Germani, G.; Burra, P.; Senzolo, M. Management of nonneoplastic portal vein
thrombosis in the setting of liver transplantation: A systematic review. Transplantation
2012
,94, 1145–1153. [CrossRef] [PubMed]
104.
Chen, H.; Liu, L.; Qi, X.; He, C.; Wu, F.; Fan, D.; Han, G. Efficacy and safety of anticoagulation in more advanced portal vein
thrombosis in patients with liver cirrhosis. Eur. J. Gastroenterol. Hepatol. 2016,28, 82–89. [CrossRef] [PubMed]
105.
Loffredo, L.; Pastori, D.; Farcomeni, A.; Violi, F. Effects of Anticoagulants in Patients with Cirrhosis and Portal Vein Thrombosis:
A Systematic Review and Meta-analysis. Gastroenterology 2017,153, 480–487.e1. [CrossRef]
106.
Francoz, C.; Valla, D.; Durand, F. Portal vein thrombosis, cirrhosis, and liver transplantation. J. Hepatol.
2012
,57, 203–212.
[CrossRef]
107.
Driever, E.G.; von Meijenfeldt, F.A.; Adelmeijer, J.; de Haas, R.J.; van den Heuvel, M.C.; Nagasami, C.; Weisel, J.W.; Fondevila, C.;
Porte, R.J.; Blasi, A.; et al. Nonmalignant portal vein thrombi in patients with cirrhosis consist of intimal fibrosis with or without
a fibrin-rich thrombus. Hepatology 2022,75, 898–911. [CrossRef]
108.
Bach, A.M.; Hann, L.E.; Brown, K.T.; Getrajdman, G.I.; Herman, S.K.; Fong, Y.; Blumgart, L.H. Portal vein evaluation with US:
Comparison to angiography combined with CT arterial portography. Radiology 1996,201, 149–154. [CrossRef]
109.
Brunaud, L.; Antunes, L.; Collinet-Adler, S.; Marchal, F.; Ayav, A.; Bresler, L.; Boissel, P. Acute mesenteric venous thrombosis:
Case for nonoperative management. J. Vasc. Surg. 2001,34, 673–679. [CrossRef]
110.
Lee, H.K.; Park, S.J.; Yi, B.H.; Yeon, E.K.; Kim, J.H.; Hong, H.S. Portal vein thrombosis: CT features. Abdom. Imaging
2008
,33,
72–79. [CrossRef]
111.
Lai, L.; Brugge, W.R. Endoscopic ultrasound is a sensitive and specific test to diagnose portal venous system thrombosis (PVST).
Am. J. Gastroenterol. 2004,99, 40–44. [CrossRef] [PubMed]
112.
Calvet, X.; Bruix, J.; Brú, C.; Ginés, P.; Vilana, R.; Solé, M.; Ayuso, M.D.C.; Bruguera, M.; Rodes, J. Natural history of hepatocellular
carcinoma in Spain. Five year’s experience in 249 cases. J. Hepatol. 1990,10, 311–317. [CrossRef]
113.
Stuart, K.E.; Anand, A.J.; Jenkins, R.L. Hepatocellular carcinoma in the United States. Prognostic features, treatment outcome,
and survival. Cancer 1996,77, 2217–2222. [CrossRef]
114.
Tublin, M.E.; Dodd, G.D.; 3rd Baron, R.L. Benign and malignant portal vein thrombosis: Differentiation by CT characteristics.
AJR Am. J. Roentgenol. 1997,168, 719–723. [CrossRef]
115.
Sandrasegaran, K.; Tahir, B.; Nutakki, K.; Akisik, F.M.; Bodanapally, U.; Tann, M.; Chalasani, N. Usefulness of conventional MRI
sequences and diffusion-weighted imaging in differentiating malignant from benign portal vein thrombus in cirrhotic patients.
AJR Am. J. Roentgenol. 2013,201, 1211–1219. [CrossRef]
116.
Sherman, C.B.; Behr, S.; Dodge, J.L.; Roberts, J.P.; Yao, F.Y.; Mehta, N. Distinguishing Tumor from Bland Portal Vein Thrombus
in Liver Transplant Candidates with Hepatocellular Carcinoma: The A-VENA Criteria. Liver Transplant.
2019
,25, 207–216.
[CrossRef]
117.
Fortea, J.I.; Zipprich, A.; Fernandez-Mena, C.; Puerto, M.; Bosoi, C.R.; Almagro, J.; Hollenbach, M.; Bañares, J.; Rodríguez-Sánchez,
B.; Cercenado, E.; et al. Enoxaparin does not ameliorate liver fibrosis or portal hypertension in rats with advanced cirrhosis. Liver
Int. 2018,38, 102–112. [CrossRef]
J. Clin. Med. 2022,11, 6435 24 of 26
118.
Amitrano, L.; Guardascione, M.A.; Menchise, A.; Martino, R.; Scaglione, M.; Giovine, S.; Romano, L.; Balzano, A. Safety and
efficacy of anticoagulation therapy with low molecular weight heparin for portal vein thrombosis in patients with liver cirrhosis.
J. Clin. Gastroenterol. 2010,44, 448–451. [CrossRef]
119.
Delgado, M.G.; Seijo, S.; Yepes, I.; Achecar, L.; Catalina, M.V.; Garcia-Criado, A.; Abraldes, J.G.; de la Peña, J.; Bañares, R.; Albillos,
A.; et al. Efficacy and safety of anticoagulation on patients with cirrhosis and portal vein thrombosis. Clin. Gastroenterol. Hepatol.
2012,10, 776–783. [CrossRef]
120.
Chung, J.W.; Kim, G.H.; Lee, J.H.; Ok, K.S.; Jang, E.S.; Jeong, S.H.; Kim, J.W. Safety, efficacy, and response predictors of
anticoagulation for the treatment of nonmalignant portal-vein thrombosis in patients with cirrhosis: A propensity score matching
analysis. Clin. Mol. Hepatol. 2014,20, 384–391. [CrossRef]
121.
Cui, S.B.; Shu, R.H.; Yan, S.P.; Wu, H.; Chen, Y.; Wang, L.; Zhu, Q. Efficacy and safety of anticoagulation therapy with different
doses of enoxaparin for portal vein thrombosis in cirrhotic patients with hepatitis B. Eur. J. Gastroenterol. Hepatol.
2015
,27, 914–919.
[CrossRef] [PubMed]
122.
Naeshiro, N.; Aikata, H.; Hyogo, H.; Kan, H.; Fujino, H.; Kobayashi, T.; Fukuhara, T.; Honda, Y.; Nakahara, T.; Ohno, A.; et al.
Efficacy and safety of the anticoagulant drug, danaparoid sodium, in the treatment of portal vein thrombosis in patients with
liver cirrhosis. Hepatol. Res. 2015,45, 656–662. [CrossRef] [PubMed]
123.
Noronha Ferreira, C.; Reis, D.; Cortez-Pinto, H.; Tato Marinho, R.; Goncalves, A.; Palma, S.; Leite, I.; Rodrigues, T.; Pedro, A.J.;
Alexandrino, P.; et al. Anticoagulation in Cirrhosis and Portal Vein Thrombosis Is Safe and Improves Prognosis in Advanced
Cirrhosis. Dig. Dis. Sci. 2019,64, 2671–2683. [CrossRef] [PubMed]
124.
Pettinari, I.; Vukotic, R.; Stefanescu, H.; Pecorelli, A.; Morelli, M.; Grigoras, C.; Sparchez, Z.; Andreone, P.; Piscaglia, F.; BO-LIVES;
et al. Clinical Impact and Safety of Anticoagulants for Portal Vein Thrombosis in Cirrhosis. Am. J. Gastroenterol.
2019
,114, 258–266.
[CrossRef] [PubMed]
125.
Bergere, M.; Erard-Poinsot, D.; Boillot, O.; Valette, P.J.; Guillaud, O.; Chambon-Augoyard, C.; Dumortier, J. Portal vein thrombosis
and liver cirrhosis: Long-term anticoagulation is effective and safe. Clin. Res. Hepatol. Gastroenterol.
2019
,43, 395–402. [CrossRef]
[PubMed]
126.
La Mura, V.; Braham, S.; Tosetti, G.; Branchi, F.; Bitto, N.; Moia, M.; Fracanzani, A.L.; Colombo, M.; Tripodi, A.; Primignani, M.;
et al. Harmful and Beneficial Effects of Anticoagulants in Patients With Cirrhosis and Portal Vein Thrombosis. Clin. Gastroenterol.
Hepatol. 2018,16, 1146–1152.e1144. [CrossRef] [PubMed]
127.
Qi, X.; De Stefano, V.; Li, H.; Dai, J.; Guo, X.; Fan, D. Anticoagulation for the treatment of portal vein thrombosis in liver cirrhosis:
A systematic review and meta-analysis of observational studies. Eur. J. Intern. Med. 2015,26, 23–29. [CrossRef]
128.
Ghazaleh, S.; Beran, A.; Aburayyan, K.; Nehme, C.; Patel, D.; Khader, Y.; Sharma, S.; Aziz, M.; Abdel-Aziz, Y.; Hammad, T.; et al.
Efficacy and safety of anticoagulation in non-malignant portal vein thrombosis in patients with liver cirrhosis: A systematic
review and meta-analysis. Ann. Gastroenterol. 2021,34, 104–110. [CrossRef]
129.
Wang, L.; Guo, X.; Xu, X.; De Stefano, V.; Plessier, A.; Noronha Ferreira, C.; Qi, X. Anticoagulation FavorsThrombus Recanalization
and Survival in Patients with Liver Cirrhosis and Portal Vein Thrombosis: Results of a Meta-Analysis. Adv. Ther.
2021
,38, 495–520.
[CrossRef]
130.
Rodriguez-Castro, K.I.; Vitale, A.; Fadin, M.; Shalaby, S.; Zerbinati, P.; Sartori, M.T.; Landi, S.; Pettinari, I.; Piscaglia, F.; Han, G.;
et al. A prediction model for successful anticoagulation in cirrhotic portal vein thrombosis. Eur. J. Gastroenterol. Hepatol.
2019
,31,
34–42. [CrossRef]
131.
Nicoară-Farcău, O.; Soy, G.; Magaz, M.; Baiges, A.; Turon, F.; Garcia-Criado, A.; Barrufet, M.; Burrel, M.; Hernández-Gea, V.;
García-Pagán, J.C. New Insights into the Pathogenesis, Risk Factors, and Treatment of Portal Vein Thrombosis in Patients with
Cirrhosis. Semin. Thromb. Hemost. 2020,46, 673–681. [CrossRef] [PubMed]
132.
Rodríguez-Castro, K.I.; Senzolo, M.; Sartori, M.T. Optimal length of anticoagulant therapy in cirrhotic patients with portal vein
thrombosis. Clin. Gastroenterol. Hepatol. 2012,10, 820–821. [CrossRef] [PubMed]
133.
Guerrero, A.; Campo, L.; Piscaglia, F.; Reiberger, T.; Han, G.; Violi, F.E.A. Anticoagulation improves overall survival through
portal vein recanalization in patients with cirrhosis and portal vein thrombosis: Individual patient data meta-analysis (IMPORTAL
study). J. Hepatol. 2021,75, S201–S293.
134.
Cerini, F.; Gonzalez, J.M.; Torres, F.; Puente, A.; Casas, M.; Vinaixa, C.; Berenguer, M.; Ardevol, A.; Augustin, S.; Llop, E.; et al.
Impact of anticoagulation on upper-gastrointestinal bleeding in cirrhosis. A retrospective multicenter study. Hepatology
2015
,62,
575–583. [CrossRef] [PubMed]
135.
Bechmann, L.P.; Sichau, M.; Wichert, M.; Gerken, G.; Kröger, K.; Hilgard, P. Low-molecular-weight heparin in patients with
advanced cirrhosis. Liver Int. 2011,31, 75–82. [CrossRef] [PubMed]
136.
Senzolo, M.; Rodriguez-Castro, K.I.; Rossetto, V.; Radu, C.; Gavasso, S.; Carraro, P.; Zerbinati, P.; Sartori, M.T.; Simioni, P. Increased
anticoagulant response to low-molecular-weight heparin in plasma from patients with advanced cirrhosis. J. Thromb. Haemost.
2012,10, 1823–1829. [CrossRef]
137.
Zhang, Y.; Zhang, M.; Tan, L.; Pan, N.; Zhang, L. The clinical use of Fondaparinux: A synthetic heparin pentasaccharide. Prog.
Mol. Biol. Transl. Sci. 2019,163, 41–53.
138.
Lobo, B.; Finch, C.; Howard, A.; Minhas, S. Fondaparinux for the treatment of patients with acute heparin-induced thrombocy-
topenia. Thromb. Haemost. 2008,99, 208–214.
J. Clin. Med. 2022,11, 6435 25 of 26
139.
Senzolo, M.; Piano, S.; Shalaby, S.; Tonon, M.; Tonello, S.; Zanetto, A.; Sacerdoti, D.; Simioni, P.; Bombonato, G.; Burra, P.; et al.
Comparison of Fondaparinux and Low-Molecular-Weight Heparin in the Treatment of Portal Vein Thrombosis in Cirrhosis. Am.
J. Med. 2021,134, 1278–1285.e2. [CrossRef]
140.
Biolato, M.; Paratore, M.; Di Gialleonardo, L.; Marrone, G.; Grieco, A. Direct oral anticoagulant administration in cirrhotic patients
with portal vein thrombosis: What is the evidence? World J. Hepatol. 2022,14, 682–695. [CrossRef]
141.
Potze, W.; Arshad, F.; Adelmeijer, J.; Blokzijl, H.; van den Berg, A.P.; Meijers, J.C.; Porte, R.J.; Lisman, T. Differential
in vitro
inhibition of thrombin generation by anticoagulant drugs in plasma from patients with cirrhosis. PLoS ONE
2014
,9, e88390.
[CrossRef] [PubMed]
142.
Potze, W.; Adelmeijer, J.; Lisman, T. Decreased
in vitro
anticoagulant potency of Rivaroxaban and Apixaban in plasma from
patients with cirrhosis. Hepatology 2015,61, 1435–1436. [CrossRef] [PubMed]
143.
Serper, M.; Weinberg, E.M.; Cohen, J.B.; Reese, P.P.; Taddei, T.H.; Kaplan, D.E. Mortality and Hepatic Decompensation in Patients
with Cirrhosis and Atrial Fibrillation Treated with Anticoagulation. Hepatology 2021,73, 219–232. [CrossRef] [PubMed]
144.
De Gottardi, A.; Trebicka, J.; Klinger, C.; Plessier, A.; Seijo, S.; Terziroli, B.; Magenta, L.; Semela, D.; Buscarini, E.; Langlet, P.; et al.
Antithrombotic treatment with direct-acting oral anticoagulants in patients with splanchnic vein thrombosis and cirrhosis. Liver
Int. 2017,37, 694–699. [CrossRef]
145.
Intagliata, N.M.; Henry, Z.H.; Maitland, H.; Shah, N.L.; Argo, C.K.; Northup, P.G.; Caldwell, S.H. Direct Oral Anticoagulants
in Cirrhosis Patients Pose Similar Risks of Bleeding When Compared to Traditional Anticoagulation. Dig. Dis. Sci.
2016
,61,
1721–1727. [CrossRef]
146.
Hum, J.; Shatzel, J.J.; Jou, J.H.; Deloughery, T.G. The efficacy and safety of direct oral anticoagulants vs traditional anticoagulants
in cirrhosis. Eur. J. Haematol. 2017,98, 393–397. [CrossRef]
147.
Semmler, G.; Pomej, K.; Bauer, D.J.M.; Balcar, L.; Simbrunner, B.; Binter, T.; Hartl, L.; Becker, J.; Pinter, M.; Quehenberger, P.; et al.
Safety of direct oral anticoagulants in patients with advanced liver disease. Liver Int. 2021,41, 2159–2170. [CrossRef]
148.
Nisly, S.A.; Mihm, A.E.; Gillette, C.; Davis, K.A.; Tillett, J. Safety of direct oral anticoagulants in patients with mild to moderate
cirrhosis: A systematic review and meta-analysis. J. Thromb. Thrombolysis 2021,52, 817–827. [CrossRef]
149.
Menichelli, D.; Ronca, V.; Di Rocco, A.; Pignatelli, P.; Marco Podda, G. Direct oral anticoagulants and advanced liver disease: A
systematic review and meta-analysis. Eur. J. Clin. Investig. 2021,51, e13397. [CrossRef]
150.
Hanafy, A.S.; Abd-Elsalam, S.; Dawoud, M.M. Randomized controlled trial of rivaroxaban versus warfarin in the management of
acute non-neoplastic portal vein thrombosis. Vasc. Pharmacol. 2019,113, 86–91. [CrossRef]
151.
Nagaoki, Y.; Aikata, H.; Daijyo, K.; Teraoka, Y.; Shinohara, F.; Nakamura, Y.; Hatooka, M.; Morio, K.; Nakahara, T.; Kawaoka, T.;
et al. Efficacy and safety of edoxaban for treatment of portal vein thrombosis following danaparoid sodium in patients with liver
cirrhosis. Hepatol. Res. 2018,48, 51–58. [CrossRef] [PubMed]
152.
Ai, M.H.; Dong, W.G.; Tan, X.P.; Xu, L.; Xu, C.; Zhang, Q.; Zhang, Y.; Li, J. Efficacy and safety study of direct-acting oral
anticoagulants for the treatment of chronic portal vein thrombosis in patients with liver cirrhosis. Eur. J. Gastroenterol. Hepatol.
2020,32, 1395–1400. [PubMed]
153.
Radosevich, P.M.; Ring, E.J.; LaBerge, J.M.; Peltzer, M.Y.; Haskal, Z.J.; Doherty, M.M.; Gordon, R.L. Transjugular intrahepatic
portosystemic shunts in patients with portal vein occlusion. Radiology 1993,186, 523–527. [CrossRef] [PubMed]
154.
Walser, E.M.; NcNees, S.W.; DeLa Pena, O.; Crow, W.N.; Morgan, R.A.; Soloway, R.; Broughan, T. Portal venous thrombosis:
Percutaneous therapy and outcome. J. Vasc. Interv. Radiol. 1998,9, 119–127. [CrossRef]
155.
Stein, M.; Link, D.P. Symptomatic spleno-mesenteric-portal venous thrombosis: Recanalization and reconstruction with endovas-
cular stents. J. Vasc. Interv. Radiol. 1999,10, 363–371. [CrossRef]
156.
Rodrigues, S.G.; Sixt, S.; Abraldes, J.G.; De Gottardi, A.; Klinger, C.; Bosch, J.; Baumgartner, I.; Berzigotti, A. Systematic review
with meta-analysis: Portal vein recanalisation and transjugular intrahepatic portosystemic shunt for portal vein thrombosis.
Aliment Pharmacol. Ther. 2019,49, 20–30. [CrossRef] [PubMed]
157.
Valentin, N.; Korrapati, P.; Constantino, J.; Young, A.; Weisberg, I. The role of transjugular intrahepatic portosystemic shunt in the
management of portal vein thrombosis: A systematic review and meta-analysis. Eur. J. Gastroenterol. Hepatol.
2018
,30, 1187–1193.
[CrossRef]
158.
Lv, Y.; Bai, W.; Li, K.; Wang, Z.; Guo, W.; Luo, B.; Wang, J.; Wang, Q.; Wang, E.; Xia, D.; et al. Anticoagulation and Transjugular
Intrahepatic Portosystemic Shunt for the Management of Portal Vein Thrombosis in Cirrhosis: A Prospective Observational Study.
Am. J. Gastroenterol. 2021,116, 1447–1464. [CrossRef]
159.
Zhan, C.; Prabhu, V.; Kang, S.K.; Li, C.; Zhu, Y.; Kim, S.; Olsen, S.; Jacobson, I.M.; Dagher, N.N.; Carney, B.; et al. Comparison of
Non-Tumoral Portal Vein Thrombosis Management in Cirrhotic Patients: TIPS Versus Anticoagulation Versus No Treatment. J.
Clin. Med. 2021,10, 2316. [CrossRef]
160.
Lv, Y.; Qi, X.; He, C.; Wang, Z.; Yin, Z.; Niu, J.; Guo, W.; Bai, W.; Zhang, H.; Xie, H.; et al. Covered TIPS versus endoscopic band
ligation plus propranolol for the prevention of variceal rebleeding in cirrhotic patients with portal vein thrombosis: A randomised
controlled trial. Gut 2018,67, 2156–2168. [CrossRef]
161.
Luo, X.; Wang, Z.; Tsauo, J.; Zhou, B.; Zhang, H.; Li, X. Advanced Cirrhosis Combined with Portal Vein Thrombosis: A
Randomized Trial of TIPS versus Endoscopic Band Ligation Plus Propranolol for the Prevention of Recurrent Esophageal Variceal
Bleeding. Radiology 2015,276, 286–293. [CrossRef] [PubMed]
J. Clin. Med. 2022,11, 6435 26 of 26
162.
Wang, Z.; Jiang, M.S.; Zhang, H.L.; Weng, N.N.; Luo, X.F.; Li, X.; Yang, L. Is Post-TIPS Anticoagulation Therapy Necessary in
Patients with Cirrhosis and Portal Vein Thrombosis? A Randomized Controlled Trial. Radiology
2016
,279, 943–951. [CrossRef]
[PubMed]
163.
Han, G.; Qi, X.; He, C.; Yin, Z.; Wang, J.; Xia, J.; Yang, Z.; Bai, M.; Meng, X.; Niu, J.; et al. Transjugular intrahepatic portosystemic
shunt for portal vein thrombosis with symptomatic portal hypertension in liver cirrhosis. J. Hepatol.
2011
,54, 78–88. [CrossRef]
[PubMed]
164.
Habib, A.; Desai, K.; Hickey, R.; Thornburg, B.; Vouche, M.; Vogelzang, R.L.; Salem, R. Portal vein recanalization-
transjugularintrahepatic portosystemic shunt using the transsplenic approach to achieve transplant candidacy in patients with
chronic portal vein thrombosis. J. Vasc. Interv. Radiol. 2015,26, 499–506. [CrossRef] [PubMed]
165.
Thornburg, B.; Desai, K.; Hickey, R.; Hohlastos, E.; Kulik, L.; Ganger, D.; Baker, T.; Abecassis, M.; Caicedo, J.C.; Ladner, D.; et al.
Pretransplantation Portal Vein Recanalization and Transjugular Intrahepatic Portosystemic Shunt Creation for Chronic Portal
Vein Thrombosis: Final Analysis of a 61-Patient Cohort. J. Vasc. Interv. Radiol. 2017,28, 1714–1721.e2. [CrossRef]
166.
Salem, R.; Vouche, M.; Baker, T.; Herrero, J.I.; Caicedo, J.C.; Fryer, J.; Hickey, R.; Habib, A.; Abecassis, M.; Koller, F.; et al.
Pretransplant Portal Vein Recanalization-Transjugular Intrahepatic Portosystemic Shunt in Patients with Complete Obliterative
Portal Vein Thrombosis. Transplantation 2015,99, 2347–2355. [CrossRef]
167.
Thornburg, B.; Desai, K.; Hickey, R.; Kulik, L.; Ganger, D.; Baker, T.; Abecassis, M.; Lewandowski, R.J.; Salem, R. Portal Vein
Recanalization and Transjugular Intrahepatic Portosystemic Shunt Creation for Chronic Portal Vein Thrombosis: Technical
Considerations. Tech. Vasc. Interv. Radiol. 2016,19, 52–60. [CrossRef] [PubMed]
168.
Boike, J.R.; Thornburg, B.G.; Asrani, S.K.; Fallon, M.B.; Fortune, B.E.; Izzy, M.J.; Verna, E.C.; Abraldes, J.G.; Allegretti, A.S.;
Bajaj, J.S.; et al. North American Practice-Based Recommendations for Transjugular Intrahepatic Portosystemic Shunts in Portal
Hypertension. Clin. Gastroenterol. Hepatol. 2022,20, 1636–1662.e1636. [CrossRef]
169.
Hollingshead, M.; Burke, C.T.; Mauro, M.A.; Weeks, S.M.; Dixon, R.G.; Jaques, P.F. Transcatheter thrombolytic therapy for acute
mesenteric and portal vein thrombosis. J. Vasc. Interv. Radiol. 2005,16, 651–661. [CrossRef]
170.
Liu, K.; Li, W.D.; Du, X.L.; Li, C.L.; Li, X.Q. Comparison of Systemic Thrombolysis Versus Indirect Thrombolysis via the Superior
Mesenteric Artery in Patients with Acute Portal Vein Thrombosis. Ann. Vasc. Surg. 2017,39, 264–269. [CrossRef]
171.
Smalberg, J.H.; Spaander, M.V.; Jie, K.S.; Pattynama, P.M.; van Buuren, H.R.; van den Berg, B.; Janssen, H.L.A.; Leebeek, F.W.G.
Risks and benefits of transcatheter thrombolytic therapy in patients with splanchnic venous thrombosis. Thromb. Haemost.
2008
,
100, 1084–1088. [CrossRef] [PubMed]
172.
Bianchini, M.; Cavani, G.; Bonaccorso, A.; Turco, L.; Vizzutti, F.; Sartini, A.; Gitto, S.; Merighi, A.; Banchelli, F.; Villa, E.; et al. Low
molecular weight heparin does not increase bleeding and mortality post-endoscopic variceal band ligation in cirrhotic patients.
Liver Int. 2018,38, 1253–1262. [CrossRef] [PubMed]
173.
Guillaume, M.; Christol, C.; Plessier, A.; Corbic, M.; Péron, J.-M.; Sommet, A.; Rautou, P.-E.; Consigny, Y.; Vinel, J.-P.; Valla, C.-D.;
et al. Bleeding risk of variceal band ligation in extrahepatic portal vein obstruction is not increased by oral anticoagulation. Eur. J.
Gastroenterol. Hepatol. 2018,30, 563–568. [CrossRef] [PubMed]
174.
Montalvá, E.; Rodríguez-Perálvarez, M.; Blasi, A.; Bonanad, S.; Gavín, O.; Hierro, L.; Lladó, L.; Llop, E.; Pozo-Laderas, J.C.;
Colmenero, J.; et al. Consensus Statement on Hemostatic Management, Anticoagulation, and Antiplatelet Therapy in Liver
Transplantation. Transplantation 2022,106, 1123–1131. [CrossRef]
... Период артериальной вазодилатации сменяется фазой «венозного спасения» с формированием венозных портопортальных коллатералей, обходящих окклюзированный сегмент, и образованием через 3-5 недель «каверномы» -отличительного признака хронического тромбоза [17,20]. ...
... УЗИ при ТВВ является методом скрининга [1,4]. Обладает чувствительностью 73-93 %, специфичностью 99 %, положительной прогностической ценностью 86-97 %, отрицательной предсказательной ценностью 98 %, что сопоставимо с ангиографией и компьютерной томографией (90 %, 99 %, 95 % и 97 % соответственно) [20,60], выявляя гиперэхогенный сигнал в просвете сосуда, расширение воротной вены, отсутствие/снижение кровотока в части или во всем просвете вены, определяет скорость и направление тока крови. Преимущества ультразвуковой допплерографии включают низкую стоимость, доступность, отсутствие облучения. ...
... Однако этот метод зависит от опыта оператора, обладает меньшей надежностью при метеоризме, ожирении, частичной окклюзии воротной вены и при распространении тромба на селезеночную и верхнюю брыжеечную вены. По данным УЗИ, трудно отличить мягкие тромбы от злокачественной инвазии воротной вены [17,20], поэтому после УЗИ рекомендовано проводить визуализационное исследование с контрастным усилением, в том числе для исключения гепатоцеллюлярной карциномы [1][2][3][4]. ...
Article
Portal vein thrombosis is the most common thrombotic complication in patients with liver cirrhosis, especially in cases of severe forms. The pathogenesis is multifactorial in nature, it determined by a change in the balance between the coagulation and anticoagulation systems. Thrombosis is often asymptomatic and is accidentally detected, although it can be complicated by varicose bleeding, intestinal ischemia, and portal biliopathy. Ultrasound Doppler examination is a screening method, as an alternative, computed tomography and magnetic resonance imaging are used. The review highlights data on epidemiology, risk factors, clinical features, and diagnosis of portal vein thrombosis in patients with liver cirrhosis. The data on the effect of portal vein thrombosis on the progression of liver cirrhosis and the survival of patients, including after liver transplantation, are presented.
... A decrease in portal FV to less than 15 cm/s measured by Doppler ultrasonography (US) is a significant predictive factor for the development of PVT 6,14 . Endothelial dysfunction is commonly present in LC patients and is associated with procedures such as sclerotherapy, portosystemic shunt surgery, and splenectomy 15 . A rebalanced coagulation concept, a delicate balance between procoagulant and anticoagulant factors, characterizes LC. ...
Article
Full-text available
Background/Aim. Portal vein thrombosis (PVT) in patients with liver cirrhosis (LC) has a prevalence of 0.6- 26%. It is most commonly discovered incidentally as part of the evaluation of LC or in the context of acute decompensation of LC due to portal hypertension. The aim of the study was to determine the prevalence of PVT in patients with LC in relation to the severity of the disease and individual elements of portal hypertension. Methods. A total of 326 patients treated for LC decompensation were included in a retrospective study. Standard laboratory analyses, abdominal ultrasonography and/or computed tomography, and esophagogastroduodenoscopy were performed. Results. The diameter of the portal vein (PV) differed between patients without esophageal varices (12.2 mm) and those with large varices (13.6 mm), p = 0.026. PVT was identified in 6.1% of patients with LC. The patients were classified according to the Child-Pugh scoring system, which has the A, B, and C categories used to assess the severity of liver disease. PVT was present in 3.0% of patients in class C and 12.0% in class B, while none of the patients in class A had PVT (p = 0.005). PVT was present in 4.4% of patients with small varices and 16.7% with large varices (p < 0.001). There was no difference in the presence of PVT between the groups of patients with and without variceal bleeding nor between groups with different degrees of ascites. A fatal outcome occurred in 29.4% of patients, but there was no difference between patients with and without PVT. Conclusion. PVT is present in more advanced stages of LC and predominantly in patients with large esophageal varices. There was no higher prevalence of PVT observed with the occurrence of variceal bleeding or with the death outcome in patients with LC.
... Вопрос о роли врожденных тромбофилий (мутация гена протромбина G20210A и мутации фактора V Лейден) в развитии ТВВ у пациентов с циррозом остается предметом дискуссии. Несмотря на то что в нескольких метаанализах показана ассоциация этих тромбофилий с риском ТВВ при циррозе, все эти работы имели смещенные результаты из-за качества включенных исследований [28]. На настоящий момент нет рекомендаций по необходимости скрининга на наследственные тромбофилии всех пациентов с циррозом и ТВВ [29]. ...
Article
Aim : to evaluate the frequency of portal vein thrombosis (PVT) and build predictive models of the development of PVT for patients with liver cirrhosis (LC) of A and B/C classes by Child-Pugh. Materials and methods . Research design is a case-control. The Case group included 130 patients with newly diagnosed PVT not caused by invasive hepatocellular carcinoma (HCC); 29 patients were assigned to class A, 101 patients were assigned to class B/C. From the database of cirrhotic patients without PVT 60 Controls for class A and 205 for B/C were selected using sratified randomization by sex, age and etiology of cirrhosis. The Mann-Whitney U -test and Pearson's chi-squared test were used to compare the groups. Odds ratios (OR) and 95 % confidence intervals (95 % CI) were calculated. Logistic regression models are constructed with the separation of the sample into training and test (0.7; 0.3). The operational characteristics of the models were calculated on the test sample; ROC analysis was carried out, the area under the ROC curve (AUC) was calculated. Results . The overall frequency of PVT was 4.1 % (95 % CI 2.7-5.8 %) in class A and 10.4 % (95 % CI 8.5-12.5 %) class B/C. Patients with class A and B/C PVT differed from the corresponding controls by more severe portal hypertension: the frequency of bleeding / number of interventions on varices compared with the control were 41/45 % vs. 7/8 % ( p < 0.001) for class A and 25.7/30.7 % vs. 16.1/16.1 % ( p < 0.05) for class B/C, ascites frequency was 24 % vs. 8 % ( p < 0.05) for class A and 89.1 % vs. 68.3 % ( p < 0.001) for class B/C. The cutoff by the portal vein diameter was the same for both classes — 13.4 mm; the spleen length was similar and amounted 17.5 mm for class A, 17.1 mm for class B/C. Patients with PVT differed from the corresponding controls by neutrophil-to-lymphocyte ratio: class A 2.33 (1.82; 3.61) vs. 1.76 (1.37; 2.20), p < 0.01, class B/C 2.49 (1.93; 3.34) vs. 2.15 (1.49; 3.26), p < 0.05. Patients of class B/C had a higher incidence of newly diagnosed malignant tumors - 23.8% (primarily HCC that does not invade the portal vein), compared with control and cases of class A - 6.3 % and 3 % ( p < 0.05), respectively. The best model for class A included variceal bleeding, ascites, portal vein diameter, absolute number of neutrophils, for class B — ascites, spleen length, portal vein diameter, malignant tumors / local factors; sensitivity, specificity, accuracy and AUC were 79.3 %, 90 %, 86.5 %, 0.897 and 73.3 %, 68.3 %, 69.9 %, 0.789, respectively. Conclusion . Independently of the Child-Pugh class of LC, the main risk factor for PVT is severe portal hypertension.
... American gastroenterological association clinical practice update and meta-analysis of direct oral anticoagulants (rivaroxaban, etc.) versus traditional anticoagulants have shown the advantages of direct oral anticoagulants, which are not only easy to use, but also have no difference in safety and bleeding risk compared with traditional anticoagulants. [29][30][31] However, few trials of direct oral anticoagulants have been conducted in liver transplant patients with end-stage liver disease. Future studies with large samples are needed to analyze the clinical anticoagulation efficacy of direct oral anticoagulants. ...
Article
Full-text available
In recent years, the association between portal vein thrombosis and liver transplantation has extensive attention from physicians worldwide. However, there is no available literature on bibliometric analysis in this research area. Herein, we aimed to conduct a bibliometric analysis to identify the hotspots and frontiers of research related to portal vein thrombosis and liver transplantation. Documents published between 2002 and 2022 were retrieved and downloaded from the Web of Science Core Collection database. VOSviewer was utilized to generate a visualization network map of authors, nations, institutions, journals, and keyword co-occurrence/clustering. Additionaly, CiteSpace was used to analyze the keywords with the strongest bursts. A total of 1272 articles and reviews were extracted from the database. The author Marco Senzolo published the largest number of papers. The United States was the most prolific country, and Hope-Bochon (France) was the top productive institution. Liver Transplantation was the most prolific journal in the field. The most commonly identified keywords in the study were cirrhosis, risk factors, portal vein thrombosis, and management, as revealed by the keyword co-occurrence analysis. It is suggested that patients with cirrhosis, portal vein thrombosis prevention, and management measures for portal vein thrombosis have been prominet topics in recent years. Furthermore, an analysis of keywords with the strongest citation bursts highlighted pediatric liver transplantation, direct oral anticoagulants, and nonalcoholic fatty liver disease as current research trends. Research in portal vein thrombosis and liver transplantation exhibits a general upward trend. The latest hot topics within this area of study involve pediatric patients and nonalcoholic fatty liver disease.
... However, SVT in cirrhotic patients is frequently asymptomatic, due to the progressive decrease in the portal vein flow velocity and the generation of collateral circulation supporting the venous flow [33]. Therefore, it could be detected incidentally in abdominal imaging performed for follow-up of cirrhosis or other diseases [34]. ...
Article
Introduction: Liver cirrhosis is accompanied by several haemostatic alterations, which contribute to the current theory of "rebalanced haemostasis". Splanchnic vein thrombosis (SVT) is a frequent complication of liver cirrhosis (17-26% of cirrhotic patients) and liver cirrhosis is a common risk factor for SVT (24-28% of SVT cases). Areas covered: This narrative review aims to describe the current state of the art on the anticoagulant treatment of cirrhotic SVT, with a particular focus on the possible role of the direct oral anticoagulants (DOACs) and recent guidelines on this topic. Expert opinion: Early anticoagulant therapy is recommended in cirrhotic patients with acute SVT, to obtain vessel recanalization and decrease the rates of portal hypertension-related complications. Gastroesophageal varices do not represent a contraindication to anticoagulation, if adequate prophylaxis of variceal bleeding is established, and varices band ligation can be safely performed without the need to stop the anticoagulant treatment. The conventional treatment of cirrhotic SVT consisted in low molecular weight heparin, as initial treatment of choice, eventually followed by vitamin K antagonists, but the DOACs can be considered as a reasonable alternative in patients with compensated liver cirrhosis.
... The last paper of this Special Issue is a review made by our group on portal vein thrombosis (PVT) in patients with cirrhosis [29]. PVT is the most common thrombotic event in this population, with increased rates in the setting of advanced liver disease. ...
Article
Full-text available
Cirrhosis is the result of sustained liver damage leading to the diffusion of hepatic fibrosis, wherein the normal hepatic architecture is replaced by abnormally organized nodules separated by fibrous septa that connect the different vascular structures of the hepatic lobule [...]
Article
In recent years, the incidence of portal vein thrombosis (PVT) in patients with cirrhosis has increased; the thrombosis ranges from asymptomatic partial to complete occlusion. Treatment is difficult. Anticoagulation therapy may be the optimal first-line treatment for patients with acute PVT who lack variceal bleeding or mesenteric ischemia. Minimally invasive treatment options include mechanical thrombectomy, chemical thrombolysis, and stent placement. However, the effectiveness and timing of anticoagulation and interventional therapies remain unclear.
Article
Full-text available
Background: A hypercoagulable state is not associated with development of portal vein thrombosis in cirrhosis, as we previously demonstrated. However, some groups demonstrated elevated levels of inflammatory markers and activation of haemostasis in the portal vein (PV) compared to post-hepatic veins, but as the liver is involved in clearance of these markers we hypothesize that interpretation of these data is not straightforward. Aim: To determine whether the PV has particular proinflammatory/hypercoagulable characteristics by comparing plasma sampled in the PV, hepatic vein (HV) and the systemic circulation. Methods: Plasma samples from 51 cirrhotic patients with portal hypertension undergoing transjugular intrahepatic portosystemic shunt placement, were taken from the PV, HV, and jugular vein (JV). Markers of inflammation (LPS, TNF-α, IL-6, TBARS), neutrophil-extracellular-traps (cfDNA, MPO-DNA), endothelial damage (VWF) and haemostasis were determined and compared between the three vascular beds. Results: Markers of inflammation were slightly, but significantly higher in the PV than in the HV and systemic circulation. VWF and markers of haemostasis were modestly elevated in the PV. Levels of multiple markers were lower in the HV compared to the PV and systemic circulation. Higher MELD score was associated with a more prothrombotic state in all three sample sites. Conclusion: In contrast to published studies, we did not detect a clear proinflammatory or prothrombotic environment in the PV of cirrhotic patients. Many markers are lowest in the HV, indicating that the low levels of these markers in the HV, at least in part, reflect clearance of those markers in the liver.
Article
Full-text available
In recent years, the traditional concept that cirrhosis-related coagulopathy is an acquired bleeding disorder has evolved. Currently, it is known that in cirrhotic patients, the hemostatic system is rebalanced, which involves coagulation factors, fibrinolysis and platelets. These alterations disrupt homeostasis, skewing it toward a procoagulant state, which can lead to thromboembolic manifestations, especially when hemodynamic and endothelial factors co-occur, such as in the portal vein system in cirrhosis. Portal vein thrombosis is a common complication of advanced liver cirrhosis that negatively affects the course of liver disease, prognosis of cirrhotic patients and success of liver transplantation. It is still debated whether portal vein thrombosis is the cause or the consequence of worsening liver function. Anticoagulant therapy is the mainstay treatment for acute symptomatic portal vein thrombosis. In chronic portal vein thrombosis, the role of anticoagulant therapy is still unclear. Traditional anticoagulants, vitamin K antagonists and low-molecular-weight heparin are standard-of-care treatments for portal vein thrombosis. In the last ten years, direct oral anticoagulants have been approved for the prophylaxis and treatment of many thromboembolic-related diseases, but evidence on their use in cirrhotic patients is very limited. The aim of this review was to summarize the evidence about the safety and effectiveness of direct oral anticoagulants for treating portal vein thrombosis in cirrhotic patients.
Article
Background and aims Alteration of platelet status associates with decompensation and death in cirrhosis, while its effect on portal vein thrombosis (PVT) remains unclear. We aimed to retrospectively investigate whether PVT associates with platelet-fibrin clot strength and platelet activation in decompensated cirrhosis. Methods Platelet-fibrin clot strength (G) was measured by thromboelastography (TEG). Platelet activation was reflected by plasma concentrations of soluble p-selectin (sPs) and a platelet aggregation test adjusted for platelet counts. Results Among 166 patients, 45 had PVT. The platelet count was significantly lower in PVT. While the G value was positively correlated with platelet count (ρ = 0.74, P < 0.01), increased G was associated with PVT after adjusting for platelet count in the logistic regression (P = 0.04). The normalized G value according to the linear relation with platelet count was calculated as follows: Gplatelet = [(G - 2622)/platelet count]. This coefficient had no correlation with platelet count and was an independent risk factor of PVT (OR = 1.03, CI95%: 1.01-1.05, P = 0.012). In two subanalyses, the collagen-induced platelet aggregation (n = 37, P = 0.029) and plasma concentration of sPs (n = 56, P = 0.001) adjusted for platelet count were significantly higher in PVT. Conclusion This study showed a positive correlation of high platelet-fibrin clot strength detected via TEG and platelet activation with PVT in decompensated cirrhosis.
Article
Patients with decompensated cirrhosis are at risk of portal vein thrombosis (PVT). We prospectively investigated whether alterations of platelet aggregation can predict PVT in decompensated cirrhosis. At baseline, all patients underwent whole blood aggregometry (Multiplate®) to assess ADP‐induced platelet aggregation. Aggregometry results were expressed as ratio between platelet aggregation and platelet count (PLT ratio). Then, patients with cirrhosis were prospectively followed for 1 year for PVT development. One‐hundred and twenty‐eight patients with decompensated cirrhosis were included (Child‐Pugh A/B/C 12/39/49%). Cumulative incidence of PVT was 14%. On multivariate analysis, PLT ratio (OR 4.5, 95%CI 2.63‐7.67; p<0.0001) and Child‐Pugh C vs. A/B (OR 4.1, 95%CI 1.18‐14.80; p=0.03) were independently associated with PVT. Discriminative ability of PLT ratio was higher than Child‐Pugh (AUC 0.92 vs 0.70, p<0.0001). A PLT ratio >0.75 had 83% sensitivity and 84% specificity for PVT. In conclusion, PLT ratio by Multiplate® seems a promising thrombotic biomarker in decompensated cirrhosis.
Article
Background & Aims Hypercoagulability and hypofibrinolysis in acutely decompensated cirrhosis (AD) may be implicated in disease progression and hemostatic complications. We conducted a prospective study to: [1] characterize hemostatic alterations in AD; [2] evaluate whether such alterations can predict ACLF and bleeding/thrombosis. Methods Hospitalized patients with AD were prospectively recruited and underwent an extensive hemostatic profiling including coagulation factors, thrombomodulin-modified thrombin generation assay with evaluation of endogenous thrombin potential (ETP; marker for plasmatic hypercoagulability), fibrinolytic factors, and plasmin-antiplasmin complex (fibrinolysis activation marker). Inflammation severity was assessed by C-reactive protein (CRP). In study part 1, we compared hemostasis in AD vs. controls (stable decompensated and compensated cirrhosis). In study part 2, we prospectively followed patients with AD for 1 year and investigated predictors of ACLF and bleeding/thrombosis. Results 169 AD patients were recruited (median MELD 20; CLIF-C AD 54). Compared to controls, AD was associated with more pronounced hypercoagulability (ETP: 871 vs. 750 vs. 605 nmol/L*min; p<0.0001), without differences in fibrinolysis activation. During follow-up, 55 patients developed ACLF. CLIF-C AD, CRP, and Child-Pugh were independently associated with ACLF. A predictive model combining these variables (Padua model) accurately identified patients at higher risk of ACLF (AUROC 0.857; 95% CI; 0.798-0.915; sensitivity 74.5%, specificity 83.3%). Notably, CRP and progression to ACLF, but not baseline coagulopathy, were associated with bleeding (n=11); CRP and antifibrinolytic factor PAI-1 >50 ng/mL were associated with thrombosis (n=14). The prognostic value of the Padua model was validated in an independent, bicentric European cohort (n=301). Conclusion Inflammation severity, and not coagulopathy, is the most important predictor of ACLF and bleeding in AD. The Padua model can be used to identify AD patients at risk of ACLF. Trial registration number NA.
Article
Background and aims In cirrhosis, decreased portal flow velocity, thrombophilia factors, and portal hypertension are considered risk factors for portal vein thrombosis (PVT). In cirrhosis, the transformation of the stellate cells causes a progressive decrease of ADAMTS-13, while VWF multimers secretion by endothelial cells is strongly enhanced. This imbalance leads to an accumulation of ultra-large VWF multimers that in sinusoidal circulation could favor PVT both in intra- and extra-hepatic branches, mostly in decompensated cirrhosis. This prospective study was aimed at identifying possible clinical, biochemical, and hemostatic factors predictive for non-tumoral PVT in a cohort of patients with compensated cirrhosis. Methods Seventynine compensated cirrhosis patients were prospectively followed for 48 months, receiving a periodic Doppler-ultrasound liver examination associated with an extensive evaluation of clinical, biochemical, and hemostatic profile. Results Five patients developed PVT (cumulative prevalence = 6.3%), occurring 4–36 months after enrollment. In logistic regression analysis, the ADAMTS-13/VWF:GpIbR ratio < 0.4 was the only independent variable significantly associated with PVT (OR 14.6, 95% C.I.:1.36–157.2, p = 0.027). A Cox-regression-analysis confirmed this finding (HR = 7.7, p = 0.027). Conclusions The ADAMTS-13/VWF ratio < 0.4 measured in compensated cirrhosis could be a reliable predictive biomarker for PVT development, paving the way to novel therapeutic strategies to prevent and treat PVT in this clinical setting.
Article
Background Portal vein thrombosis (PVT) may be associated with negative outcomes in patients with liver cirrhosis. However, the prevalence and incidence of PVT in liver cirrhosis are heterogeneous among studies and have not been sufficiently determined yet. Methods The PubMed, EMBASE, and Cochrane Library databases were searched. Eligible studies would explore the prevalence and/or incidence of PVT in liver cirrhosis without hepatocellular carcinoma or abdominal surgery. Pooled proportion with 95% confidence interval (CI) was calculated using a random-effect model. Factors associated with the presence/occurrence of PVT were also extracted. Results Among the 8549 papers initially identified, 74 were included. Fifty-four studies explored the prevalence of PVT in liver cirrhosis with a pooled prevalence of 13.92% (95%CI=11.18–16.91%). Based on cross-sectional data, Child-Pugh class B/C, higher D-dimer, ascites, and use of non-selective beta-blockers (NSBBs) were associated with the presence of PVT in liver cirrhosis. Twenty-three studies explored the incidence of PVT in liver cirrhosis with a pooled incidence of 10.42% (95%CI=8.16–12.92%). Based on cohort data, Child-Pugh class B/C, higher model of end-stage liver disease score, higher D-dimer, lower platelets count, decreased portal flow velocity, ascites, use of NSBBs, and moderate or high-risk esophageal varices could predict the occurrence of PVT in liver cirrhosis. Conclusion Approximately one seventh of cirrhotic patients have PVT, and one tenth will develop PVT. Progression of liver cirrhosis and portal hypertension seems to be in parallel with the risk of PVT. Prospective studies with detailed information about classification and extension of PVT in liver cirrhosis are needed.
Article
Background & Aims Studies on platelet aggregation in cirrhosis are controversial because interpretation of platelet function is challenged by thrombocytopenia. We conducted a prospective study to investigate whole blood platelet aggregation in cirrhosis and its association with liver-related outcomes. Methods Platelet aggregation was assessed by whole blood aggregometry (Multiplate®). To overcome the influence of platelet count and compare cirrhosis with thrombocytopenia versus controls with normal platelet count, we calculated a ratio between platelet aggregation and platelet count (PLT ratio). Then, we prospectively followed patients with cirrhosis and ascertained predictors of decompensation, transplantation, and death. Results Two-hundred and three cirrhosis patients were prospectively recruited (77% decompensated). PLT ratio was significantly higher in cirrhosis than in chronic hepatitis and healthy subjects (0.44 vs. 0.25 and 0.26, respectively; p<0.0001). In cirrhosis, the ratio increased with disease severity (Child-Pugh class C>B>A) and was particularly elevated in decompensated patients with severe thrombocytopenia. Among decompensated patients, 65 had further decompensation, underwent transplantation, or died during a 6-month follow-up. On multivariate analysis, PLT ratio (OR: 1.87, 95%CI: 1.23-2.84; p=0.003) and MELD score (OR: 1.05, 95%CI: 1.01-1.08; p=0.01) were independently associated with outcome. The relative risk of events was 7.5-fold higher in patients with PLT ratio >0.75 versus patients with PLT ratio <0.25 (RR: 7.5, 95% CI: 2.5-21.9; p=0.003). The increased PLT ratio, its discriminative ability for composite outcome, and the prognostic value of PLT ratio >0.75 were confirmed in an independent cohort of hospitalized patients with decompensated cirrhosis (n=41). Conclusions Patients with cirrhosis, particularly when decompensated, exhibit significantly increased whole blood platelet aggregation. Decompensated patients with PLT ratio >0.75 have >80% probability of further decompensation, transplantation, or liver-related death within 6 months. Lay summary In patients with advanced, chronic liver disease (i.e. cirrhosis), previous studies have suggested that platelets (i.e. circulating blood cells that help form clots to stop bleeding) are dysfunctional. In particular, these studies suggested that platelet aggregation (the process by which platelets adhere to each other to form clots) is reduced. Since platelet aggregation is important for clot formation, it has been hypothesized that alterations of platelet aggregation may be responsible for the increased risk of bleeding observed in patients with cirrhosis. Our study demonstrates: 1) that platelet aggregation in patients with cirrhosis is higher than in healthy individuals; 2) that platelet aggregation in patients with decompensated cirrhosis (i.e. those who have already experienced some complications of cirrhosis) is particularly elevated and associated with risk of cirrhosis progression towards further complications and death. Trial registration number NA
Article
The prevention and management of bleeding and thrombosis in patients with cirrhosis poses several difficult clinical questions. These Clinical Practice Guidelines have been developed to provide practical guidance on debated topics, including current views on haemostasis in liver disease, controversy regarding the need to correct thrombocytopenia and abnormalities in the coagulation system in patients undergoing invasive procedures, and the need for thromboprophylaxis in hospitalised patients with haemostatic abnormalities. Multiple recommendations in this document are based on interventions that the panel feels are not useful, even though widely applied in clinical practice.